Subscribe or renew today

Every print subscription comes with full digital access

Science News

A man wearing a blue-green shirt and a red sash around his waist rides a dark brown horse in pursuit of a riderless white horse. Three other reddish horses run across a plain covered in straw-colored grass.

Horses may have been domesticated twice. Only one attempt stuck

Genetic evidence suggests that the ancestors of domestic horses were bred for mobility about 4,200 years ago.

Scientists are fixing flawed forensics that can lead to wrongful convictions

Thomas cech’s ‘the catalyst’ spotlights rna and its superpowers, more stories in genetics.

A chimera pig embryo

50 years ago, chimeras gave a glimpse of gene editing’s future

Advances in gene editing technology have led to the first successful transplant of a pig kidney into a human.

Several ferns with forest in the background

The largest known genome belongs to a tiny fern

Though 'Tmesipteris oblanceolata' is just 15 centimeters long, its genome dwarfs humans’ by more than 50 times.

Here’s why some pigeons do backflips

Meet the scientist homing in on the genes involved in making parlor roller pigeons do backward somersaults.

Two chimpanzees hang from a rope with two hands above a grassy field. Both are facing away from the camera.

A genetic parasite may explain why humans and other apes lack tails

Around 25 million years ago, a stretch of DNA inserted itself into an ancestral ape’s genome, an event that might have taken our tails away.

Stacks of long tubes of various lengths are seen. Inside the tubes is a bright purple "filling". This is the long part of a nerve fiber called an axon. Around those fibers are thick tubes colored brownish-gray that form an insulating sheath around the nerve. Some wispy strands of connective tissue lays over some of the tubes. Connective tissue is colored hot pink.

Ancient viruses helped speedy nerves evolve

A retrovirus embedded in the DNA of some vertebrates helps turn on production of a protein needed to insulate nerve cells, aiding speedy thoughts.

A young female-presenting person with allergies sneezes into a white handkerchief. They have brown skin and black hair pulled back into a ponytail. They are wearing a light yellow shirt and a backpack with black straps with a neon green camping roll strapped across their shoulders. Trees in various shades of green are blurred in the background.

Newfound immune cells are responsible for long-lasting allergies

A specialized type of immune cell appears primed to make the type of antibodies that lead to allergies, two research groups report.

A photograph of Krystal Tsosie smiling in her white lab coat, which has an embroidered tortoise on it.

Geneticist Krystal Tsosie advocates for Indigenous data sovereignty

A member of the Navajo Nation, she believes Indigenous geneticists have a big role to play in protecting and studying their own data.

A skull with an arrow shot through the nose is displayed on a black background. The Danish bog skull, known as Porsmose Man, dates to around 4,600 years ago.

How ancient herders rewrote northern Europeans’ genetic story

New DNA analyses show the extent of the Yamnaya people’s genetic reach starting 5,000 years ago and how it made descendants prone to diseases like MS.

A woman with long dark hair wearing a white T-shirt holds her right hand over her mouth while her left hand braces her against the wall. The image conveys nauseousness.

Fetuses make a protein that causes morning sickness in pregnancy

A hormone called GDF15 triggers a part of the brain involved in nausea and vomiting, a new study finds. Blocking its action may lead to treatments.

Subscribers, enter your e-mail address for full access to the Science News archives and digital editions.

Not a subscriber? Become one now .

Suggestions or feedback?

MIT News | Massachusetts Institute of Technology

  • Machine learning
  • Social justice
  • Black holes
  • Classes and programs

Departments

  • Aeronautics and Astronautics
  • Brain and Cognitive Sciences
  • Architecture
  • Political Science
  • Mechanical Engineering

Centers, Labs, & Programs

  • Abdul Latif Jameel Poverty Action Lab (J-PAL)
  • Picower Institute for Learning and Memory
  • Lincoln Laboratory
  • School of Architecture + Planning
  • School of Engineering
  • School of Humanities, Arts, and Social Sciences
  • Sloan School of Management
  • School of Science
  • MIT Schwarzman College of Computing

Scientists develop a rapid gene-editing screen to find effects of cancer mutations

Press contact :, media download.

Pastel illustration shows two hands, creating a circle, and one hand holds a pencil and draws a DNA strand. More DNA strands are around the hands.

*Terms of Use:

Images for download on the MIT News office website are made available to non-commercial entities, press and the general public under a Creative Commons Attribution Non-Commercial No Derivatives license . You may not alter the images provided, other than to crop them to size. A credit line must be used when reproducing images; if one is not provided below, credit the images to "MIT."

Pastel illustration shows two hands, creating a circle, and one hand holds a pencil and draws a DNA strand. More DNA strands are around the hands.

Previous image Next image

Tumors can carry mutations in hundreds of different genes, and each of those genes may be mutated in different ways — some mutations simply replace one DNA nucleotide with another, while others insert or delete larger sections of DNA.

Until now, there has been no way to quickly and easily screen each of those mutations in their natural setting to see what role they may play in the development, progression, and treatment response of a tumor. Using a variant of CRISPR genome-editing known as prime editing, MIT researchers have now come up with a way to screen those mutations much more easily.

The researchers demonstrated their technique by screening cells with more than 1,000 different mutations of the tumor suppressor gene p53, all of which have been seen in cancer patients. This method, which is easier and faster than any existing approach, and edits the genome rather than introducing an artificial version of the mutant gene, revealed that some p53 mutations are more harmful than previously thought.

This technique could also be applied to many other cancer genes, the researchers say, and could eventually be used for precision medicine, to determine how an individual patient’s tumor will respond to a particular treatment.

“In one experiment, you can generate thousands of genotypes that are seen in cancer patients, and immediately test whether one or more of those genotypes are sensitive or resistant to any type of therapy that you’re interested in using,” says Francisco Sanchez-Rivera, an MIT assistant professor of biology, a member of the Koch Institute for Integrative Cancer Research, and the senior author of the study.

MIT graduate student Samuel Gould is the lead author of the paper , which appears today in Nature Biotechnology .

Editing cells

The new technique builds on research that Sanchez-Rivera began 10 years ago as an MIT graduate student. At that time, working with Tyler Jacks, the David H. Koch Professor of Biology, and then-postdoc Thales Papagiannakopoulos, Sanchez-Rivera developed a way to use CRISPR genome-editing to introduce into mice genetic mutations linked to lung cancer.

In that study, the researchers showed that they could delete genes that are often lost in lung tumor cells, and the resulting tumors were similar to naturally arising tumors with those mutations. However, this technique did not allow for the creation of point mutations (substitutions of one nucleotide for another) or insertions.

“While some cancer patients have deletions in certain genes, the vast majority of mutations that cancer patients have in their tumors also include point mutations or small insertions,” Sanchez-Rivera says.

Since then, David Liu, a professor in the Harvard University Department of Chemistry and Chemical Biology and a core institute member of the Broad Institute, has developed new CRISPR-based genome editing technologies that can generate additional types of mutations more easily. With base editing, developed in 2016, researchers can engineer point mutations, but not all possible point mutations. In 2019, Liu, who is also an author of the Nature Biotechnology study, developed a technique called prime editing, which enables any kind of point mutation to be introduced, as well as insertions and deletions.

“Prime editing in theory solves one of the major challenges with earlier forms of CRISPR-based editing, which is that it allows you to engineer virtually any type of mutation,” Sanchez-Rivera says.

When they began working on this project, Sanchez-Rivera and Gould calculated that if performed successfully, prime editing could be used to generate more than 99 percent of all small mutations seen in cancer patients.

However, to achieve that, they needed to find a way to optimize the editing efficiency of the CRISPR-based system. The prime editing guide RNAs (pegRNAs) used to direct CRISPR enzymes to cut the genome in certain spots have varying levels of efficiency, which leads to “noise” in the data from pegRNAs that simply aren’t generating the correct target mutation. The MIT team devised a way to reduce that noise by using synthetic target sites to help them calculate how efficiently each guide RNA that they tested was working.

“We can design multiple prime-editing guide RNAs with different design properties, and then we get an empirical measurement of how efficient each of those pegRNAs is. It tells us what percentage of the time each pegRNA is actually introducing the correct edit,” Gould says.

Analyzing mutations

The researchers demonstrated their technique using p53, a gene that is mutated in more than half of all cancer patients. From a dataset that includes sequencing information from more than 40,000 patients, the researchers identified more than 1,000 different mutations that can occur in p53.

“We wanted to focus on p53 because it’s the most commonly mutated gene in human cancers, but only the most frequent variants in p53 have really been deeply studied. There are many variants in p53 that remain understudied,” Gould says.

Using their new method, the researchers introduced p53 mutations in human lung adenocarcinoma cells, then measured the survival rates of these cells, allowing them to determine each mutation’s effect on cell fitness.

Among their findings, they showed that some p53 mutations promoted cell growth more than had been previously thought. These mutations, which prevent the p53 protein from forming a tetramer — an assembly of four p53 proteins — had been studied before, using a technique that involves inserting artificial copies of a mutated p53 gene into a cell.

Those studies found that these mutations did not confer any survival advantage to cancer cells. However, when the MIT team introduced those same mutations using the new prime editing technique, they found that the mutation prevented the tetramer from forming, allowing the cells to survive. Based on the studies done using overexpression of artificial p53 DNA, those mutations would have been classified as benign, while the new work shows that under more natural circumstances, they are not.

“This is a case where you could only observe these variant-induced phenotypes if you're engineering the variants in their natural context and not with these more artificial systems,” Gould says. “This is just one example, but it speaks to a broader principle that we’re going to be able to access novel biology using these new genome-editing technologies.”

Because it is difficult to reactivate tumor suppressor genes, there are few drugs that target p53, but the researchers now plan to investigate mutations found in other cancer-linked genes, in hopes of discovering potential cancer therapies that could target those mutations. They also hope that the technique could one day enable personalized approaches to treating tumors.

“With the advent of sequencing technologies in the clinic, we'll be able to use this genetic information to tailor therapies for patients suffering from tumors that have a defined genetic makeup,” Sanchez-Rivera says. “This approach based on prime editing has the potential to change everything.”

The research was funded, in part, by the National Institute of General Medical Sciences, an MIT School of Science Fellowship in Cancer Research, a Howard Hughes Medical Institute Hanna Gray Fellowship, the V Foundation for Cancer Research, a National Cancer Institute Cancer Center Support Grant, the Ludwig Center at MIT, the Koch Institute Frontier Research Program via the Casey and Family Foundation Cancer Research Fund, Upstage Lung Cancer, and the Michael (1957) and Inara Erdei Cancer Research Fund, and the MIT Research Support Committee.

Share this news article on:

Related links.

  • Francisco Sanchez-Rivera
  • Koch Institute
  • Department of Biology

Related Topics

  • Broad Institute

Related Articles

Microscopic view of pink DNA strand is in the center while blue blobs bind to its left and right sides.

Gene-editing technique could speed up study of cancer mutations

(Left to right) Tyler Jacks, Francisco Sanchez-Rivera, and Thales Papagiannakopoulos.

Fast modeling of cancer mutations

Previous item Next item

More MIT News

Ashesh Rambachan converses with a student in the front of a classroom.

A data-driven approach to making better choices

Read full story →

On the left, Erik Lin-Greenberg talks, smiling, with two graduate students in his office. On the right, Tracy Slatyer sits with two students on a staircase, conversing warmly.

Paying it forward

Portrait photo of John Fucillo posing on a indoor stairwell

John Fucillo: Laying foundations for MIT’s Department of Biology

Graphic of hand holding a glowing chip-based 3D printer

Researchers demonstrate the first chip-based 3D printer

Drawing of old English church with British Pound signs overlaid in some blank areas.

The unexpected origins of a modern finance tool

Primordial black hole forming amid a sea of color-charged quarks and gluons

Exotic black holes could be a byproduct of dark matter

  • More news on MIT News homepage →

Massachusetts Institute of Technology 77 Massachusetts Avenue, Cambridge, MA, USA

  • Map (opens in new window)
  • Events (opens in new window)
  • People (opens in new window)
  • Careers (opens in new window)
  • Accessibility
  • Social Media Hub
  • MIT on Facebook
  • MIT on YouTube
  • MIT on Instagram

MIT Technology Review

  • Newsletters

The first gene-editing treatment: 10 Breakthrough Technologies 2024

Sickle-cell disease is the first illness to be beaten by CRISPR, but the new treatment comes with an expected price tag of $2 to $3 million.

  • Antonio Regalado archive page

a scientist looks at a tall strand of DNA in a suspension of liquid. A hose sends the liquid back to an IV and into the arm of a patient seated comfortably in a domestic chair with two nice plants and a happy, observant cat.

CRISPR Therapeutics, Editas Medicine, Precision BioSciences, Vertex Pharmaceuticals

The first gene-editing cure has arrived. Grateful patients are calling it “life changing.”

It was only 11 years ago that scientists first developed the potent DNA-snipping technology called CRISPR. Now they’ve brought CRISPR out of the lab and into real medicine with a treatment that cures the symptoms of sickle-cell disease.

Sickle-cell is caused by inheriting two bad copies of one of the genes that make hemoglobin. Symptoms include bouts of intense pain, and life expectancy with the disease is just 53 years. It affects 1 in 4,000 people in the US, nearly all of them African-American. 

So how did this disease become CRISPR’s first success ? A fortuitous fact of biology is part of the answer. Our bodies harbor another way to make hemoglobin that turns off when we’re born. Researchers found that a simple DNA edit to cells from the bone marrow could turn it back on.

Many CRISPR treatments are in trials, but in 2022, Vertex Pharmaceuticals, based in Boston, was first to bring one to regulators for approval. That treatment was for sickle-cell. After their bone marrow was edited, nearly all the patients who volunteered in the trial were pain free. 

Good news. But the expected price tag of the gene-editing treatment is $2 to $3 million. And Vertex has no immediate plans to offer it in Africa—where sickle-cell disease is most common, and where it still kills children.

The company says this is because the treatment regimen is so complex. It involves a hospital stay; doctors remove the bone marrow, edit the cells, and then transplant them back. In countries that still struggle to cover basic health needs, the procedure remains too demanding. So simpler, cheaper ways to deliver CRISPR could come next. 

Biotechnology and health

What’s next for mdma.

The FDA is poised to approve the notorious party drug as a therapy. Here’s what it means, and where similar drugs stand in the US. 

  • Cassandra Willyard archive page

Beyond Neuralink: Meet the other companies developing brain-computer interfaces

Companies like Synchron, Paradromics, and Precision Neuroscience are also racing to develop brain implants

My biotech plants are dead

Two ‘Firefly petunias’ perished in a shipping misadventure, but other customers have had better luck.

The effort to make a breakthrough cancer therapy cheaper

CAR-T cells could revolutionize the treatment of a wide variety of diseases, if only we can make them cheaper.

Stay connected

Get the latest updates from mit technology review.

Discover special offers, top stories, upcoming events, and more.

Thank you for submitting your email!

It looks like something went wrong.

We’re having trouble saving your preferences. Try refreshing this page and updating them one more time. If you continue to get this message, reach out to us at [email protected] with a list of newsletters you’d like to receive.

  • Skip to main content
  • Keyboard shortcuts for audio player

Shots - Health News

  • Your Health
  • Treatments & Tests
  • Health Inc.
  • Public Health

The CRISPR Revolution

He inherited a devastating disease. a crispr gene-editing breakthrough stopped it.

Rob Stein, photographed for NPR, 22 January 2020, in Washington DC.

Patrick Doherty volunteered for a new medical intervention of gene-editor infusions for the treatment of genetically-based diseases. Patrick Doherty hide caption

Patrick Doherty volunteered for a new medical intervention of gene-editor infusions for the treatment of genetically-based diseases.

Patrick Doherty had always been very active. He trekked the Himalayas and hiked trails in Spain.

But about a year and a half ago, he noticed pins and needles in his fingers and toes. His feet got cold. And then he started getting out of breath any time he walked his dog up the hills of County Donegal in Ireland where he lives.

"I noticed on some of the larger hill climbs I was getting a bit breathless," says Doherty, 65. "So I realized something was wrong."

Doherty found out he had a rare, but devastating inherited disease — known as transthyretin amyloidosis — that had killed his father. A misshapen protein was building up in his body, destroying important tissues, such as nerves in his hands and feet and his heart.

Doherty had watched others get crippled and die difficult deaths from amyloidosis.

"It's terrible prognosis," Doherty says. "This is a condition that deteriorates very rapidly. It's just dreadful."

So Doherty was thrilled when he found out that doctors were testing a new way to try to treat amyloidosis. The approach used a revolutionary gene-editing technique called CRISPR , which allows scientists to make very precise changes in DNA.

"I thought: Fantastic. I jumped at the opportunity," Doherty says.

On Saturday, researchers reported the first data indicating that the experimental treatment worked, causing levels of the destructive protein to plummet in Doherty's body and the bodies of five other patients treated with the approach.

"I feel fantastic," Doherty says. "It's just phenomenal."

The advance is being hailed not just for amyloidosis patients but also as a proof-of-concept that CRISPR could be used to treat many other, much more common diseases. It's a new way of using the innovative technology.

"This is a major milestone for patients," says Jennifer Doudna of the University of California, Berkeley, who shared a Nobel Prize for her work helping develop CRISPR.

"While these are early data, they show us that we can overcome one of the biggest challenges with applying CRISPR clinically so far, which is being able to deliver it systemically and get it to the right place," Doudna says.

CRISPR has already been shown to help patients suffering from the devastating blood disorders sickle cell disease and beta thalassemia . And doctors are trying to use it to treat cancer and to restore vision to people blinded by a rare genetic disorder.

But those experiments involve taking cells out of the body, editing them in the lab, and infusing them back in or injecting CRISPR directly into cells that need fixing.

The study Doherty volunteered for is the first in which doctors are simply infusing the gene-editor directly into patients and letting it find its own way to the right gene in the right cells. In this case, it's cells in the liver making the destructive protein.

"This is the first example in which CRISPR-Cas9 is injected directly into the bloodstream — in other words systemic administration — where we use it as a way to reach a tissue that's far away from the site of injection and very specifically use it to edit disease-causing genes," says John Leonard, the CEO of Intellia Therapeutics , which is sponsoring the study.

Doctors infused billions of microscopic structures known as nanoparticles carrying genetic instructions for the CRISPR gene-editor into four patients in London and two in New Zealand. The nanoparticles were absorbed by their livers, where they unleashed armies of CRISPR gene-editors. The CRISPR editor homed in on the target gene in the liver and sliced it, disabling production of the destructive protein.

Within weeks, the levels of protein causing the disease plummeted, especially in the volunteers who received a higher dose. Researchers reported at the Peripheral Nerve Society Annual Meeting and in a paper published in The New England Journal of Medicine .

"It really is exciting," says Dr. Julian Gillmore , who is leading the study at the University College London, Royal Free Hospital.

"This has the potential to completely revolutionize the outcome for these patients who have lived with this disease in their family for many generations. It's decimated some families that I've been looking after. So this is amazing," Gillmore says.

The patients will have to be followed longer, and more patients will have to be treated, to make sure the treatment's safe, and determine how much it's helping, Gillmore stresses. But the approach could help those struck by amyloidosis that isn't inherited, which is a far more common version of the disease, he says.

Moreover, the promising results potentially open the door for using the same approach to treatment of many other, more common diseases for which taking cells out of the body or directly injecting CRISPR isn't realistic, including heart disease, muscular dystrophy and brain diseases such as Alzheimer's.

"This is really opening a new era as we think about gene-editing where we can begin to think about accessing all kinds of different tissue in the body via systemic administration," Leonard says.

Other scientists who are not involved in the research agree.

"This is a wonderful day for the future of gene-editing as a medicine," agree Fyodor Urnov , a professor of genetics at the University of California, Berkeley. "We as a species are watching this remarkable new show called: our gene-edited future."

Doherty says he started feeling better within weeks of the treatment and has continued to improve in the weeks since then.

"I definitely feel better," he told NPR. "I'm speaking to you from upstairs in our house. I climbed stairs to get up here. I would have been feeling breathless. I'm thrilled."

  • Alzheimer's disease & dementia
  • Arthritis & Rheumatism
  • Attention deficit disorders
  • Autism spectrum disorders
  • Biomedical technology
  • Diseases, Conditions, Syndromes
  • Endocrinology & Metabolism
  • Gastroenterology
  • Gerontology & Geriatrics
  • Health informatics
  • Inflammatory disorders
  • Medical economics
  • Medical research
  • Medications
  • Neuroscience
  • Obstetrics & gynaecology

Oncology & Cancer

  • Ophthalmology
  • Overweight & Obesity
  • Parkinson's & Movement disorders
  • Psychology & Psychiatry
  • Radiology & Imaging
  • Sleep disorders
  • Sports medicine & Kinesiology
  • Vaccination
  • Breast cancer
  • Cardiovascular disease
  • Chronic obstructive pulmonary disease
  • Colon cancer
  • Coronary artery disease
  • Heart attack
  • Heart disease
  • High blood pressure
  • Kidney disease
  • Lung cancer
  • Multiple sclerosis
  • Myocardial infarction
  • Ovarian cancer
  • Post traumatic stress disorder
  • Rheumatoid arthritis
  • Schizophrenia
  • Skin cancer
  • Type 2 diabetes
  • Full List »

Last update: 4 hours ago

Researchers detail molecular pathway that impacts pancreatic cancer progression and treatment response

  • Date 6 hours 12 hours 1 day 3 days all
  • Rank Last day 1 week 1 month all
  • LiveRank Last day 1 week 1 month all
  • Popular Last day 1 week 1 month all

Genetics news

Researchers at UNC Lineberger Comprehensive Cancer Center and colleagues have established the most comprehensive molecular portrait of the workings of KRAS, a key cancer-causing gene or "oncogene," and how its activities ...

4 hours ago

Study uncovers how cancer stem cells spread and resist treatment

NDORMS researchers have identified a critical axis that controls the formation and behavior of cancer stem cells (CSCs), a subpopulation of cells that influence how lethal the cancer can be, its resistance to chemotherapy, ...

6 hours ago

Long-read RNA sequencing reveals key gene expressions in Alzheimer's disease

Researchers at the University of Kentucky Sanders-Brown Center on Aging are working to develop a pre-symptomatic disease diagnostic tool for Alzheimer's disease.

9 hours ago

latest genetics research

Genetics study points to potential treatments for restless leg syndrome

Scientists have discovered genetic clues to the cause of restless leg syndrome, a condition common among older adults. The discovery could help identify those individuals at greatest risk of the condition and point to potential ...

Jun 5, 2024

latest genetics research

Study suggests father's diet before conception influences children's health

Dr. Raffaele Teperino, head of the "Environmental Epigenetics" research group at Helmholtz Munich, along with his research team, has examined the impact of paternal diet on children's health—specifically, the influence ...

latest genetics research

New insights on cellular clones and inflammation in bones

As humans age, hematopoietic stem cells—the immature precursor cells that give rise to all blood and immune cells—accumulate mutations. Some of the mutations allow these stem cells to self-renew and expand more effectively ...

latest genetics research

Prostate cancer study reveals molecular switch linked to lineage plasticity, therapy resistance

Two genes working in tandem play a critical role in shaping the identity and behavior of prostate cancer cells and their response to treatment, UT Southwestern Medical Center researchers report.

latest genetics research

Study finds maternal obesity in mice increases microRNA levels in the hypothalamus in offspring, leading to overeating

Maternal obesity impacts the eating behaviors of offspring via long-term overexpression of the microRNA miR-505-5p, according to a study publishing June 4 in the open-access journal PLOS Biology by Laura Dearden and Susan ...

Jun 4, 2024

latest genetics research

Rare disease's mutation could explain more common conditions

TREX1 is a gene that is supposed to direct the maintenance of the entire body's DNA, but new research shows that when people are born with mutated TREX1, it causes catastrophic damage to the DNA over time, resulting in a ...

latest genetics research

New role of tumor suppressor STAT3β discovered in leukemia

The protein STAT3β has a positive influence on the course of acute myeloid leukemia (AML) and could serve as a prognostic marker in the future. These are the outstanding results of a cancer research study at the Karl Landsteiner ...

latest genetics research

Novel software combines gene activity and tissue location to decode disease mechanisms

In disease research, it's important to know gene expression and where in a tissue the expression is happening, but marrying the two sets of information can be challenging.

latest genetics research

Muscle disorder caused by key protein mutations uncovered in new study

A recent study has found that the SMCHD1 protein plays a key role in controlling how genes are processed, which affects the progression of Facioscapulohumeral Muscular Dystrophy (FSHD). This discovery of SMCHD1's function ...

latest genetics research

Shared genetic links between sleep and neuropsychiatric conditions may lead to new treatments

Disturbed sleep is very common in almost all neuropsychiatric and neurodevelopmental conditions (NDPCs), such as autism, attention deficit and hyperactivity disorder, schizophrenia, and bipolar disorder.

latest genetics research

New method offers faster, more accurate pathogen identification, even in complex DNA sequences

A team of researchers from the Case Western Reserve University School of Medicine has developed a new method for target DNA sequence amplification, testing and analysis.

Jun 3, 2024

latest genetics research

Cracking the aging code: Insights into lipid changes

Researchers at the RIKEN Center for Integrative Medical Sciences (IMS) have discovered numerous age-related changes in the lipid metabolism of mice, across both organs and sexes. Among these changes was the selective accumulation, ...

latest genetics research

New analytical tool can improve understanding of heritable human traits and diseases

Researchers from the University of Oslo have developed an innovative method to improve our understanding of heritable human traits and diseases. The analytical tool, called GSA-MiXeR, is designed to make sense of genetic ...

latest genetics research

Genetic changes identified as key to childhood lupus

Australian and Chinese researchers have jointly discovered a genetic key to the development of the devastating autoimmune condition childhood lupus.

latest genetics research

Chromatin openness sheds new light on prostate cancer plasticity

Treatment resistance caused by cancer cell plasticity constitutes a major challenge in the treatment of prostate cancer. A recent study from the University of Eastern Finland Institute of Biomedicine published in Nucleic ...

latest genetics research

Targeting leukemia's survival route: A novel approach to overcoming leukemia recurrence

Researchers from Karolinska Institutet have, in collaboration with the University of Eastern Finland and Lund University, published a study in Genome Biology demonstrating that targeting cell cycle and cell fate regulatory ...

latest genetics research

Study finds more women in oil-rich Gulf countries battle with breast cancer

Breast cancer incidence has surged in the oil-rich Gulf (GCC) states, with the disease developing its own localized clinical and pathological features, setting them apart from those found in women with breast cancer in western ...

latest genetics research

10 hours ago

latest genetics research

5 hours ago

latest genetics research

7 hours ago

latest genetics research

8 hours ago

latest genetics research

E-mail newsletter

SciTechDaily

  • June 6, 2024 | Protectors of the Ice? Strange Giant Viruses Discovered on Greenland Ice Sheet
  • June 6, 2024 | Quantum Pioneers: How Magnetic Quivers Are Rewriting the Rules of Particle Physics
  • June 6, 2024 | James Webb Unmasks the Carbon-Rich Secrets of Protoplanetary Disks
  • June 6, 2024 | Just Three Atoms Thick – Scientists Have Developed the World’s Thinnest Lens
  • June 6, 2024 | From Corneas to Hearts: Revolutionary Jelly-Like Material Promises To Mend Human Organs

Genetics News

Genetics is the study of genes, heredity, and genetic variation in living organisms. Our Genetics category provides the latest news, research, and discoveries in this exciting field, including the latest breakthroughs in genetic engineering, gene therapy, and personalized medicine. From the study of genetic diseases to the genetics of complex traits, our coverage of Genetics provides a comprehensive look at the latest advancements in genetic research. We also cover the latest developments in genetic technology, including advances in sequencing, gene editing, and gene expression analysis. Whether you’re a geneticist, medical professional, or just interested in the latest advancements in genetics research, our Genetics category has everything you need to stay up-to-date and informed.

Arabian Horse

Science June 6, 2024

The Dawn of Horse Power: Using DNA To Trace Back 4,200 Years

Domestic horses, originating from the western Russian steppes about 4,200 years ago, transformed Eurasian societies by facilitating faster trade and interactions. An international research team…

Sad Fat Boy

Health June 5, 2024

Study: Treatment-Resistant Depression Linked to BMI

Recent research highlights a genetic contribution to treatment-resistant depression, linking it with several health disorders and suggesting new treatment possibilities based on shared genetic factors….

Abstract Planet DNA Art Concept

Science June 5, 2024

Unlocking the Genetic Giant: Tiny Fern Has the Largest Genome of Any Organism on Earth

The Tmesipteris oblanceolata fern sets a new record for the largest genome, impacting plant growth and adaptation insights. A New Caledonian fern species was awarded…

DNA Genetics Evolution Artist's Concept

Biology June 4, 2024

Breaking Evolution’s Rules: Scientists Uncover Surprising Reemergence of Once-Lost Biological Traits

Recent research shows that genes linked to lost traits in stick insects may be preserved, enabling their potential re-emergence over evolutionary time. Traits can disappear…

Evolutionary Context of Anglerfish Immunogenomic Degradation

Sexual Parasitism: Anglerfish’s Strategy To Conquer the Deep Sea Amid Global Warming

Research published in the scientific journal Current Biology reveals that sexual parasitism in anglerfishes originated during a major global warming event that led to their…

Genetics DNA Mutation Concept

Health May 31, 2024

How a Single Gene Mutation is Rewriting the Book on Intellectual Disability

Researchers have discovered a neurodevelopmental disorder linked to mutations in the RNU4-2 gene, a non-coding gene, which could impact tens of thousands globally. This finding…

Mutated Amino Acids in the AFF3 Protein

Biology May 30, 2024

Developmental Disorders: Researchers Link Protein Levels to Severe Intellectual Deficiencies

New research reveals that both the excess and the deficiency of a single protein can lead to severe intellectual deficiencies. The discovery offers critical insights…

Great Ape Genetic Chromosome Sequencing Art

Biology May 29, 2024

Surprising Evolutionary Insights Revealed by First Complete Chromosome Sequences From Great Apes

Complete X and Y chromosome sequences from six different primate species have been successfully mapped, revealing a rich diversity among these species and providing deeper…

Darkness Eye Night

Biology May 28, 2024

Johns Hopkins Scientists Solve 30-Year Biological Mystery of Night Blindness

Johns Hopkins scientists identified how the G90D mutation in the rhodopsin gene causes night blindness, offering potential therapeutic targets. In what they believe is a…

1606 Virginia Map

Science May 26, 2024

New Research Reveals That the First Permanent English Settlers in North America Ate Dogs

Researchers discovered that six dogs from the Virginia Company Period in Jamestown—the first permanent English settlement in North America—possessed Indigenous ancestry and were eaten by…

CRISPR Genome Editing

Biology May 25, 2024

Simple New Strategy Improves Safety and Precision of CRISPR Gene Editing

KAUST researchers have improved CRISPR gene editing safety by reducing harmful DNA deletions and enhancing repair mechanisms, advancing towards safer genetic treatments. A simple and…

Alzheimer's Brain Scan Art Concept

Biology May 24, 2024

Researchers Identify New Genetic Form of Alzheimer’s Disease

Scientists have discovered that nearly all individuals with two copies of the APOE4 gene will develop signs of Alzheimer’s disease, suggesting a distinct genetic form…

Cell in Cell Phenomena

Biology May 23, 2024

Across the Entire Tree of Life – “Cell Cannibalism” Is Much More Common Than Scientists Thought

New research reveals that cell-in-cell phenomena, where one cell consumes another, are common across all life forms and important for normal biological functions, not just…

Deep Sea Anglerfish Illustration

Sexual Parasitism: Yale Scientists Unveil the Mating Mysteries of Deep-Sea Anglerfish

Yale researchers have uncovered how sexual parasitism among deep-sea anglerfish, involving males permanently attaching to females, facilitated their adaptation from shallow-water habitats to the deep-sea…

mRNA RISC RNAi

Health May 23, 2024

Harnessing RNAi: Alnylam’s Path From Lab Discovery to Life-Changing Treatments

The Journey From Research Discovery to Impactful New Treatments Alnylam Pharmaceuticals, founded by MIT professors and former postdocs, has developed five FDA-approved RNAi medicines, significantly…

Biochemistry Protein Art

Biology May 17, 2024

A Breakthrough in Protein Genetics: Scientists Unveil How Pseudogenes Evolve

Rice University’s Peter Wolynes and his research team have made a significant breakthrough in understanding the evolution of specific genetic sequences known as pseudogenes. Their…

Cancer Cell Death Concept

Health May 16, 2024

Assassin Gene Discovery Shifts Cancer Treatment Paradigms – “Very Unexpected Finding”

Scientists have discovered that the way cancer cells die from chemotherapy appears to be different than previously understood. New research highlights the Schlafen11 gene’s role…

DNA Genetics Mutation Concept

Biology May 14, 2024

Gene Linked to Learning Difficulties Has Direct Impact on Learning and Memory

A gene previously associated with intellectual disability has been discovered to control learning and memory in mice. The gene, called KDM5B has previously been linked…

ScienceDaily

Genetics study points to potential treatments for restless leg syndrome

Scientists have discovered genetic clues to the cause of restless leg syndrome, a condition common among older adults. The discovery could help identify those individuals at greatest risk of the condition and point to potential ways to treat it.

Restless leg syndrome can cause an unpleasant crawling sensation in the legs and an overwhelming urge to move them. Some people experience the symptoms only occasionally, while others get symptoms every day. Symptoms are usually worse in the evening or at night-time and can severely impair sleep.

Despite the condition being relatively common -- up to one in 10 older adults experience symptoms, while 2-3% are severely affected and seek medical help -- little is known about its causes. People with restless leg syndrome often have other conditions, such as depression or anxiety, cardiovascular disorders, hypertension, and diabetes, but the reason why is not known.

Previous studies had identified 22 genetic risk loci -- that is, regions of our genome that contain changes associated with increased risk of developing the condition. But there are still no known 'biomarkers' -- such as genetic signatures -- that could be used to objectively diagnose the condition.

To explore the condition further, an international team led by researchers at the Helmholtz Munich Institute of Neurogenomics, Institute of Human Genetics of the Technical University of Munich (TUM) and the University of Cambridge pooled and analysed data from three genome-wide association studies. These studies compared the DNA of patients and healthy controls to look for differences more commonly found in those with restless leg syndrome. By combining the data, the team was able to create a powerful dataset with more than 100,000 patients and over 1.5 million unaffected controls.

The results of the study are published today in Nature Genetics .

Co-author Dr Steven Bell from the University of Cambridge said: "This study is the largest of its kind into this common -- but poorly understood -- condition. By understanding the genetic basis of restless leg syndrome, we hope to find better ways to manage and treat it, potentially improving the lives of many millions of people affected worldwide."

The team identified over 140 new genetic risk loci, increasing the number known eight-fold to 164, including three on the X chromosome. The researchers found no strong genetic differences between men and women, despite the condition being twice as common in women as it is men -- this suggests that a complex interaction of genetics and the environment (including hormones) may explain the gender differences we observe in real life.

Two of the genetic differences identified by the team involve genes known as glutamate receptors 1 and 4 respectively, which are important for nerve and brain function. These could potentially be targeted by existing drugs, such as anticonvulsants like perampanel and lamotrigine, or used to develop new drugs. Early trials have already shown positive responses to these drugs in patients with restless leg syndrome.

The researchers say it would be possible to use basic information like age, sex, and genetic markers to accurately rank who is more likely to have severe restless leg syndrome in nine cases out of ten.

To understand how restless leg syndrome might affect overall health, the researchers used a technique called Mendelian randomisation. This uses genetic information to examine cause-and-effect relationships. It revealed that the syndrome increases the risk of developing diabetes.

Although low levels of iron in the blood are thought to trigger restless leg syndrome -- because they can lead to a fall in the neurotransmitter dopamine -- the researchers did not find strong genetic links to iron metabolism. However, they say they cannot completely rule it out as a risk factor.

Professor Juliane Winkelmann from TUM, one of senior authors of the study, said: "For the first time, we have achieved the ability to predict restless leg syndrome risk. It has been a long journey, but now we are empowered to not only treat but even prevent the onset of this condition in our patients."

  • Restless Leg Syndrome
  • Sleep Disorder Research
  • Down Syndrome
  • Diseases and Conditions
  • Personalized Medicine
  • Birth Defects
  • Heart Disease
  • Adult attention-deficit disorder
  • Runner's knee
  • Personalized medicine
  • Asperger syndrome
  • Drug discovery
  • Alzheimer's disease

Story Source:

Materials provided by University of Cambridge . The original text of this story is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License . Note: Content may be edited for style and length.

Journal Reference :

  • Barbara Schormair, Chen Zhao, Steven Bell, Maria Didriksen, Muhammad S. Nawaz, Nathalie Schandra, Ambra Stefani, Birgit Högl, Yves Dauvilliers, Cornelius G. Bachmann, David Kemlink, Karel Sonka, Walter Paulus, Claudia Trenkwalder, Wolfgang H. Oertel, Magdolna Hornyak, Maris Teder-Laving, Andres Metspalu, Georgios M. Hadjigeorgiou, Olli Polo, Ingo Fietze, Owen A. Ross, Zbigniew K. Wszolek, Abubaker Ibrahim, Melanie Bergmann, Volker Kittke, Philip Harrer, Joseph Dowsett, Sofiene Chenini, Sisse Rye Ostrowski, Erik Sørensen, Christian Erikstrup, Ole B. Pedersen, Mie Topholm Bruun, Kaspar R. Nielsen, Adam S. Butterworth, Nicole Soranzo, Willem H. Ouwehand, David J. Roberts, John Danesh, Brendan Burchell, Nicholas A. Furlotte, Priyanka Nandakumar, Amélie Bonnefond, Louis Potier, Christopher J. Earley, William G. Ondo, Lan Xiong, Alex Desautels, Markus Perola, Pavel Vodicka, Christian Dina, Monika Stoll, Andre Franke, Wolfgang Lieb, Alexandre F. R. Stewart, Svati H. Shah, Christian Gieger, Annette Peters, David B. Rye, Guy A. Rouleau, Klaus Berger, Hreinn Stefansson, Henrik Ullum, Kari Stefansson, David A. Hinds, Emanuele Di Angelantonio, Konrad Oexle, Juliane Winkelmann. Genome-wide meta-analyses of restless legs syndrome yield insights into genetic architecture, disease biology and risk prediction . Nature Genetics , 2024; DOI: 10.1038/s41588-024-01763-1

Cite This Page :

Explore More

  • 'Tweezer-Like' Bionic Tools Feel Right
  • Odd Planet-Forming Disks Around Low-Mass Stars
  • Toward Blood Stem Cell Self-Renewal
  • Restored Hearing and Speech in Kids Born Deaf
  • Babies and AI Both Learn Key Foundation Models
  • Myelination May Drive Drug Addiction
  • Freshwater On Earth 4 Billion Years Ago
  • Extended Battle: 3,500-Year-Old Mycenaean Armor
  • Oral Insulin Drops: Relief for Diabetes Patients
  • AIs Are Irrational, but Not Like Humans

Trending Topics

Strange & offbeat.

  • Share full article

Advertisement

Supported by

Guest Essay

The Long-Overlooked Molecule That Will Define a Generation of Science

latest genetics research

By Thomas Cech

Dr. Cech is a biochemist and the author of the forthcoming book “The Catalyst: RNA and the Quest to Unlock Life’s Deepest Secrets,” from which this essay is adapted.

From E=mc² to splitting the atom to the invention of the transistor, the first half of the 20th century was dominated by breakthroughs in physics.

Then, in the early 1950s, biology began to nudge physics out of the scientific spotlight — and when I say “biology,” what I really mean is DNA. The momentous discovery of the DNA double helix in 1953 more or less ushered in a new era in science that culminated in the Human Genome Project, completed in 2003, which decoded all of our DNA into a biological blueprint of humankind.

DNA has received an immense amount of attention. And while the double helix was certainly groundbreaking in its time, the current generation of scientific history will be defined by a different (and, until recently, lesser-known) molecule — one that I believe will play an even bigger role in furthering our understanding of human life: RNA.

You may remember learning about RNA (ribonucleic acid) back in your high school biology class as the messenger that carries information stored in DNA to instruct the formation of proteins. Such messenger RNA, mRNA for short, recently entered the mainstream conversation thanks to the role they played in the Covid-19 vaccines. But RNA is much more than a messenger, as critical as that function may be.

Other types of RNA, called “noncoding” RNAs, are a tiny biological powerhouse that can help to treat and cure deadly diseases, unlock the potential of the human genome and solve one of the most enduring mysteries of science: explaining the origins of all life on our planet.

Though it is a linchpin of every living thing on Earth, RNA was misunderstood and underappreciated for decades — often dismissed as nothing more than a biochemical backup singer, slaving away in obscurity in the shadows of the diva, DNA. I know that firsthand: I was slaving away in obscurity on its behalf.

In the early 1980s, when I was much younger and most of the promise of RNA was still unimagined, I set up my lab at the University of Colorado, Boulder. After two years of false leads and frustration, my research group discovered that the RNA we’d been studying had catalytic power. This means that the RNA could cut and join biochemical bonds all by itself — the sort of activity that had been thought to be the sole purview of protein enzymes. This gave us a tantalizing glimpse at our deepest origins: If RNA could both hold information and orchestrate the assembly of molecules, it was very likely that the first living things to spring out of the primordial ooze were RNA-based organisms.

That breakthrough at my lab — along with independent observations of RNA catalysis by Sidney Altman at Yale — was recognized with a Nobel Prize in 1989. The attention generated by the prize helped lead to an efflorescence of research that continued to expand our idea of what RNA could do.

In recent years, our understanding of RNA has begun to advance even more rapidly. Since 2000, RNA-related breakthroughs have led to 11 Nobel Prizes. In the same period, the number of scientific journal articles and patents generated annually by RNA research has quadrupled. There are more than 400 RNA-based drugs in development, beyond the ones that are already in use. And in 2022 alone, more than $1 billion in private equity funds was invested in biotechnology start-ups to explore frontiers in RNA research.

What’s driving the RNA age is this molecule’s dazzling versatility. Yes, RNA can store genetic information, just like DNA. As a case in point, many of the viruses (from influenza to Ebola to SARS-CoV-2) that plague us don’t bother with DNA at all; their genes are made of RNA, which suits them perfectly well. But storing information is only the first chapter in RNA’s playbook.

Unlike DNA, RNA plays numerous active roles in living cells. It acts as an enzyme, splicing and dicing other RNA molecules or assembling proteins — the stuff of which all life is built — from amino acid building blocks. It keeps stem cells active and forestalls aging by building out the DNA at the ends of our chromosomes.

RNA discoveries have led to new therapies, such as the use of antisense RNA to help treat children afflicted with the devastating disease spinal muscular atrophy. The mRNA vaccines, which saved millions of lives during the Covid pandemic, are being reformulated to attack other diseases, including some cancers . RNA research may also be helping us rewrite the future; the genetic scissors that give CRISPR its breathtaking power to edit genes are guided to their sites of action by RNAs.

Although most scientists now agree on RNA's bright promise, we are still only beginning to unlock its potential. Consider, for instance, that some 75 percent of the human genome consists of dark matter that is copied into RNAs of unknown function. While some researchers have dismissed this dark matter as junk or noise, I expect it will be the source of even more exciting breakthroughs.

We don’t know yet how many of these possibilities will prove true. But if the past 40 years of research have taught me anything, it is never to underestimate this little molecule. The age of RNA is just getting started.

Thomas Cech is a biochemist at the University of Colorado, Boulder; a recipient of the Nobel Prize in Chemistry in 1989 for his work with RNA; and the author of “The Catalyst: RNA and the Quest to Unlock Life’s Deepest Secrets,” from which this essay is adapted.

The Times is committed to publishing a diversity of letters to the editor. We’d like to hear what you think about this or any of our articles. Here are some tips . And here’s our email: [email protected] .

Follow the New York Times Opinion section on Facebook , Instagram , TikTok , WhatsApp , X and Threads .

Green fern on forest floor with brown leaves.

Tiny Fern Has World’s Largest Genome

A small South Pacific fern boasts more than 50 times as many base pairs as the human genome

Max Kozlov, Nature magazine

Illustration of active RNA molecules behind machines

Revolutionary Genetics Research Shows RNA May Rule Our Genome

Scientists have recently discovered thousands of active RNA molecules that can control the human body

Philip Ball

Two whiteflies against a green background

Stolen Bacterial Genes Helped Whiteflies to Become the Ultimate Pests

Rather than relying on bacteria, whiteflies cut out the middleman and acquired their own genes to process nitrogen

Rohini Subrahmanyam

Sugar glider, mid-air on black background

How Sugar Gliders Got Their Wings

Several marsupial species, including sugar gliders, independently evolved a way to make membranes that allow them to glide through the air

Viviane Callier

Top view of beetle.

Unraveling the Secrets of This Weird Beetle’s 48-Hour Clock

New research examines the molecular machinery behind a beetle’s strange biological cycle

Andrew Chapman

A seated woman in a courtroom holds a photo of family members

Forensic Genealogy Offers Families the Gift of Closure

The forensic scientist’s toolbox is growing thanks to creative methods that generate reliable leads, analyze evidence, identify suspects and solve cold cases

Nancy La Vigne

Plasmodium falciparum microscopic image.

Ancient Malaria Genome from Roman Skeleton Hints at Disease’s History

Genetic information from ancient Roman remains is helping to reveal how malaria has moved and evolved alongside people

Tosin Thompson, Nature magazine

Brown giant panda approaching on leafy ground.

Rare Brown Panda Mystery Solved after 40 Years

Chinese researchers have found the gene responsible for the brown-and-white fur of a handful of giant pandas

Xiaoying You, Nature magazine

Colorful DNA helix

What Do You Mean, Bisexual People Are ‘Risk-Taking’? Why Genetic Studies about Sexuality Can Be Fraught

A recent study on risk-taking and bisexuality made assumptions that some experts don’t agree with.

Tulika Bose, Lauren Leffer, Timmy Broderick

Coccyx, computer illustration

How Humans Lost Their Tails

A newly discovered genetic mechanism helped eliminate the tails of human ancestors

This Genetically Engineered Petunia Glows in the Dark and Could Be Yours for $29

The engineered “firefly petunia” emits a continuous green glow thanks to genes from a light-up mushroom

Katherine Bourzac, Nature magazine

Dalmation standing outside, head turns towards tail

Why Do Dogs Wag Their Tail?

Is your dog’s tail-wagging a side effect of domestication, or did humans select for it?

Tom Metcalfe

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Published: 24 August 2020

The road ahead in genetics and genomics

  • Amy L. McGuire 1 ,
  • Stacey Gabriel 2 ,
  • Sarah A. Tishkoff   ORCID: orcid.org/0000-0002-1339-5959 3 , 4 ,
  • Ambroise Wonkam   ORCID: orcid.org/0000-0003-1420-9051 5 , 6 ,
  • Aravinda Chakravarti   ORCID: orcid.org/0000-0002-4264-2285 7 ,
  • Eileen E. M. Furlong   ORCID: orcid.org/0000-0002-9544-8339 8 ,
  • Barbara Treutlein   ORCID: orcid.org/0000-0002-3299-5597 9 ,
  • Alexander Meissner   ORCID: orcid.org/0000-0001-8646-7469 2 , 10 , 11 , 12 ,
  • Howard Y. Chang   ORCID: orcid.org/0000-0002-9459-4393 13 ,
  • Núria López-Bigas   ORCID: orcid.org/0000-0003-4925-8988 14 , 15 , 16 ,
  • Eran Segal   ORCID: orcid.org/0000-0002-6859-1164 17 &
  • Jin-Soo Kim   ORCID: orcid.org/0000-0003-4847-1306 18  

Nature Reviews Genetics volume  21 ,  pages 581–596 ( 2020 ) Cite this article

81k Accesses

112 Citations

342 Altmetric

Metrics details

  • Genetic techniques

In celebration of the 20th anniversary of Nature Reviews Genetics , we asked 12 leading researchers to reflect on the key challenges and opportunities faced by the field of genetics and genomics. Keeping their particular research area in mind, they take stock of the current state of play and emphasize the work that remains to be done over the next few years so that, ultimately, the benefits of genetic and genomic research can be felt by everyone.

The contributors

Amy L. McGuire is the Leon Jaworski Professor of Biomedical Ethics and Director of the Center for Medical Ethics and Health Policy at Baylor College of Medicine. She has received numerous teaching awards at Baylor College of Medicine, was recognized by the Texas Executive Women as a Woman on the Move in 2016 and was invited to give a TedMed talk titled “There is No Genome for the Human Spirit” in 2014. In 2020, she was elected as a Hastings Center Fellow. Her research focuses on ethical and policy issues related to emerging technologies, with a particular focus on genomic research, personalized medicine and the clinical integration of novel neurotechnologies.

Stacey Gabriel is the Senior Director of the Genomics Platform at the Broad Institute since 2012 and has led platform development, execution and operation since its founding. She is Chair of Institute Scientists and serves on the institute’s executive leadership team. She is widely recognized as a leader in genomic technology and project execution. She has led the Broad’s contributions to numerous flagship projects in human genetics, including the International HapMap Project, the 1000 Genomes Project, The Cancer Genome Atlas, the National Heart, Lung, and Blood Institute’s Exome Sequencing Project and the TOPMed programme. She is Principal Investigator of the Broad’s All of Us (AoU) Genomics Center and serves on the AoU Program Steering Committee.

Sarah A. Tishkoff is the David and Lyn Silfen University Associate Professor in Genetics and Biology at the University of Pennsylvania, Philadelphia, USA, and holds appointments in the School of Medicine and the School of Arts and Sciences. She is a member of the US National Academy of Sciences and a recipient of an NIH Pioneer Award, a David and Lucile Packard Career Award, a Burroughs/Wellcome Fund Career Award and an American Society of Human Genetics Curt Stern Award. Her work focuses on genomic variation in Africa, human evolutionary history, the genetic basis of adaptation and phenotypic variation in Africa, and the genetic basis of susceptibility to infectious disease in Africa.

Ambroise Wonkam is Professor of Medical Genetics, Director of GeneMAP (Genetic Medicine of African Populations Research Centre) and Deputy Dean Research in the Faculty of Health Sciences, University of Cape Town, South Africa. He has successfully led numerous NIH- and Wellcome Trust-funded projects over the past decade to investigate clinical variability in sickle cell disease, hearing impairment genetics and the return of individual findings in genetic research in Africa. He won the competitive Clinical Genetics Society International Award for 2014 from the British Society of Genetic Medicine. He is president of the African Society of Human Genetics.

Aravinda Chakravarti is Director of the Center for Human Genetics and Genomics, the Muriel G. and George W. Singer Professor of Neuroscience and Physiology, and Professor of Medicine at New York University School of Medicine. He is an elected member of the US National Academy of Sciences, the US National Academy of Medicine and the Indian National Science Academy. He has been a key participant in the Human Genome Project, the International HapMap Project and the 1000 Genomes Project. His research attempts to understand the molecular basis of multifactorial disease. He was awarded the 2013 William Allan Award by the American Society of Human Genetics and the 2018 Chen Award by the Human Genome Organization.

Eileen E. M. Furlong is Head of the Genome Biology Department at the European Molecular Biology Laboratory (EMBL) and a member of the EMBL Directorate. She is an elected member of the European Molecular Biology Organization (EMBO) and the Academia Europaea, and a European Research Council (ERC) advanced investigator. Her group dissects fundamental principles of how the genome is regulated and how it drives cell fate decisions during embryonic development, including how developmental enhancers are organized and function within the 3D nucleus. Her work combines genetics, (single-cell) genomics, imaging and computational approaches to understand these processes. Her research has advanced the development of genomic methods for use in complex multicellular organisms.

Barbara Treutlein is Associate Professor of Quantitative Developmental Biology in the Department of Biosystems Science and Engineering of ETH Zurich in Basel, Switzerland. Her group uses and develops single-cell genomics approaches in combination with stem cell-based 2D and 3D culture systems to study how human organs develop and regenerate and how cell fate is regulated. For her work, Barbara has received multiple awards, including the Friedmund Neumann Prize of the Schering Foundation, the Dr. Susan Lim Award for Outstanding Young Investigator of the International Society of Stem Cell Research and the EMBO Young Investigator Award.

Alexander Meissner is a scientific member of the Max Planck Society and currently Managing Director of the Max Planck Institute (MPI) for Molecular Genetics in Berlin, Germany. He heads the Department of Genome Regulation and is a visiting scientist in the Department of Stem Cell and Regenerative Biology at Harvard University. Before his move to the MPI, he was a tenured professor at Harvard University and a senior associate member of the Broad Institute, where he co-directed the epigenomics programme. In 2018, he was elected as an EMBO member. His laboratory uses genomic tools to study developmental and disease biology with a particular focus on epigenetic regulation.

Howard Y. Chang is the Virginia and D. K. Ludwig Professor of Cancer Genomics at Stanford University and an investigator at the Howard Hughes Medical Institute. He is a physician–scientist who has focused on deciphering the hidden information in the non-coding genome. His laboratory is best known for studies of long non-coding RNAs in gene regulation and development of new epigenomic technologies. He is an elected member of the US National Academy of Sciences, the US National Academy of Medicine, and the American Academy of Arts and Sciences.

Núria López-Bigas is ICREA research Professor at the Institute for Research in Biomedicine and Associate Professor at the University Pompeu Fabra. She obtained an ERC Consolidator Grant in 2015 and was elected as an EMBO member in 2016. Her work has been recognized with the prestigious Banc de Sabadell Award for Research in Biomedicine, the Catalan National Award for Young Research Talent and the Career Development Award from the Human Frontier Science Program. Her research focuses on the identification of cancer driver mutations, genes and pathways across tumour types and in understanding the mutational processes that lead to the accumulation of mutations in cancer cells.

Eran Segal is Professor in the Department of Computer Science and Applied Mathematics at the Weizmann Institute of Science, heading a multidisciplinary laboratory with extensive experience in machine learning, computational biology and analysis of heterogeneous high-throughput genomic data. His research focuses on the microbiome, nutrition and genetics, and their effect on health and disease and aims to develop personalized medicine based on big data from human cohorts. He has published more than 150 publications and received several awards and honours for his work, including the Overton and the Michael Bruno awards. He was recently elected as an EMBO member and as a member of the Israel Young Academy.

Jin-Soo Kim is Director of the Center for Genome Engineering in the Institute for Basic Science in Daejon, South Korea. He has received numerous awards, including the 2017 Asan Award in Medicine, the 2017 Yumin Award in Science and the 2019 Research Excellence Award (Federation of Asian and Oceanian Biochemists and Molecular Biologists). He was featured as one of ten Science Stars of East Asia in Nature ( 558 , 502–510 (2018)) and has been recognized as a highly cited researcher by Clarivate Analytics since 2018. His work focuses on developing tools for genome editing in biomedical research.

Similar content being viewed by others

latest genetics research

A brief history of human disease genetics

latest genetics research

Molecular quantitative trait loci

latest genetics research

A new era in functional genomics screens

Making genomics truly equitable.

Amy McGuire. For the field of genetics and genomics, the first decade of the twenty-first century was a time of rapid discovery, transformative technological development and plummeting costs. We moved from mapping the human genome, an international endeavour that took more than a decade and cost billions of dollars, to sequencing individual genomes for a mere fraction of the cost in a relatively short time.

During the subsequent decade, the field turned towards making sense of the vast amount of genomic information being generated and situating it in the context of one’s environment, lifestyle and other non-genetic factors. Much of the hype that characterized the previous decade was tempered as we were reminded of the exquisite complexity of human biology. A vision of medicine driven by genetically determined risk predictions was replaced with a vision of precision in which genetics, environment and lifestyle all converge to deliver the right treatment to the right patient at the right time 1 .

As we embark on the third decade of this century, we are now faced with the prospect of being able not only to more accurately predict disease risk and tailor existing treatments on the basis of genetic and non-genetic factors but also to potentially cure or even eliminate some diseases entirely with gene-editing technologies.

These advancements raise many ethical and policy issues, including concerns about privacy and discrimination, the right of access to research findings and direct-to-consumer genetic testing, and informed consent. Significant investment has been made to better understand the risks and benefits of clinical genomic testing, and there has been vigorous debate about the ethics of human gene editing, with many prominent scientists and bioethicists calling for a moratorium on human germline editing until it is proven to be safe and effective and there is broad societal consensus on its appropriate application 2 .

These are all important issues that we need to continue to explore, but as the technologies that have been developed and tested at warp speed over the past two decades begin to be integrated into routine clinical care, it is imperative that we also confront one of the most difficult and fundamental challenges in genomics, in medicine and in society — rectifying structural inequities and addressing factors that privilege some while disadvantaging others. The genomics of the future must be a genomics for all, regardless of ethnicity, geography or ability to pay.

This audacious goal of making genomics truly equitable requires multifaceted solutions. The disproportionate burden of illness and death among racial and ethnic minorities associated with the global COVID-19 pandemic 3 and recent protests against police brutality towards African American citizens 4 have strengthened the antiracism movement and amplified demands for racial equity.

To be part of this movement and effect change will require humility. We must actively listen and learn from each other, especially when it is uncomfortable and our own complicity may be implicated. It will require solidarity and a recognition that we are all connected through our common humanity. And it will require courage. It may seem like a platitude, but it is true that nothing will change unless actual change is made. If we continue to do things as they have always been done, we will end up where we have always been. It is time to step into the discomfort and dare to do something different.

So what can we do differently to make genomics more equitable? I propose three areas where we should focus attention to address this important question. First, we must ensure equitable representation in genomic research. Examining 2,511 studies involving nearly 35 million samples from the GWAS Catalog in 2016, Popejoy and Fullerton found that the vast majority (81%) come from individuals of European descent, with only 5% coming from non-Asian minority populations 5 . This has created an ‘information disparity’ that has an impact on the reliability of clinical genomic interpretation for under-represented minorities 6 . The US National Institutes of Health (NIH) has invested in efforts to increase diversity in genomic research, but to be successful these efforts must be accompanied by serious attention to earning the trust of disadvantaged and historically mistreated populations. This will require, at a minimum, more meaningful engagement, improved transparency, robust systems of accountability, and a commitment to creating opportunities that promote and support a genomics workforce that includes scientists and clinicians from under-represented populations.

It is insufficient to achieve diverse representation in genomic research; however, there must also be equitable access to the fruits of that research. An analysis of the US Centers for Disease Control and Prevention’s 2018 Behavioural Risk Factor Surveillance System found that non-elderly adults from self-identified racial or ethnic minority groups are significantly less likely to see a doctor because of cost than non-elderly white adults 7 . This finding reflects how the structure and financing of health care in the United States perpetuates inequities and contributes to the larger web of social injustice that is at the heart of the problem. Even when socio-economic factors are controlled for, racial disparities in access to genetic services persist 8 . Large-scale, sustained research is needed to better understand and actively address the multitude of factors that contribute to this, including issues related to structural racism, mistrust, implicit and explicit bias, a lack of knowledge of genetic testing, and concerns about misuse of genetic information.

Finally, and perhaps most daunting, we must strive to achieve more equitable outcomes from genomic medicine. Many racial and ethnic minorities disproportionately experience chronic disease and premature death compared with white individuals. Disparities also exist by gender, sexual orientation, age, disability status, socio-economic status and geographical location. Health outcomes are heavily influenced by social, economic and environmental factors. Thus, although providing more equitable access to genomic services and ensuring more equitable representation in genomic research are necessary first steps, they are not enough 9 . Genomics can only be part of the solution if it is integrated with broader social, economic and political efforts aimed at addressing disparities in health outcomes. For genomics to be truly equitable, it must operate within a just health-care system and a just society.

we must strive to achieve more equitable outcomes from genomic medicine

Genome sequencing at population scale

Stacey Gabriel. Twenty years ago, I finished a PhD project that involved laboriously sequencing one gene — a rather complicated one, RET — in a couple of hundred people to catalogue pathogenic variants for Hirschsprung disease. This work required designing primers on the basis of genome sequence data as they were gradually released, amplifying the gene exon by exon (all 20!), running sequencing gels and manually scoring sequence changes. The notion of sequencing the whole genome to catalogue sequence changes was something to wish for in our wildest dreams.

Thanks to great strides in technology and the hard work of geneticists, engineers, epidemiologists and clinicians, much progress has been made; huge numbers of genomes (and exomes) have been sequenced across the world. Disease gene-finding projects such as my graduate work are now done routinely, rather than one gene at a time, using whole-exome or whole-genome sequencing (WGS) in families and affected individuals, enabling the identification of genes and causative mutations in thousands of Mendelian diseases and some complex diseases.

But the real promise of genome sequencing lies in true population-scale sequencing, ultimately at the scale of tens of millions of individuals, whereby genome sequencing of unselected people enables the unbiased, comprehensive study of our genome and the variation therein. It provides a ‘lookup table’ to catalogue disease-causing and benign variants (our ‘allelic series’). The genome sequence should become part of the electronic health record; it is a stable, persistent source of information about a person akin to physical measurements such as weight or blood pressure, exposures such as smoking or alcohol use, and (in many ways better than) self-reported family history.

the real promise of genome sequencing lies in true population-scale sequencing, ultimately at the scale of tens of millions of individuals

What can we learn? What needs to be solved? Even fairly small numbers of genomes aggregated in a consistent and searchable form have enabled a new way to use and interpret genomic data, just in the past couple of years providing a glimpse at the future. Efforts such as gnomAD 10 are a start — this database contains data from more than 15,000 genomes and 125,0000 exomes. With this resource, the frequency of genetic variants within populations is readily available. A clinician interpreting the genome of a patient can ask whether a variant has been observed before. The data provide a starting point for assessing the functional impact of classes of genetic variation and the ability to ask questions about ‘missing’ genetic variation where there is constraint.

Coupled with clinical data, building up population-scale databases of genomic plus clinical information will fuel the application of better risk interpretation using polygenic risk scores (PRSs) 11 . More routine WGS will shorten the ‘diagnostic odyssey’, in which patients suffer through rounds of testing and parents are left uncertain about future reproductive planning. More efficient clinical trials might be built using genomic information. With existing genomic information on all individuals in a health system, trials could be designed in a way that selects individuals most likely to have an event. This enrichment could provide more promising, shorter, smaller and cheaper trial design.

These databases must also rapidly be built in such a way that is representative of the population, representing the actual racial and ethnic diversity, not just what was available as banked sample collections. These are well known to be predominantly European-descent samples and thus preclude application of risk prediction tools in non-white individuals and have limited the ability to find population-specific genetic associations, such as those that have been demonstrated in type 2 diabetes mellitus (T2DM) 12 .

We have to solve important issues — data sharing, privacy and getting the data to scale. Sharing genomic and clinical data is of key importance to drive forward discovery and our understanding of how to use these data in the health-care setting. To do this well and responsibly, trust must be built and maintained through adherence to the rights of privacy, protection and non-discrimination. Progress is being made through the creation of data platforms and the development of frameworks for data protection and sharing; for example, by the work of the Global Alliance for Genomics and Health (GA4GH).

Several large biobanks are already being established to launch population-scale efforts. The UK Biobank is a vanguard programme that contains genotype data, questionnaire-based health and physical measurements on 500,000 individuals and some linkage to their medical records. Other efforts such as the All of Us research programme have been launched with goals directed at true population-based representation, and biobanks that link genomic data to comprehensive medical records in specific health-care systems (for example, Geisinger) or in specific countries or regions (for example, Estonia and Iceland) are also under way.

A big piece of this puzzle is generating comprehensive genome sequence data in these programmes and far beyond. For this aim, large-scale, affordable sequencing is key. No problem, right? Is sequencing not always getting cheaper? The problem is that this assumption is no longer true. We have got to where we are today because for a long time, from 2008 to 2013, sequencing costs dropped exponentially. However, in recent years, the sequencing cost curve has flattened, as is apparent in publicly reported cost estimates provided by the US National Human Genome Research Institute 13 . The cost per megabase of sequence data has remained largely unchanged since around 2016, hovering around a list price of US$0.01 per megabase, which translates to a US$1,000 genome. Gone are the days of our field touting the impressive decrease of cost in comparison with Moore’s law, and this development is worrying.

Some discounting does happen at considerable volume, and whole genomes can be priced in the range of US$500 to US$700. However, large projects (more than 500,000 samples) sequenced at these prices are few and far between, and are generally dependent on pharmaceutical or biotech funding, which can bring with it restrictions on data sharing. It is my belief that a fivefold to sevenfold reduction in total costs is needed to unlock more sequencing at the population scale and, ultimately, for genome sequencing to be more widely applied in the health-care setting. At US$100 per genome, the cost represents less than 1% of the annual average health-care expenditure per person in the United States, and a genome sequence is a one-time investment that can be referenced again and again over the entire lifespan of a person. Getting that cost curve down will be important to inspire health-care systems to adopt genome sequencing routinely.

I see three main drivers that will get us to US$100 per genome: innovation, scale and competition.

Innovation . Generating sequence data requires multiple components, and there are multiple areas ripe for innovation. Sample preparation can be improved through more efficient methods that decrease the labour required, or miniaturization can decrease the cost of the reagents used in library preparation. Developments to decrease data processing costs are also ripe for innovation. Recently, we showed that processing using optimized computing power lowered the time and cost of creating a sequence file by ~50% (S.G., unpublished observations). While decreases in the costs of sample preparation and data processing are important, they represent a small component of the total cost. Roughly 70% of the cost of sequencing a human genome is the sequencing reagent (flow cell) and the instrument. Appreciable cost decrease is made possible only by decreasing these marginal costs, as was demonstrated in the period from 2010 to 2014, when flow-cell densities doubled and sequencing cost dropped by an order of magnitude (US$100 per gigabase to US$10 per gigabase).

Scale . One component of cost is the fixed cost borne by the sequencing centre or the sequencing vendor. With high scale, centres can become more efficient and offset costs such as the costs of personnel, equipment and facilities. Scale can also result in volume discounting of the reagents, although this process is tightly controlled and approached cautiously depending on overall market dynamics.

Competition . Innovation and scale can only achieve so much. The cost of generating the data (the cost per gigabase) dominates and thus must come down considerably. The current market requires alternative options to drive this advance. Presently, the market for short-read sequencing is lacking viable, proven competition that would force flow-cell densities and machine yield to be increased and put pressures on volume discounting. While options for long-read sequencing exist and play a role in particular applications, such as de novo sequencing and structural variant resolution, they are at present far from cost competitive and, therefore, do not apply pressure to bring down the cost of routine WGS.

We need innovation, great economies of scale and/or real competition to come to play in the marketplace. When it comes to sequencing technology, particularly at a large scale, we cannot be complacent and work around the current barriers to realize small gains and one-off wins. This might involve specific types of investment beyond just financial ones; adopting and vetting new technology requires time, creativity, commitment and patience. It is a challenge for our community to take on now. In 5 years’ time, I hope we can look back at the era of the US$100 genome and progress towards real population-scale databases that fuel discovery, enriching our knowledge of the human allelic series and, importantly, the routine use of genomic data in the health-care setting.

A global view of human evolution

Sarah Tishkoff. The past 10 years saw an exponential increase in SNP array and high-coverage WGS data owing to innovations in genomic technologies. It is now possible to generate WGS data from tens of thousands of individuals (for example, GenomeAsia 100K 14 and NIH TOPMed 15 ). An increase in medical biobanks with access to electronic health records (for example, the UK Biobank 16 , the Million Veteran Project 17 and BioBank Japan 18 ) is enabling the mapping of hundreds of genetic associations with complex traits and diseases, as well as phenome-wide association studies 19 to map pleiotropic associations of phenotypes with genes. The genetic associations identified in these and other studies have been used to calculate PRSs for predicting complex phenotypes and risk of diseases.

Yet despite these advances, as of 2019, nearly 80% of individuals in genome-wide association studies (GWAS) were of European ancestries, ~10% were of East Asian ancestries, ~2% were of African ancestries, ~1.5% were of Hispanic ancestries and less than 1% were of other ancestries 20 . There is also a strong European bias in genomic reference databases, such as gnomAD and GTEx . These biases limit our knowledge of genetic risk factors for disease in ethnically diverse populations and could exacerbate health inequities 20 . Furthermore, PRSs that were estimated using European data do not accurately predict phenotypes and disease risk in non-European populations, performing worst in individuals with African ancestry 21 . The lack of transportability of PRSs across ethnic groups is likely due to differences in patterns of linkage disequilibrium and haplotype structure (resulting in different SNPs tagging causal variants), differences in allele frequencies, gene × gene effects and gene × environment effects. It is also possible that the genetic architecture of complex traits and diseases may differ across ethnic groups owing to different demographic histories and adaptation to diverse environments.

Although there have been initiatives to increase inclusion of ethnically diverse populations in human genomics research (for example, the NIH TOPMed 15 and H3Africa consortia), Indigenous populations remain under-represented. Great care must be taken to ensure that genomic research of minority and Indigenous populations is conducted in an ethical manner. This involves establishing partnerships with local research scientists, being sensitive to local customs and cultural concerns, obtaining both community and individual consent, and returning results to communities that participated when possible. In addition, there should be training and capacity building so that genomic research can be conducted locally, where feasible.

A particular area of focus in the future should be developing tools and resources that make genomic data and analyses accessible in low- and middle-income countries. We have to ensure that all people benefit from the genomics revolution and advances in precision medicine and gene editing. Thus, several of the biggest challenges in the next decade will be (1) to increase inclusion of ethnically diverse populations in human genomics research; (2) the generation of more diverse reference genomes using methods that generate long sequencing reads, and haplotype phasing, to account for the large amount of structural variation that likely exists within and between populations; (3) the training of a more diverse community of genomic research scientists; and (4) the development of better methods for accurately predicting phenotypes and genetic risk across ethnically diverse populations and for distinguishing gene × environment effects.

The inclusion of ethnically diverse populations, including Indigenous populations, is also critical for reconstructing human evolutionary history and understanding the genetic basis of adaptation to diverse environments and diets. While there have been a number of success stories for identifying genes of large effect that play a role in local adaptation (for example, lactose tolerance and sickle cell disease (SCD) associated with malaria resistance), identifying signatures of polygenic selection has been considerably more challenging 22 . Genomic signatures of polygenic adaptation are based on the ability to detect subtle shifts in allele frequencies at hundreds or thousands of loci with minor effect on the phenotype of a complex trait and to determine whether that shift is a result of demography or natural selection. A more daunting challenge arises from the same issues of portability of PRSs described earlier — variants associated with a complex trait may not tag well across ethnic groups and/or the genetic architecture of a trait may differ in different populations. Furthermore, it has recently been shown that uncorrected population stratification can result in a false signal of polygenic selection 23 . For example, several studies have identified signatures of polygenic adaptation for height across European populations (selection for increased height in northern Europeans and for decreased height in southern Europeans). However, it was recently shown that these results were influenced by population structure that could not be easily corrected using standard approaches, particularly for SNPs below genome-wide levels of significance 23 . When this analysis was repeated with variants identified in a more homogenous set of individuals of European ancestry from the UK Biobank, these signatures of polygenic adaptation were erased 23 . Thus, methods for detecting polygenic adaptation that are less biased by population structure and by population ascertainment bias will need to be developed in the future. These studies will also benefit from inclusion of more ethnically diverse populations in GWAS and identification of better tag SNPs as described earlier. A challenge of inclusion of minority populations in GWAS is that sample sizes are often small relative to majority populations. However, the high levels of genetic diversity and extremes of phenotypic diversity observed in some populations, particularly those from Africa, make them particularly informative for GWAS. For example, a GWAS of skin pigmentation in fewer than 1,600 Africans was informative for identifying novel genetic variants that affect skin colour, including a previously uncharacterized gene, MFSD12 (ref. 24 ). Thus, genomic studies in the future must make inclusion of minority populations a priority.

A challenge in both GWAS and selection scans has been the identification of causal genetic variants that directly have an impact on variable traits. Most of these variants are in non-coding regions of the genome. The development of high-throughput approaches, such as massively parallel luciferase expression assays to identify gene regulatory regions and high-throughput CRISPR screens in vitro and in vivo to identify functional variants influencing the trait of interest, will be useful 25 . There is also a need to better understand cell type-specific variation and gene regulation at the single-cell level, including response to stimuli such as immune, pharmacological and nutrient challenges, in ethnically diverse populations. However, these approaches are still limited by the need to have informative cell lines. This can be particularly challenging to obtain for Indigenous populations living in remote regions. Improvements in the differentiation of induced pluripotent stem cells (iPS cells) into assorted cell types and into organoids will be important for facilitating functional genomic studies. Establishment of iPS cells and organoids from diverse non-human primate species will also be informative for comparative genomic studies to identify the evolution of human-specific traits such as brain development and cognition. However, iPS cell-derived cells may not accurately reflect the impact of mutations acting on developmental phenotypes, which will require development of more efficient in vivo approaches in model organisms.

Perhaps the biggest revolution in the study of recent human evolutionary history has been the development of methods that make it feasible to sequence and/or obtain targeted genotypes from ancient DNA samples. The generation of high-coverage reference genomes for archaic hominid species such as Neanderthals and Denisovans, located in Eurasia, has made it feasible to identify archaic introgressed segments within the genomes of non-Africans. Some of these regions have been shown to play a role in adaptive traits such as adaptation to high altitude and immune response 26 . Furthermore, there has been an explosion of studies of ancient genetic variation in Europeans within the past 30,000 years that has demonstrated a much more complex model of the peopling of Europe, and the recent evolution of adaptive traits, than previously known from the archaeological record or from studies of modern populations 27 . The biggest challenge has been the inability to get high-quality ancient DNA from regions with a tropical climate, such as Africa and Asia. While there has been success in analysing DNA samples as old as 15,000 years in Africa, which has been informative for tracing recent migration and admixture events 28 , the lack of a more ancient African reference genome makes it very challenging to detect archaic introgression, which currently relies on statistical modelling approaches. Thus, the biggest challenge in the next 10 years will be the successful sequencing of ancient DNA more than 20,000 years old from all regions of the world, so that we may have a better understanding of the complex web of population histories from across the globe.

African genomics — the next frontier

Ambroise Wonkam. To fully meet the potential of global genetic medicine, research into African genomic variation is a scientific imperative, with equitable access being a major challenge to be addressed. Studying African genomic variation represents the next frontier of genetic medicine for three major reasons: ancestry, ecology and equity.

On the basis of a ‘pan-genome’ generated from 910 individuals of African descent, at least 300 million DNA variants (10%) are not found in the current human reference genome 29 , and 2–19% of the genome of ancestral Africans derives from poorly investigated archaic populations that diverged before the split of Neanderthals and modern humans 30 . Neanderthal genome contributions make up ~2% of the genome in present-day Europeans and are enriched for variations in genes involved in dermatological phenotypes, neuropsychiatric disorders and immunological functions 31 . Once technical challenges in sequencing poor-quality DNA have been overcome and approaches to investigate the genomic contribution of African archaic populations have been refined, it is likely that associations between variants in ancient African DNA and human traits or diseases will be found, providing insights that can benefit modern-day humans.

As a consequence of the 300,000–500,000 years of genomic history of modern humans in Africa, ancestral African populations are the most genetically diverse in the world. By contrast, there is an extreme genetic bottleneck, resulting in much less variation, in all non-African populations who evolved from the thousands of humans who migrated out of Africa approximately 70,000 years ago. Current PRSs, which aim to predict the risk for an individual of a specific disease on the basis of the genetic variants that individual harbours, exhibit a bias regarding usability and transferability across populations, as most PRSs do not account for multiple alleles that are either limited or of high frequency among Africans. A GWAS on the genetic susceptibility to T2DM identified a previously unreported African-specific significant locus, while showing transferability of 32 established T2DM loci 32 . In addition, nonsense mutations found commonly among Africans in PCSK9 , which are rare in Europeans 33 , are associated with a 40% reduction in plasma levels of low-density lipoprotein, supporting PCSK9 as a target for dyslipidaemia therapeutics. In the largest GWAS meta-analysis for 34 complex traits, conducted in 14,345 Africans, several loci had limited transferability among cohorts 34 , further illustrating that genomic variation is highest among Africans compared with other populations. As a consequence, linkage disequilibrium is lower in Africans, which improves fine mapping and identification of causative variants. Indeed, while only 2.4% of participants in large GWAS are African individuals, they account for 7% of all associations 35 . Moreover, whole-exome sequencing of nearly 1,000 African study participants of Xhosa ancestry with schizophrenia found very rare damaging mutations in multiple genes 36 , a finding that could be replicated in a Swedish cohort of 5,000 individuals. In comparison, results for the Xhosa cohort yielded larger effect sizes, which shows that for the same number of cases and controls, the greater genetic variation in African populations provides more power to detect genotype–phenotype relationships. Therefore, millions of African genomes must be sequenced, with genotyping and analysis tools optimized for their interrogation.

Greater availability of African genomes will improve our understanding of genomic variation and complex trait associations in all populations but will also support research into common monogenic diseases. The discovery of a single African origin of the SCD mutation, about 5,000–7,000 years ago, not only suggested recent migration and admixture events between Africans and Mediterranean and/or Middle Eastern populations but also enhanced our understanding of genetic variation in general as well as its potential impact on haemoglobinopathies 37 . For example, variants in the HBB -like gene cluster linked with high levels of fetal haemoglobin have been associated with less severe SCD; because the level of fetal haemoglobin is under genetic control, it is amenable to therapeutic manipulation by gene editing 38 . Moreover, knowledge of an individual’s genetic variants can have an impact on secondary prevention of and treatment strategies for SCD. For example, variants in APOL1 and HMOX1 and co-inheritance of α-thalassaemia are associated with kidney dysfunctions 39 ; stroke in SCD is associated with targeted genetic variants used in a Bayesian model; and overall SCD mortality has been associated with circulating transcriptomic profiles. It is estimated that 75% of the 305,800 babies with SCD born each year are born in Africa; SCD in Africa will serve as a model for understanding the impact of genetic variation on common monogenic traits and help to illustrate the multiple layers of genomic medicine implementation.

Greater availability of African genomes will improve our understanding of genomic variation and complex trait associations in all populations

Exploring African genomic diversity will also increase discovery of novel variants and genes for rare monogenic conditions. Indeed, allelic and locus heterogeneity display important differences in African individuals compared with other populations; for example, mutations in GJB2 account for nearly 50% of cases of congenital non-syndromic hearing impairment among Eurasians but are nearly non-existent in Africans, and there is evidence that novel variants in hearing impairment-associated genes are more likely to be found in Africans than in populations of European or Asian ancestries 40 . Higher fertility rate, consanguinity practices and regional genetic bottlenecks will improve novel gene discovery for monogenic diseases in Africa, as well as disease–gene pair curation, and will address existing challenges surrounding database biases and inference of variant deleteriousness, which have led to the misclassification of variants.

Differential population genomic variant frequencies are shaped by natural evolutionary selection as an adaptation to environmental pressures. The African continent follows a North–South axis, which is associated with variable climates and biodiversity, both motors of natural selection. This specific African ecology has shaped genetic variation accordingly, which can have a detrimental or positive impact on health. Obvious examples are variants that cause SCD but confer resistance to malaria 37 , APOL1 variants that are protective against trypanosomes (the parasites that cause sleeping sickness) 41 and variants of OSBPL10 and RXRA that protect against dengue fever 42 . Unfortunately, APOL1 variants also increase susceptibility to chronic kidney disease in populations of African ancestry 39 , 41 . A better understanding of the functional impact of genetic variants specific to African populations, particularly those that have been selected under environmental pressure, and the way they interact with each other is needed and will have a positive impact on genetic medicine practice. Moreover, immunogenetic studies among Africans will further our understanding of natural selection and responses to emerging infectious diseases, such as COVID-19.

The scientific imperative of genomic research of African populations is expected to enhance genetic medicine knowledge and practice in Africa but will face the challenges of overburdened and under-resourced public health-care systems, and often absent ethical, legal and social implication frameworks 43 , requiring international collaboration to be managed. Developing an African genomics workforce will be necessary to meet the major need for research across the lifespan for cohorts of millions of individuals with complex or monogenic diseases. Such endeavours can thrive on the foundation of recently established initiatives such as H3Africa. Indeed, equitable access for Africans is essential if African genomics is to reach its full potential as the next frontier of global genetic medicine.

Decoding multifactorial phenotypes

Aravinda Chakravarti. We live in a time of great technological progress in genomics and computing. And we live in a time when ‘genetics’ is a household word, with a public increasingly adept at understanding its relevance to their own lives. Not surprisingly, the study of genetics is being reinvented, rediscovered and reshaped, and we are beginning to understand the science of human heredity at a resolution that was impossible before.

The most significant genetics puzzle today, in my view, is the dissection of ‘family resemblance’ of complex phenotypes, both for intellectual (raison d'être of genetics) and practical (disease diagnosis and therapy) reasons. We have long known that family resemblance arises from shared alleles, declining as genetic relationship wanes, but the precise molecular components and composition of this resemblance are still poorly understood. At the turn of the twentieth century, the components were a matter of bitter and acrimonious debate 44 between the ‘Mendelians’ and the ‘Biometricians’, until the opposing views were reconciled by Ronald Fisher’s 1918 analysis 45 that complex inheritance could be explained through segregation of many genes, each individually Mendelian. In 1920, its publication delayed by World War I, this notion was elegantly demonstrated by the experimental studies of Altenburg and Muller using truncate wing , an “inconstant and modifiable character” 46 in Drosophila .

Fisher’s model assumed an infinite number of genes additively contributing to a trait, with common genetic variation at each component locus comprising two alleles that differ only slightly in their genetic effects 45 ; these genetic assumptions were quite contrary to what was then known 44 . Throughout the past century, this view matured, as segregation analyses of human phenotypes taught us that — beyond the effects of some major genes — most trait variation was polygenic, modulated by family-specific and random environmental factors 47 . Today, we have empirical evidence from GWAS, which use dense maps of genetic variants on hundreds of thousands of individuals measured for many traits and diseases, that the genetic architecture of most multifactorial traits is from common sequence variants with small allelic differences at thousands of sites across the genome 48 . This replacement of a pan-Mendelian view with a pan-polygenic view of traits is one of the most important contributions of genomics to genetics. Unfortunately, this mapping success has not clarified the number of genes involved, the identity of those genes or how those genes specify the phenotype. Indeed, some have concluded that many of the mapped GWAS loci are unrelated to the core biology of each phenotype 49 . Thus, for a deeper understanding, we need radically different approaches to understand complex trait biology in contrast to merely expanding GWAS in larger and larger samples.

for a deeper understanding, we need radically different approaches to understand complex trait biology

Yet, the most significant biology to emerge from GWAS is that most of the likely trait-causing variants fall outside coding sequences, in regulatory elements, most frequently enhancers 50 , 51 . This important finding has uncovered four new genetic puzzles. First, the non-coding regulatory machinery is vast; how much of this regulation is compromised, and how does it affect phenotypes? Second, regulatory changes affect RNA expression at many genes and protein expression at others; how does a cell ‘read’ these numerous changes as specific signals? Third, how is this coordinated expression response translated into cellular responses affecting phenotypes? Fourth, if specific environmental factors affect the same phenotype, which components do they dysregulate? In my opinion, we need to answer these questions for specific traits and diseases to truly understand their polygenic biology. Finally, these explanations must also answer the question of why some traits are decidedly Mendelian whereas others are not.

The questions of tomorrow will need to focus on four areas: the biology of enhancers and the transcription factors that bind them 51 ; the effect of genetic variation in enhancers 50 ; gene regulatory networks (GRNs) that regulate expression of multiple genes 52 ; and how GRN changes lead to specific cellular responses 53 . Despite many advances, the number of enhancers regulating expression of a specific gene remains unknown. How many enhancers are cell type specific versus ubiquitous? How many are constitutive rather than stage specific? And do they act additively or synergistically in gene expression? Additionally, which cognate transcription factors bind these enhancers, with what dynamics and how are they regulated 54 ? These details of a gene’s ‘enhancer code’ are critical for assessing its relative effect on a trait. Next, how does enhancer sequence variation affect a gene’s activity? Does such variation affect transcription factor binding only or its interaction with the promoter? Is the enhancer variant’s effect evident in all cellular states or only some? Is variation in only one enhancer sufficient to alter gene expression, or are multiple changes in multiple elements necessary?

Additional critical questions include which genes are involved in the core pathway underlying a trait, and how do we identify them 49 ? Elegant work has shown how genes are regulated within integrated modular GRNs, whereby one gene’s product is required in a subsequent step by another gene, with feedback interactions 52 . These GRNs comprise elements from the genome, transcriptome and proteome, with rate-limiting steps that require regulation. As our work on Hirschsprung disease has shown 50 , 53 , a GRN is composed of core genes, is the logic diagram of regulation of a major rate-limiting cellular step, is enriched in coding and enhancer disease variants with disease susceptibility scaling with increasing number of variants, and with disease resulting from effects on its rate-limiting gene product 53 . That is, the GRN integrates the expression of multiple genes. Finally, we need to understand how GRN changes alter cell properties and behaviour. I speculate that rate-limiting steps in GRNs are major regulators of broad cell properties, be they differentiation, migration, proliferation or apoptosis, the cellular integrator of GRN variation. Thus, genetic variation across the genome affects enhancers dysregulating many genes, but only when they dysregulate GRNs through rate-limiting steps do they affect cell and tissue biology 55 . This offers the promise of a mechanistic understanding of human polygenic disease.

The way forward for complex trait biology, including disease, is to shift our approach from reverse to forward genetics, using genome-wide approaches to cell type-specific gene perturbation. I believe we can construct cell-type GRNs en masse, inclusive of their enhancers, transcription factors and feedback or feedforward interactions, to then assay functionally defined variation in phenotypes. But, even this approach will be insufficient. We need to test our success by solving at least a few complex traits completely and demonstrating their veracity using a synthetic biology approach to recapitulate the phenotype in a model system; similarly to the field of chemistry, analysis has to be followed by de novo synthesis. Our genomic technologies are getting up to the task to enable this advance; as geneticists, are we?

Enhancers and embryonic development

Eileen Furlong. The work of my group sits at the interface of genome regulation and animal development, and there have been many exciting advances in both during the past decade. Developmental biology studies fundamental processes such as tissue and organ development and how complexity emerges through the combined action of cell communication, movement and mechanical forces. After the discovery that differentiated cells could be reprogrammed to a naive embryonic stem cell-like state, the past decade has witnessed an explosion in in vitro cellular reprogramming and differentiation studies. Organoids are a very exciting extension of this. The extent to which these fairly simple systems can self-organize and generate complexity 56 is one of the unexpected surprises of the past 5–10 years. The buzz around stem cells has also renewed interest in cellular plasticity in vivo and has uncovered an unexpected degree of transdifferentiation and dedifferentiation 57 . In the mouse heart, for example, cardiomyocytes dedifferentiate and proliferate to regenerate heart tissue when damaged within the first week after birth 58 .

Our understanding of the molecular changes that accompany differentiation has hugely advanced owing to the jump in scale, resolution and sensitivity of next-generation sequencing technologies over the past decade. This has led to a flood of studies in embryonic stem cells, iPS cells and embryos that revealed new concepts underlying genome regulation by measuring transcript diversity, transcription factor occupancy, chromatin accessibility and conformation, and chromatin, DNA and RNA modifications. The future challenge will be to connect this information to the physical characteristics of cells and how they form complex tissues. New technologies that solve many challenges of working with embryos will help, including CRISPR to engineer genomes, optogenetics to perturb proteins, lattice light-sheet and selective plane illumination microscopy to image processes in vivo, and low-input methods to overcome issues with scarce material. Particularly exciting to me are recent advances in single-cell genomics, which, although they are in their early days, will dramatically change the way we study embryogenesis. Many new insights have already emerged, including the discovery of unknown cell types and new developmental trajectories for well-established cell types. Even the concept of ‘cell identity’ has come into question.

Cell identities are largely driven by transcription factors, which act through cis -regulatory elements called ‘enhancers.’ One of the most exciting unsolved mysteries, in my opinion, is how enhancers relay information to their target genes. The textbook view of enhancers is of elements with exclusive function that regulate a specific target gene through direct promoter interactions, which occur sequentially if multiple enhancers are involved. However, emerging concepts in the past decade question many of these ‘dogmas’. Some enhancers have dual functions, whereas others may even regulate two genes. Enhancer–promoter communication is now viewed in the light of spatial genome organization, including topologically associating domains (TADs) and membraneless nuclear microcompartments (that is, hubs or condensates) 59 . Being present within the same TAD likely increases the frequency of enhancer–promoter interactions, but how a specific enhancer finds its correct promoter within a TAD, or when TADs are rearranged 60 , 61 , remains a mystery. Hubs or condensates are dynamic microcompartments 62 that contain high local concentrations of proteins, including transcription factors and the transcriptional machinery. One potential implication of condensates is that enhancers may not need to ‘directly’ touch a gene’s promoter to regulate transcription — rather, it may be sufficient to come in close proximity within the same condensate. Presumably, once proteins reach a critical concentration, transcription will be initiated. While this model fits a lot of emerging data, there are still many open questions. What is the required distance between an enhancer and a promoter to trigger transcription? Does this distance differ for different enhancers 63 depending on their transcription factor–DNA affinities? Do different chromatin environments 64 influence the process? At some loci, mutation of a single transcription factor-binding site in a single enhancer can have dramatic effects on gene expression and development. It is difficult to reconcile such cases with a shared condensate model, as other proteins bound to the enhancers and promoter should still phase separate. By contrast, there are many examples where mutation of a single transcription factor-binding site, or even an entire enhancer, has minimal impact on the expression of a gene. These observations suggest that there may be different types of loci, with requirements for different types of chromatin topologies and local nuclear environments, which will be important to tease apart in the coming years.

The genetic dissection of model loci in the 1990s and the first decade of the twenty-first century led to much of our understanding of how genes are regulated. The power of genomics in the past few decades has captured regulatory information for all genes genome-wide, providing more unbiased views of regulatory signatures, leading to new models of gene regulation. What is missing is empirical testing at a large scale. A major challenge is to move to more systematic in vivo functional dissection in organisms. CRISPR-based pooled screens have advanced the interrogation of genomic regions in cell culture systems. However, scaling functional assays in embryos remains a huge challenge. The task is enormous — even long-standing model organisms, such as Drosophila and mice, lack knockout strains for all protein-coding genes, and the number of regulatory elements is at least an order of magnitude higher. There has been little progress in developing scalable methods to quantify the contribution of a transcription factor’s input to an enhancer’s activity, and gene expression, in embryos. More systematic unbiased data will uncover more generalizable regulatory principles, increase our predictive abilities of gene regulation and developmental programmes, and enhance our understanding of the impact of genetic variation.

A major challenge is to move to more systematic in vivo functional dissection in organisms

Perhaps the most promising and exciting prospects in the coming years are to use single-cell genomics, imaging and the integration of the two to dissect the amazing complexity of embryonic development. Single-cell genomics can reveal information about developmental transitions in a way that was unfeasible before. When combined with temporal information, such data can reconstruct developmental trajectories 65 , 66 and identify the regulatory regions and transcription factors likely responsible for each transition 67 . The scale and unbiased nature of the data, profiling tens to hundreds of thousands of cells, provides much richer information than anyone envisaged just 5 years ago, bringing a new level of inference and causal modelling. The ability to measure single-cell parameters in situ (called ‘spatial omics’) will be transformative in the context of developing embryos to reveal the functional impact of spatial gradients, inductive signals and cell–cell interactions, and to move to digital 4D embryos. Combining these approaches with genetic perturbations holds promise to decode developmental programmes as they unfold. Will this bring us to a predictive understanding of the regulatory networks driving embryonic development during the next decade? ‘Simple’ model organisms are a fantastic test case to determine the types and scale of data required and to develop the computational framework to build predictive networks. The systematic functional dissection of gene regulation and true integration of single-cell genomics with single-cell imaging will bring many exciting advances in our understanding of the programmes driving embryonic development in the coming years.

Spatial multi-omics in single cells

Barbara Treutlein. Incredibly, the first single-cell transcriptome was sequenced just over a decade ago 68 ! Since this milestone, transcriptomes of millions of cells have been sequenced and analysed from diverse organisms, tissues and other cellular biosystems, and these maps of cell states are revolutionizing the life sciences. The technologies and associated computational methods have matured and been democratized to such an extent that nearly all laboratories can apply the approach to their particular system or question.

Of course, the transcriptome is not enough, and protocols have already been developed to measure chromatin accessibility, histone modifications, protein abundances, cell lineages and other features linked to genome activity in single cells 69 . Currently, many studies use dissociation-based single-cell genomics methods, where the spatial context is disrupted to facilitate the capture of single cells for downstream processing. Methods are improving to measure genomic features in situ 70 , as well as to computationally map features to spatial contexts 71 , 72 . The stage is set for the next phase of single-cell genomics, where spatial registration of multimodal genome activity across molecular, cellular and tissue or ecosystem scales will enable virtual reconstructions with extraordinary resolution and predictive capacity. These virtual maps will rely on multi-omic profiling of healthy and perturbed tissues and organisms, which presents major challenges and opportunities for innovation.

Cell throughput remains a challenge, and it is unclear what role dissociation-based single-cell sequencing protocols will play in the future. These protocols are fairly easy to implement, and laboratories around the world can execute projects with tens of thousands of cells analysed per experiment. However, there are scenarios in which measuring millions of cells per experiment would be desired, such as in perturbation screens. Combinatorial barcoding methods push cell-throughput boundaries 73 ; however, it is unclear how to scale full transcriptome sequencing economically to millions of cells using current sequencing technologies. ‘Compressed sensing’ modalities — whereby a limited, selected and/or random number of features are measured per cell, and high-dimensional feature levels are recovered through inference or similarity to a known reference — provide an interesting possibility to increasing cell throughput 74 .

Most single-cell transcriptome protocols are currently limited to priming the polyadenylation track present on all cellular mRNAs; however, this approach leads to biased sampling of highly expressed mRNAs. Clever innovations for random or targeted RNA enrichment could be a way to build up composite representations of cell states. Image-based in situ sequencing methods provide a means for increasing the number ofcells measured per experiment, as millions of cells can be imaged without a substantial increase in financial cost, although imaging time is a limiting factor. There remains a lot of room for experimental and computational optimizations to measure the transcriptome, random barcodes, DNA conformations and protein abundances from the micrometre scale to the centimetre scale spatially, and it will be interesting to see how methods for spatial registration advance over the next 5 years.

Currently, most high-throughput measurements are performed on cell suspensions or on intact tissues using one modality. That said, studies are emerging that measure several features from the same cell; for example, mRNA and chromatin accessibility 75 or mRNA and lineage 76 . To build virtual maps, independent measurements from different cells can be integrated with use of data integration tools 77 , although it can be difficult to align cell states across modalities in particular in developing systems. Therefore, the ultimate goal is to directly measure as many features as possible (for example, RNA, lineage, chromatin, proteins and DNA methylation) in the same cell 78 , ideally with spatial resolution. Furthermore, combining genetic and pharmacological perturbation screens with single-cell multi-omic measures will be informative to understand cell state landscapes and underlying regulatory networks for each cell type. The CRISPR–Cas field continues to develop creative tools for precise single-locus editing and other manipulations 79 , and incorporation of these toolkits with single-cell sequencing readouts will certainly bring new mechanistic insight.

Life forms are inherently dynamic, and each cell has a story to tell. Static measurements do not provide sufficient insight into the mechanisms that give rise to each cell state observed in a tissue. Computational approaches to stitch together independent measurements across time can be used to reconstruct potential histories; however, these are indirect inferences. Long-term live imaging in 2D cultures using confocal microscopy and in 3D tissues using light-sheet microscopy provides morphology, behaviour, location and, in some cases, molecular information on the history of a cell. Indeed, such long-term imaging experiments revealed that cell fates or states can be predicted from cell behaviour across many generations 80 . Cell tracking combined with end point single-cell genomics experiments can help to understand how cell states came to be; however, these experiments lack molecular resolution of the intermediates. There are strategies using CRISPR–Cas systems to capture highly prevalent RNAs inside cells at given times and insert these RNAs into DNA for storage and subsequent readout 81 . Together with live tracking and end-point single-cell genomics, such methods could provide unprecedented insight into cell histories.

My vision is that the emerging technologies described above can be applied to human 2D cell culture and 3D organoid biosystems to understand human development and disease mechanisms. My team and others are working to build virtual human organs that are based on high-throughput, multimodal single-cell genomics data. Organoid counterparts provide opportunities to perturb the system and understand lineage histories. Together, the next generation of single-cell genomics methods and human organoid technologies will provide unprecedented opportunities to develop new therapies for human disease.

the next generation of single-cell genomics methods and human organoid technologies will provide unprecedented opportunities

Unravelling the layers of the epigenome

Alexander Meissner. Around 1975, the idea that 5-methylcytosine could provide a mechanism to control gene expression gained traction, despite little knowledge of its genomic distribution or the associated enzymes 82 . With similarly limited genomic information or knowledge of the players involved, the histone code hypothesis was put forward in 2000 to explain how multiple different covalent modifications of chromatin may be coordinated to direct specific regulatory functions 83 . Tremendous progress has been made since, and the list of core epigenetic regulators that have been discovered and characterized seems largely complete 84 .

DNA sequencing has continued to dominate the past decade and contributed to an exponential growth of genome-wide maps of all layers of regulation. In the early days, individual CpG sites could be measured by restriction enzymes, whereas now we have generated probably well over a trillion cytosine methylation measurements. An equally astonishing number of genome-wide data sets have been collected for transcriptomes, histone modifications, transcription factor occupancy and DNA accessibility. Furthermore, the number of single-cell transcriptome and epigenome data sets continues to grow at an unprecedented pace.

On the basis of this overabundance of data across many normal and diseased cell states, for instance, we now clearly understand the non-random distribution of cytosine methylation across many different organisms. These maps have helped to refine our understanding of its relationship to gene expression, including the realization that only a few promoters are normally controlled via this modification, whereas gene bodies are actively targeted, and most dynamic changes occur at distal regulatory sites. Similar insights exist for many core histone modifications, and, in general, we have an improved appreciation of the epigenetic writers, readers and erasers involved. Over the past decade, we have seen substantially integrated and multilayered epigenomic analyses that provide a fairly comprehensive picture of epigenomic landscapes, including their dynamics across development and disease.

Additional innovation is now needed around data access and sharing. As noted, there is certainly no shortage of data, but to enable individual researchers to generate and verify hypotheses quickly improved tools are required to access and browse these data. Over the past decade, large coordinated projects such as ENCODE , the Roadmap Epigenomics Project and Blueprint Epigenome have initiated such efforts, but it remains a reality that data are not at everyone’s fingertips quite yet.

Moreover, despite decades of steady and recently accelerated progress, many important questions remain regarding the molecular coordination and developmental functions of these epigenetic modifications. For instance, cytosine methylation at gene bodies has been preserved for more than a billion years of evolution and yet its precise function is still under investigation. How and why did genomic methylation switch to a global mechanism in vertebrates compared with the selected methylation observed in invertebrates? What is the precise function of this modification in each of its regulatory contexts, and how are its ubiquitously acting enzymes recruited to specific sites in the genome? The latter is particularly timely given recent observations that enhancers, but also some repetitive elements, show ongoing recruitment of both de novo methylation and demethylation activity. Moreover, extraembryonic tissues show redirected activity that shares notable similarities with the long observed altered DNA methylation landscape found across most cancer types 85 . Lastly, it is abundantly clear that DNA methylation is essential for mammalian development; but despite us knowing this for nearly three decades, it is not clear how and why developing knockout embryos die. The specific developmental requirements are also largely true for many histone-modifying enzymes; however, it remains incompletely understood how exactly these modifications interact to support gene regulation.

A decade ago it seemed likely that we would answer questions such as these using newly gained sequencing power as a potent tool for generating hypotheses. However, for the most part, epigenomic analyses have expanded a highly valuable, but still largely descriptive, understanding of numerous epigenetic layers. So one may ask, what is different now and why should we expect to answer these questions in the coming years?

Technological innovation has always played a key role in biology, and some broadly applicable, recent breakthroughs will enable us to drive progress in the coming years. These include the transfer of the bacterial innate immunity CRISPR–Cas system as a universal genome-targeting tool 86 as well as for base editing, epigenome editing and various genome manipulations. Similarly, new fast-acting endogenous protein degradation systems have been developed that further enhance our ability to probe for precise function 87 . The past decade also saw major improvements in imaging technologies as well as cell and molecular biology, moving from the 2D space into the 3D space with both organoid cell culture models 88 and chromosome conformation capture approaches for exploring nuclear organization 89 .

Another major shift included the reappreciation that membraneless organelles are a widespread mechanism of cellular organization 90 . In particular, there have been many advances in our understanding of how condensates form and function, including for transcriptional regulation. Together with known properties of modified histones on DNA and the fact that many epigenetic regulators also contain intrinsically disordered regions, it is reasonable to assume that these physical properties will have a major impact on our understanding of chromatin. Importantly, changes in topology have been linked to disease 91 , and similar connections have been reported recently for condensates 92 . This will likely be an exciting area to follow in the coming years.

there have been many advances in our understanding of how condensates form and function, including for transcriptional regulation

Lastly, our research continues to be more and more reliant on multidisciplinary skills, with mathematics, physics, chemistry and computer science playing an ever-more central role in biology, which will require some rethinking in training and institutional organization to accomplish our goals. Going forward, we will need more functional integration, which in part due to the aforementioned selected discoveries is now very tractable. In particular, more refined perturbation of gene activity, which for many chromatin regulators should be separated into catalytic and regulatory functions, together with readouts at multiple levels of resolution will bring us closer to the insights needed. We recently exemplified this with a pipeline that explores epigenetic regulator mutant phenotypes at single-cell resolution 93 . From these studies, we may be able to understand how epigenetic regulators interact with the environment to influence or protect the organismal phenotype, connecting detailed molecular genetics to classical theories of epigenetic phenomena.

As we approach the 100-year anniversary of the detection of 5-methylcytosine in DNA 94 , it seems we can hope to declare at least for some layers of the epigenome that we fully understand the rules under which they operate. This may enable the exploration of more precise therapeutic interventions, for instance by redirecting chromatin modifiers rather than blocking their universal catalytic activities, which are shared between normal and diseased states. Of course, looking back at predictions made just 10 years ago 95 , one should expect many additional unforeseen advances that are just as difficult to predict now as they were back then.

Long non-coding RNAs: a time to build

Howard Chang. Long non-coding RNAs (lncRNAs) are the dominant transcriptional output of many eukaryotic genomes. Although studies over the past decade have revealed diverse mechanisms and disease implications for many lncRNAs, the vast majority of lncRNAs remain mysterious. The fundamental challenge is that we lack the knowledge to systematically transform lncRNA sequence into function. Progress in the next decade may come from a paradigm shift from ‘reading’ to ‘writing’ lncRNAs.

Gene regulation was once thought to be the exclusive province of proteins. Intense efforts for disease diagnosis and treatment focused almost entirely on protein-coding genes and their products, ignoring the vast majority of the genome. Even at the time of the completion of the Human Genome Project, only a handful of functional lncRNAs were known that silenced the expression of neighbouring genes. Thus, it was widely believed that the genome contained mostly ‘junk’, which sometimes made RNA as transcriptional noise.

The human genome is currently estimated to encode nearly 60,000 lncRNAs, ranging from several hundred to tens of thousands of bases, that apparently do not function by encoding proteins 96 . Studies over the past decade discovered that many lncRNAs act at the interface between chromatin modification machinery and the genome. Specific lncRNAs can act as guides, scaffolds or decoys to control the recruitment of specific chromatin modification enzymes or transcription factors to DNA or their dismissal from DNA 97 . lncRNAs can activate as well as silence genes, and these RNAs can target neighbouring genes as a function of local chromosomal folding (in cis ) or at a distance throughout the genome (in trans ). Detailed dissections of individual lncRNAs have revealed that lncRNAs are composed of modular RNA motifs that enable one lncRNA to connect proteins that read, write or erase specific chromatin marks. These findings have galvanized substantial excitement about lncRNAs; laboratories around the world are now investigating the roles of lncRNAs in diverse systems, ranging from control of flowering time in plants to mutations in human genetic disorders.

Nonetheless, the notable progress to date can be viewed as anecdotal — each lncRNA is its own story. When a new lncRNA sequence is recognized in a genome database or RNA profiling experiment, we are still in the dark about what may happen to the cell or organism (if anything) when the lncRNA is removed. Indeed, efforts to ‘read’ lncRNAs have been the dominant experimental strategy over the past two decades. Systematic efforts in the ENCODE, FANTOM and emerging cell atlas consortia have mapped the transcriptional landscape, transcript isoforms and, more recently, single-cell expression profiles of lncRNAs. These powerful data are now combined with genome-scale CRISPR-based methods to inactivate tens of thousands of lncRNAs, one at a time, to observe possible cell defects 98 , 99 . However, many challenges remain. Positive hits require further exploratory studies to define possible mechanisms of action, and we lack a principled strategy to combine lncRNA knockouts to address genetic redundancy and compensation.

A potentially fruitful and complementary direction is the pivot from ‘reading’ to ‘writing’ long RNA scripts. On the basis of the systematic dissection of RNA sequences and secondary structures in lncRNAs, we and others believe that the information in lncRNAs resembles that on a billboard (in which keywords and catchphrases are repeated) rather than a finely honed legal document (where every comma counts). Small units of RNA shapes are repeated within lncRNAs to build up the meaning in the lncRNA billboard, but these RNA shapes can be rearranged in different orders or locations without affecting meaning. These insights have allowed scientists to recognize lncRNA genes from different species that perform the same function even though the primary sequences bear little similarity 100 . Moreover, investigators were able to strip down lncRNAs to their essential ‘words’, composed of these key repeating shapes and one-tenth the size of the original lncRNA, which still functioned in vivo to control chromatin state over a whole chromosome 100 , 101 . Finally, it is now possible to successfully create synthetic lncRNAs. By adding RNA shapes to carefully chosen RNA templates, investigators are starting to create designer lncRNAs that can regulate chromatin in vivo 100 , suffice to partly rescue the physiological lncRNA gene knockout 102 , or target RNAs to specific cytotopic locations within the cell 103 , 104 .

The shift from reading to writing lncRNAs will challenge us on the technical front, leading to potential transformative technologies. Current technologies for massively parallel reporter gene assays are built on short sequence inserts. A plan to build tens of thousands of synthetic lncRNAs will require accurate long DNA or RNA synthesis. These designer sequences will need to be placed into the appropriate locations in the genome and controlled to have proper developmental expression, splicing pattern and RNA chemical modifications. Landmark studies using the XIST lncRNA, which normally silences the second X chromosome in female cells, to silence the ectopic chromosome 21 in Down syndrome cells highlight the biomedical promise of such an approach 105 .

As the field develops technologies for large-scale creation and testing of synthetic lncRNAs, we can rigorously test our understanding of the information content in the language of RNA sequences and shapes. The next decade promises to be an exciting time for building non-coding RNAs and to create entirely new tools to manipulate gene function for biology and medicine.

FAIR genomics to track tumorigenesis

Núria López-Bigas. Cancer research is one of the fields that has probably benefited the most from the technological and methodological advances of genomics. In the span of less than two decades, the field has witnessed an incredible boost in the generation of cancer genomic, epigenomic and transcriptomic data of patients’ tumours, both in bulk and more recently at the single-cell level. My dream as a cancer researcher is to have a full understanding of the path that cells follow towards tumorigenesis. Which events in the life of an individual, a tissue and a particular cell lead to the malignant transformation of some cells? Of course I do not expect to have a deterministic answer, as this is not a deterministic process. Instead we should aim for a quantitative or probabilistic understanding of the key events that drive tumorigenesis. We have solid epidemiological evidence showing that smoking increases the probability of lung cancer, exposure to the Sun raises the probability of developing melanoma and some anticancer treatments increase the probability of secondary neoplasms. But which specific mechanisms at the molecular and cellular levels influence these increases?

One first clear goal of cancer genomics is to catalogue all genes involved in tumorigenesis across different tissues. Although this is a daunting task, it is actually feasible 106 . By analysing the mutational patterns of genes across tumours, one can identify those with significant deviations from what is expected under neutrality, which indicates that these mutations provide a selective advantage in tumorigenesis and are thus driver mutations. We can imagine a future in which through the systematic analysis of millions of sequenced tumour genomes this catalogue or compendium moves closer and closer to completion. For this to happen, not only do we need genome sequencing to expand — this process is already in motion in research, clinical settings and the pharmaceutical industry — but more importantly the resulting data must be made FAIR (findable, accessible, interoperable and reusable) 107 . To this end, consortia and initiatives that promote, catalyse and facilitate the sharing of genomic data, such as the Beyond 1 Million Genomes consortium, the GA4GH or the cBioPortal for Cancer Genomics , are necessary.

Of note, cataloguing genes and mutations involved in cancer development, albeit a very important first step, is still far from the final goal of understanding how and under which conditions they drive tumorigenesis. Framing cancer development as a Darwinian evolutionary process helps me to navigate the path towards this final objective. As is true of any Darwinian process, its two key features are variation and selection. Thanks to the past 15 years of cancer genomics, we now have a much better grasp of the origin of somatic genetic variation between cells across different tissues. The study of the variability in the number, type and genomic distribution of mutations across tumours provides a window into the life history of cells across the somatic tissues of an individual 108 , 109 . In addition, recent studies sequencing the genome of healthy cells in different tissues 110 , 111 , 112 have shown that mutations accumulate in hundreds and thousands in our cells in normal conditions over time. These studies have also detected positive selection in some genes across healthy tissues. Hence, positive selection is a pervasive process that operates not only in tumorigenesis but also in healthy tissues, where it is a hallmark of somatic development of skin, oesophagus, blood and other tissues. Take, for example, clonal haematopoiesis: it results from a continuous Darwinian evolutionary process in which over time (with age) some haematopoietic cells harbouring mutations in certain blood development genes, such as DNMT3A and TET2 , outcompete other cells in the compartment 113 , 114 . This process is part of normal haematopoietic development. Problems arise only when this process gets out of control, leading to leukaemia in the case of blood, or a malignant tumour in solid tissues. Why is it only in rare cases that this ubiquitous interplay between variation and selection becomes uncontrollable and results in full-blown tumorigenesis? Which events, beside known tumorigenic mutations, drive this process?

we now have a much better grasp of the origin of somatic genetic variation between cells across different tissues

If we have learnt something in recent years, it is that virtually all tumours harbour driver mutations 115 , 116 , 117 , implying that driver genomic events are necessary. However, they are clearly not sufficient for tumorigenesis to occur. So, what are these other triggers of the tumorigenic process? What happens in the lung cells of a smoker or in the haematopoietic cells of a patient treated with chemotherapy that increases their chances to become malignant? Epigenetic modifications and changes in selective constraints, such as evolutionary bottlenecks, for example, at the time of chemotherapy, may be part of the answer.

For the near future, my dream is to see a further increase in FAIR cancer genomics data to help us disentangle the step-by-step game of variation and selection in our tissues that leads to tumorigenesis and likely other ageing-related diseases.

Integrating genomics into medicine

Eran Segal. The past 20 years in genomics have been extraordinary. We developed high-throughput sequencing and learned how to use it to efficiently sequence full genomes and measure gene expression and epigenetic marks at the genome-wide scale and even at the single-cell level 118 . Using these capabilities, we created unprecedented catalogues of novel genomes, functional DNA elements and non-coding RNAs from all kingdoms of life 119 . But — perhaps with the exception of cancer 120 and gene therapy for some monogenic diseases 121 — genomics has yet to deliver on its promise to have an impact on our everyday life. For example, drugs and diagnostics are still being developed in the traditional way, with screening assays to find lead compounds for targets typically arising from animal studies, without involving genomics in any of the steps. Moreover, when the global COVID-19 pandemic hit, the genome of the spreading severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was rapidly sequenced, but why some infected individuals exhibit severe disease and others do not remains unknown.

Indeed, our next challenge is to translate the incredible resources and technologies developed in genomics into an improved understanding of health and disease. This improved understanding should transform the field of medicine to use genomics in its transition to personalized medicine, which promises individualized treatment by targeting the right medication to the right person at the right time on the basis of that person’s unique profile. By continuing to focus on more and more measurements and the creation of more atlases and catalogues, we run the danger of drowning in ever-growing amounts of data and correlative findings. Walking down this path can lead to an endless endeavour, as bulk measurements can always be replaced with single-cell ones, or measures at higher temporal and spatial resolution, across more conditions and wider biological contexts.

Instead, we should use genomics to tackle big unanswered questions such as what causes the variation that we see across people in phenotypes, disease susceptibility and drug responses? What is the relative contribution of genetic, epigenetic, microbiome and environmental factors? How are their effects mediated, and what would be the effect of different interventions? Ultimately, we should strive to use genomics to generate actionable and personalized insights that lead to better health. We are now at an inflexion point in genomics that allows us for the first time to apply it to study human biology and realize these ambitious aims 122 .

At the cellular level, we can use iPS cells from patients to derive cellular models of multiple diseases and prioritize treatments based on measuring both their cellular and molecular response (for example, gene expression and epigenetics) to existing drugs and drug combinations. We can even use massively parallel assays to separately measure the effect of each of tens of thousands of rationally designed mutations, including patient-specific mutations, as we have done, for example, in testing the effect of all clinically identified mutations in TP53 on cellular function 123 . Measuring the molecular effects of directed mutations in genes encoding transcription factors and signalling molecules and in other genes can reveal the underlying pathways and regulatory networks of the disease studied and identify putative therapeutic targets. The application of such approaches to fields that are still poorly understood, such as neurodegenerative diseases, can be particularly impactful.

But we can be much more ambitious and directly profile large cohorts of human individuals using diverse ‘omics’ assays. As molecular changes typically precede clinical disease manifestations, longitudinal measurements coupled with clinical phenotyping have the potential of identifying novel disease diagnostics and therapeutic targets. Indeed, biobanks that track large samples of hundreds of thousands of individuals have recently emerged and are proving highly informative 124 . However, at the molecular level their focus has thus far been on genetics. Technological advances and cost reductions now allow us to obtain much deeper person-specific multi-omic profiles that include transcriptome, proteome, methylome, microbiome, immune system and metabolome measurements. Having these data on the same individual and at multiple time points can reveal which omic layer is more perturbed and informative for each disease and identify associations between molecular markers and disease.

The challenge in using such observational data from human cohorts is to identify which of the associations are causal. One way to address this is to wisely select the nature and type of the associations studied. For example, in working with microbiome data, we can move from analyses at the level of species composition to analyses at the level of SNPs in bacterial genes. Such associations are more specific and more likely to be causal, as in the case of a SNP in the dadH bacterial gene, which correlated with metabolism of the primary medication to treat Parkinson disease and the gut microbiota from patients 125 . Another approach is to use longitudinal measurements and separation of time to emulate target trials from observational data 126 . For example, we can select distinct subsets from the cohort that match on several known risk factors (for example, age or body mass index) but differ on a marker of interest (for example, expression of a gene or presence of an epigenetic mark), and compare future disease onset or progression in these two populations. Similarly, retrospective analysis of baseline multi-omic measurements from participants in randomized clinical trials may identify markers that distinguish responders from non-responders and be used for patient stratification or for identifying additional putative targets.

Ultimately, biomarkers identified from observational cohorts need to be tested in randomized clinical trials to establish causality and assess efficacy. In the case of microbial strains extracted from humans, we may be able to skip animal testing and go directly to human trials. In other cases, such as when human genes are being manipulated, we will need to start with cell culture assays and animal testing before performing clinical trials in humans. However, in all cases, tested omic targets should have already shown associations in human individuals, thus making them more likely to be relevant and succeed in trials, as is the case with drug targets for which genetic evidence links them to the disease 127 .

Beyond these scientific challenges, there is the challenge of engaging the public and diverse ethnic and socio-economic groups to participate in such large-scale multi-omic profiling endeavours even before we can present them with immediate benefits. We can start with incentives in the form of informational summary reports of the data measured and gradually move towards carefully and responsibly conveyed actionable insights as we learn more.

Overcoming the aforementioned challenges is not an easy task, but with the breathtaking advances that genomics has undergone in the past two decades, the time may be right to tackle them. Success can transform genomics from being applied mostly in research settings to having it become an integral and inseparable part of medicine.

CRISPR genome editing enters the clinic

Jin-Soo Kim. In the past several years, genome editing has come of age 128 , in particular because of the repurposing of CRISPR systems. Genomic DNA can be modified in a targeted manner in vivo or in vitro with high efficiency and precision, potentially enabling therapeutic genome editing for the treatment of both genetic and non-genetic diseases. All three types of programmable nucleases developed for genome editing, namely zinc-finger nucleases, transcription activator-like effector nucleases and CRISPR nucleases, are now under clinical investigation. In the next several years, we will be able to learn whether these genome-editing tools will be effective and safe enough to treat patients with an array of diseases, including HIV infection, leukaemia, blood disorders and hereditary blindness, heralding a new era in medicine.

If the history of the development of novel drugs or treatments such as gene therapy and monoclonal antibodies is any guide, the road to therapeutic genome editing is likely to be bumpy but ultimately worth travelling. Key questions related to medical applications of programmable nucleases concern their mode of delivery, specificity, on-target activity and immunogenicity. First, in vivo delivery (or direct delivery into patients) of genes or mRNAs encoding programmable nucleases or preassembled Cas9 ribonucleoproteins can be a challenge, given the large size of these nucleases. Ex vivo (or indirect) delivery is, in general, more efficient than in vivo delivery but is limited to cells from blood or bone marrow, which can be collected with ease, edited in vitro and transfused back into patients. Ongoing developments of nanoparticles and viral vectors are expected to enhance and expand in vivo genome editing in tissues or organs not readily accessible with current delivery systems, such as the brain.

Second, programmable nucleases, including CRISPR nucleases, can cause unwanted on-target and off-target mutations, which may contribute to oncogenesis. Several cell-based and cell-free methods have been developed to identify genome-wide CRISPR off-target sites in an unbiased manner 129 , 130 , 131 . But it remains a challenge to validate off-target activity at sites with low mutation frequencies (less than 0.1%) in a population of cells, owing to the intrinsic error rates of current sequencing technologies. Even at on-target sites, CRISPR–Cas9 can induce unexpected outcomes such as large deletions of chromosomal segments 132 . It will be important to understand the mechanisms behind the unusual on-target activity and to measure and reduce the frequencies of such events.

Last but not least, Cas9 and other programmable nucleases can be immunogenic, potentially causing undesired innate and adaptive immune responses. In this regard, it makes sense that initial clinical trials have focused on ex vivo delivery of Cas9 ribonucleoproteins into T cells or in vivo gene editing in the eye, an immunologically privileged organ. Cas9 epitope engineering or novel Cas9 orthologues derived from non-pathogenic bacteria may avoid some of the immune responses, offering therapeutic modalities for in vivo genome editing in tissues or organs with little or no immune privilege.

Base editing 133 , 134 and prime editing 135 are promising new approaches that may overcome some of the limitations of nuclease-mediated genome editing. Base editors and prime editors are composed of a Cas9 nickase, rather than the wild-type Cas9 nuclease, and a nucleobase deaminase and a reverse transcriptase, respectively. Because a nickase, unlike a nuclease, produces DNA single-strand breaks or nicks, but not double-strand breaks (DSBs), base editors and prime editors are unlikely to induce large deletions at on-target sites and chromosomal rearrangements resulting from non-homologous end joining (NHEJ) repair of concurrent on-target and off-target DSBs. Furthermore, when it comes to gene correction rather than gene disruption, these new types of gene editors are much more efficient and ‘cleaner’ than DSB-producing nucleases because they neither require donor template DNA nor rely on error-prone NHEJ; in human cells, DSBs are preferentially repaired by NHEJ, leading to small insertions or deletions (indels), rather than by homologous recombination involving donor DNA.

Base editors and prime editors are also well suited for germline editing and in utero editing (that is, gene editing in the fetus), which should be done with caution, in full consideration of ethical, legal and societal issues. In principle, CRISPR–Cas9 can be used for the correction of pathogenic mutations in human embryos; however, donor DNA is seldom used as a repair template in human embryos 136 . Recurrent or non-recurrent de novo mutations are responsible for the vast majority of genetic diseases. Cell-free fetal DNA in the maternal blood can be used to detect these de novo mutations in fetuses, which are absent in the parents. Some de novo mutations are manifested even before birth, leading to miscarriage, disability or early death after birth; it is often too late and inefficient to attempt gene editing in newborns. These mutations could be corrected in utero using base editors or prime editors without inducing unwanted indels and without relying on inefficient homologous recombination. Compared with germline editing or preimplantation genetic diagnosis, in utero editing, if proven safe and effective in the future, should be ethically more acceptable because it does not involve the creation or destruction of human embryos.

As promising and powerful as they are, current versions of base editors and prime editors can be further optimized and improved. For instance, Cas9 evolved in microorganisms as a nuclease rather than a nickase. Current Cas9 nickases used for base editing (D10A SpCas9 variant) and prime editing (H840A variant) can be engineered to increase their activities and specificities. In parallel, deaminase and reverse transcriptase moieties in base editors and prime editors, respectively, can be engineered or replaced with appropriate orthologues to increase the efficiency and scope of genome editing. It has been shown that base editors can cause both guide RNA-dependent and guide RNA-independent DNA or RNA off-target mutations, raising concerns for their applications in medicine. Prime editors may also cause unwanted on-target and off-target mutations, which must be carefully studied before moving on to therapeutic applications.

Biomedical researchers are now equipped with powerful tools for genome editing. I expect that these tools will be developed further and applied more broadly in both research and medicine in the coming years.

Collins F. The director of the NIH lays out his vision of the future of medical science. Time https://time.com/5709207/medical-science-age-of-discovery (2019).

The National Academies of Sciences, Engineering, and Medicine Organizing Committee for the International Summit on Human Gene Editing. On human gene editing: international summit statement. The National Academies of Sciences, Engineering, and Medicine https://www.nationalacademies.org/news/2015/12/on-human-gene-editing-international-summit-statement (2015).

Centers for Disease Control and Prevention. COVID-19 in racial and ethnic minority groups. CDC https://www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/racial-ethnic-minorities.html (2020).

Edwards, F., Lee, H. & Esposito, M. Risk of being killed by police use of force in the United States by age, race–ethnicity, and sex. Proc. Natl Acad. Sci. USA 116 , 16793–16798 (2019).

CAS   PubMed   PubMed Central   Google Scholar  

Popejoy, A. B. & Fullerton, S. M. Genomics is failing on diversity. Nature 538 , 161–164 (2016).

Popejoy, A. B. et al. The clinical imperative for inclusivity: race, ethnicity, and ancestry (REA) in genomics. Hum. Mutat. 39 , 1713–1720 (2018).

PubMed   PubMed Central   Google Scholar  

Artiga, S. & Orgera, K. Key facts on health and health care by race and ethnicity. Kaiser Family Foundation https://www.kff.org/report-section/key-facts-on-health-and-health-care-by-race-and-ethnicity-coverage-access-to-and-use-of-care/ (2019).

Armstrong, K., Micco, E., Carney, A., Stopfer, J. & Putt, M. Racial differences in the use of BRCA1/2 testing among women with a family history of breast or ovarian cancer. JAMA 293 , 1729–1736 (2005).

CAS   PubMed   Google Scholar  

Bonham, V. L., Callier, S. L. & Royal, C. D. Will precision medicine move us beyond race? N. Engl. J. Med. 374 , 2003–2005 (2016).

Karczewski, K. J. et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581 , 434–443 (2020).

Khera, A. V. et al. Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nat. Genet. 50 , 1219–1224 (2018).

The SIGMA Type 2 Diabetes Consortium. Sequence variants in SLC16A11 are a common risk factor for type 2 diabetes in Mexico. Nature 506 , 97–101 (2014).

Google Scholar  

Wetterstrand, K. A. DNA sequencing costs: data from the NHGRI genome sequencing program (GSP). National Human Genome Research Institute https://www.genome.gov/sequencingcostsdata (2019).

Wall, J. D. et al. The GenomeAsia 100K Project enables genetic discoveries across Asia. Nature 576 , 106–111 (2019).

CAS   Google Scholar  

Kowalski, M. H. et al. Use of >100,000 NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium whole genome sequences improves imputation quality and detection of rare variant associations in admixed African and Hispanic/Latino populations. PLoS Genet. 15 , e1008500 (2019).

Bycroft, C. et al. The UK Biobank resource with deep phenotyping and genomic data. Nature 562 , 203–209 (2018).

Gaziano, J. M. et al. Million veteran program: a mega-biobank to study genetic influences on health and disease. J. Clin. Epidemiol. 70 , 214–223 (2016).

PubMed   Google Scholar  

Nagai, A. et al. Overview of the BioBank Japan Project: study design and profile. J. Epidemiol. 27 , S2–S8 (2017).

Denny, J. C. et al. Systematic comparison of phenome-wide association study of electronic medical record data and genome-wide association study data. Nat. Biotechnol. 31 , 1102–1110 (2013).

Sirugo, G., Williams, S. M. & Tishkoff, S. A. The missing diversity in human genetic studies. Cell 177 , 1080 (2019).

Martin, A. R. et al. Clinical use of current polygenic risk scores may exacerbate health disparities. Nat. Genet. 51 , 584–591 (2019).

McQuillan, M. A., Zhang, C., Tishkoff, S. A. & Platt, A. The importance of including ethnically diverse populations in studies of quantitative trait evolution. Curr. Opin. Genet. Dev. 62 , 30–35 (2020).

Sohail, M. et al. Polygenic adaptation on height is overestimated due to uncorrected stratification in genome-wide association studies. eLife 8 , e39702 (2019).

Crawford, N. G. et al. Loci associated with skin pigmentation identified in African populations. Science 358 , eaan8433 (2017).

Gasperini, M., Tome, J. M. & Shendure, J. Towards a comprehensive catalogue of validated and target-linked human enhancers. Nat. Rev. Genet. 21 , 292–310 (2020).

Racimo, F., Sankararaman, S., Nielsen, R. & Huerta-Sánchez, E. Evidence for archaic adaptive introgression in humans. Nat. Rev. Genet. 16 , 359–371 (2015).

Skoglund, P. & Mathieson, I. Ancient genomics of modern humans: the first decade. Annu. Rev. Genomics Hum. Genet. 19 , 381–404 (2018).

Vicente, M. & Schlebusch, C. M. African population history: an ancient DNA perspective. Curr. Opin. Genet. Dev. 62 , 8–15 (2020).

Sherman, R. M. et al. Assembly of a pan-genome from deep sequencing of 910 humans of African descent. Nat. Genet. 51 , 30–35 (2019).

Durvasula, A. et al. Recovering signals of ghost archaic introgression in African populations. Sci. Adv. 12 , eaax5097 (2020).

Skov, L. et al. The nature of Neanderthal introgression revealed by 27,566 Icelandic genomes. Nature 582 , 78–83 (2020).

Adeyemo, A. A. et al. ZRANB3 is an African-specific type 2 diabetes locus associated with beta-cell mass and insulin response. Nat. Commun. 10 , 3195 (2019).

Cohen, J. et al. Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9 . Nat. Genet. 37 , 161–165 (2005).

Gurdasani, D. et al. Uganda genome resource enables insights into population history and genomic discovery in Africa. Cell 179 , 984–002.e36 (2019).

Gurdasani, D. et al. Genomics of disease risk in globally diverse populations. Nat. Rev. Genet. 20 , 520–535 (2019).

Gulsuner, S. et al. Genetics of schizophrenia in the South African Xhosa. Science 367 , 569–573 (2020).

Shriner, D. & Rotimi, C. N. Whole-genome-sequence-based haplotypes reveal single origin of the sickle allele during the Holocene wet phase. Am. J. Hum. Genet. 102 , 547–556 (2018).

Wu, Y. et al. Highly efficient therapeutic gene editing of human haematopoietic stem cells. Nat. Med. 25 , 776–783 (2019).

Geard, A. et al. Clinical and genetic predictors of renal dysfunctions in sickle cell anaemia in Cameroon. Br. J. Haematol. 178 , 629–639 (2017).

Lebeko, K. et al. Targeted genomic enrichment and massively parallel sequencing identifies novel nonsyndromic hearing impairment pathogenic variants in Cameroonian families. Clin. Genet. 90 , 288–290 (2016).

Genovese, G. et al. Association of trypanolytic ApoL1 variants with kidney disease in African Americans. Science 329 , 841–845 (2010).

Sierra, B. et al. OSBPL10, RXRA and lipid metabolism confer African-ancestry protection against dengue haemorrhagic fever in admixed Cubans. PLoS Pathog. 13 , e1006220 (2017).

Wonkam, A. & de Vries, J. Returning incidental findings in African genomics research. Nat. Genet. 52 , 17–20 (2020).

Provine, W. B. The Origins of Theoretical Population Genetics (University of Chicago Press, 1971)

Fisher, R. A. The correlation between relatives on the supposition of Mendelian inheritance. Trans. R. Soc. Edinb. 52 , 399–433 (1918).

Altenburg, E. & Muller, H. J. The genetic basis of truncate wing – an inconstant and modifiable character in Drosophila. Genetics 5 , 1–59 (1920).

Morton, N. E. Analysis of family resemblance. I. Introduction. Am. J. Hum. Genet. 26 , 318–330 (1974).

Visscher, P. M. et al. 10 Years of GWAS discovery: biology, function and translation. Am. J. Hum. Genet. 101 , 5–22 (2017).

Boyle, E. A., Li, Y. I. & Pritchard, J. K. An expanded view of complex traits: from polygenic to omnigenic. Cell 169 , 1177–1186 (2017).

Emison, E. S. et al. A common, sex-dependent mutation in a putative RET enhancer underlies Hirschsprung disease susceptibility. Nature 434 , 857–863 (2005).

Maurano, M. T. et al. Systematic localization of common disease-associated variation in regulatory DNA. Science 337 , 1190–1195 (2012).

Davidson, E. Emerging properties of animal gene regulatory networks. Nature 468 , 911–920 (2010).

Chatterjee, S. et al. Enhancer variants synergistically drive dysregulation of the RET gene regulatory network in Hirschsprung disease. Cell 167 , 355–368 (2016).

Segal, E., Raveh-Sadka, T., Schroeder, M., Unnerstall, U. & Gaul, U. Predicting expression patterns from regulatory sequence in Drosophila segmentation. Nature 451 , 535–540 (2008).

Chakravarti, A. & Turner, T. N. Revealing rate-limiting steps in complex disease biology: The crucial importance of studying rare, extreme-phenotype families. Bioessays 38 , 578–586 (2016).

Lancaster, M. A. et al. Cerebral organoids model human brain development and microcephaly. Nature 501 , 373–379 (2013).

Rothman, J. & Jarriault, S. Developmental plasticity and cellular reprogramming in caenorhabditis elegans. Genetics 213 , 723–757 (2019).

Porrello, E. R. et al. Transient regenerative potential of the neonatal mouse heart. Science 331 , 1078–1080 (2011).

Mir, M., Bickmore, W., Furlong, E. E. M. & Narlikar, G. Chromatin topology, condensates and gene regulation: shifting paradigms or just a phase? Development 146 , dev182766 (2019).

Ghavi-Helm, Y. et al. Highly rearranged chromosomes reveal uncoupling between genome topology and gene expression. Nat. Genet. 51 , 1272–1282 (2019).

Despang, A. et al. Functional dissection of the Sox9-Kcnj2 locus identifies nonessential and instructive roles of TAD architecture. Nat. Genet. 51 , 1263–1271 (2019).

Hnisz, D., Shrinivas, K., Young, R. A., Chakraborty, A. K. & Sharp, P. A. A phase separation model for transcriptional control. Cell 169 , 13–23 (2017).

Shrinivas, K. et al. Enhancer features that drive formation of transcriptional condensates. Mol. Cell 75 , 549–561 e547 (2019).

Narlikar, G. J. Phase-separation in chromatin organization. J. Biosci. 45 , 5 (2020).

Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566 , 496–502 (2019).

Farrell, J. A. et al. Single-cell reconstruction of developmental trajectories during zebrafish embryogenesis. Science 360 , eaar3131 (2018).

Cusanovich, D. A. et al. The cis-regulatory dynamics of embryonic development at single-cell resolution. Nature 555 , 538–542 (2018).

Tang, F. et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat. Methods 6 , 377–382 (2009).

Camp, J. G., Platt, R. & Treutlein, B. Mapping human cell phenotypes to genotypes with single-cell genomics. Science 365 , 1401–1405 (2019).

Lein, E., Borm, L. E. & Linnarsson, S. The promise of spatial transcriptomics for neuroscience in the era of molecular cell typing. Science 358 , 64–69 (2017).

Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33 , 495–502 (2015).

Achim, K. et al. High-throughput spatial mapping of single-cell RNA-seq data to tissue of origin. Nat. Biotechnol. 33 , 503–509 (2015).

Cao, J. et al. Comprehensive single-cell transcriptional profiling of a multicellular organism. Science 357 , 661–667 (2017).

Cleary, B., Cong, L., Cheung, A., Lander, E. S. & Regev, A. Efficient generation of transcriptomic profiles by random composite measurements. Cell 171 , 1424–1436 e1418 (2017).

Cao, J. et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science 361 , 1380–1385 (2018).

Kester, L. & van Oudenaarden, A. Single-cell transcriptomics meets lineage tracing. Cell Stem Cell 23 , 166–179 (2018).

Stuart, T. & Satija, R. Integrative single-cell analysis. Nat. Rev. Genet. 20 , 257–272 (2019).

Zhu, C., Preissl, S. & Ren, B. Single-cell multimodal omics: the power of many. Nat. Methods 17 , 11–14 (2020).

Anzalone, A. V., Koblan, L. W. & Liu, D. R. Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol . (2020).

Loeffler, D. et al. Asymmetric lysosome inheritance predicts activation of haematopoietic stem cells. Nature 573 , 426–429 (2019).

Schmidt, F., Cherepkova, M. Y. & Platt, R. J. Transcriptional recording by CRISPR spacer acquisition from RNA. Nature 562 , 380–385 (2018).

Holliday, R. & Pugh, J. E. DNA modification mechanisms and gene activity during development. Science 187 , 226–232 (1975).

Strahl, B. D. & Allis, C. D. The language of covalent histone modifications. Nature 403 , 41–45 (2000).

Jambhekar, A., Dhall, A. & Shi, Y. Roles and regulation of histone methylation in animal development. Nat. Rev. Mol. Cell Biol. 20 , 625–641 (2019).

Smith, Z. D. et al. Epigenetic restriction of extraembryonic lineages mirrors the somatic transition to cancer. Nature 549 , 543–547 (2017).

Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337 , 816–821 (2012).

Nabet, B. et al. The dTAG system for immediate and target-specific protein degradation. Nat. Chem. Biol. 14 , 431–441 (2018).

Clevers, H. Modeling development and disease with organoids. Cell 165 , 1586–1597 (2016).

Dekker, J., Marti-Renom, M. A. & Mirny, L. A. Exploring the three-dimensional organization of genomes: interpreting chromatin interaction data. Nat. Rev. Genet. 14 , 390–403 (2013).

Banani, S. F., Lee, H. O., Hyman, A. A. & Rosen, M. K. Biomolecular condensates: organizers of cellular biochemistry. Nat. Rev. Mol. Cell Biol. 18 , 285–298 (2017).

Lupianez, D. G. et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 161 , 1012–1025 (2015).

Basu, S. et al. Unblending of transcriptional condensates in human repeat expansion disease. Cell 181 , 1062–1079 e1030 (2020).

Grosswendt, S. et al. Epigenetic regulator function through mouse gastrulation. Nature 584 , 102–108 (2020).

Johnson, T. B. & Coghill, R. D. Researches on pyrimidines. C111. The discovery of 5-methyl-cytosine in tuberculinic acid, the nucleic acid of the tubercle bacillus. J. Am. Chem. Soc. 47 , 2838–2844,47 (1925).

Heard, E. et al. Ten years of genetics and genomics: what have we achieved and where are we heading? Nat. Rev. Genet. 11 , 723–733 (2010).

Quinn, J. J. & Chang, H. Y. Unique features of long non-coding RNA biogenesis and function. Nat. Rev. Genet. 17 , 47–62 (2016).

Kopp, F. & Mendell, J. T. Functional classification and experimental dissection of long noncoding RNAs. Cell 172 , 393–407 (2018).

Liu, S. J. et al. CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells. Science 355 , eaah7111 (2017).

Rubin, A. J. et al. Coupled single-cell CRISPR screening and epigenomic profiling reveals causal gene regulatory networks. Cell 176 , 361–376.e17 (2019).

Quinn, J. J. et al. Rapid evolutionary turnover underlies conserved lncRNA-genome interactions. Genes. Dev. 30 , 191–207 (2016).

Kirk, J. M. et al. Functional classification of long non-coding RNAs by k-mer content. Nat. Genet. 50 , 1474–1482 (2018).

Carter, A. C. et al. Spen links RNA-mediated endogenous retrovirus silencing and X chromosome inactivation. eLife 9 , e54508 (2020).

Lubelsky, Y. & Ulitsky, I. Sequences enriched in Alu repeats drive nuclear localization of long RNAs in human cells. Nature 555 , 107–111 (2018).

Shukla, C. J. et al. High-throughput identification of RNA nuclear enrichment sequences. EMBO J. 37 , e98452 (2018).

Czerminski, J. T. & Lawrence, J. B. Silencing Trisomy 21 with XIST in neural stem cells promotes neuronal differentiation. Dev. Cell 52 , 294–308 e3 (2020).

Martínez-Jiménez, F. et al. A compendium of mutational cancer driver genes. Nat. Rev. Cancer https://doi.org/10.1038/s41568-020-0290-x (2020).

Article   PubMed   Google Scholar  

Wilkinson, M. et al. The FAIR guiding principles for scientific data management and stewardship. Sci. Data 3 , 160018 (2016).

Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578 , 94–101 (2020).

Gonzalez-Perez, A., Radhakrishnan, S. & Lopez-Bigas, N. Local determinants of the mutational landscape of the human genome. Cell 177 , 101–114 (2019).

Martincorena, I. et al. High burden and pervasive positive selection of somatic mutations in normal human skin. Science 348 , 880–886 (2015).

Martincorena, I. et al. Somatic mutant clones colonize the human esophagus with age. Science 362 , 911–917 (2018).

Yokoyama, A. et al. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature 565 , 312–317 (2019).

Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371 , 2477–2487 (2014).

Jaiswal, S. et al. Age related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371 , 2488–2498 (2014).

Sabarinathan, R. et al. The whole-genome panorama of cancer drivers. Preprint at bioRxiv https://doi.org/10.1101/190330 (2017).

Pich, O. et al. The mutational footprints of cancer therapies. Nat. Genet. 51 , 1732–1740 (2019).

Campbell, P. J. et al. Pan-cancer analysis of whole genomes. Nature 578 , 82–93 (2020).

Rozenblatt-Rosen, O., Stubbington, M. J. T., Regev, A. & Teichmann, S. A. The Human Cell Atlas: from vision to reality. Nature 550 , 451–453 (2017).

ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489 , 57–74 (2012).

Damodaran, S. et al. Cancer Driver Log (CanDL): catalog of potentially actionable cancer mutations. J. Mol. Diagn. 17 , 554–559 (2015).

High, K. A. & Roncarolo, M. G. Gene therapy. N. Engl. J. Med. 381 , 455–464 (2019).

Shilo, S., Rossman, H. & Segal, E. Axes of a revolution: challenges and promises of big data in healthcare. Nat. Med. 26 , 29–38 (2020).

Kotler, E. et al. A systematic p53 mutation library links differential functional impact to cancer mutation pattern and evolutionary conservation. Mol. Cell 71 , 873 (2018).

Swanson, J. M. The UK Biobank and selection bias. Lancet 380 , 110 (2012).

Maini Rekdal, V., Bess, E. N., Bisanz, J. E., Turnbaugh, P. J. & Balskus, E. P. Discovery and inhibition of an interspecies gut bacterial pathway for levodopa metabolism. Science 364 , eaau6323 (2019).

Hernán, M. A. & Robins, J. M. Using big data to emulate a target trial when a randomized trial is not available. Am. J. Epidemiol. 183 , 758–764 (2016).

Nelson, M. R. et al. The support of human genetic evidence for approved drug indications. Nat. Genet. 47 , 856–860 (2015).

Kim, J.-S. Genome editing comes of age. Nat. Protoc. 11 , 1573–1578 (2016).

Kim, D. et al. Digenome-seq: genome-wide profiling of CRISPR-Cas9 off-target effects in human cells. Nat. Methods 12 , 237–243 (2015).

Tsai, S. Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33 , 187–197 (2015).

Wienert, B. et al. Unbiased detection of CRISPR off-targets in vivo using DISCOVER-Seq. Science 364 , 286–289 (2019).

Kosicki, M., Tomberg, K. & Bradley, A. et al. Repair of double-strand breaks induced by CRISPR-Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 36 , 765–771 (2018).

Komor, A. C. et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533 , 420–424 (2016).

Nishida, K. et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 353 , aaf8729 (2016).

Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576 , 149–157 (2019).

Ma, H. et al. Correction of a pathogenic gene mutation in human embryos. Nature 548 , 413–419 (2017).

Download references

Acknowledgements

A.C. acknowledges that the ideas in his contribution were developed through studies on Hirschsprung disease and thanks the many trainees who have contributed to this work over the past 5 years. A.L.M. acknowledges A. Gutierrez, K. Kostick, G. Lazaro, M. Majumder, K. Munoz, S. Pereira, H. Smith and P. Zuk for feedback. A.M. thanks D. Hnisz, Z. D. Smith, J. Charlton and H. Kretzmer for feedback and the Max Planck Society for funding. A.W. is supported by NIH awards U54HG009790, U01HG009716, U01HG007459 and U24HL135600, and Wellcome Trust award H3A/18/001, and states that the funders had no role in study design, and analysis, decision to publish or preparation of the manuscript. B.T. acknowledges J. G. Camp for helpful discussions. E.E.M.F. is very grateful to A. Ephrussi, M. Mir, M. Perino, Y. Kherdjemil, T. Pollex and S. Secchia for useful comments. E. E. M. F is supported by European Research Council (Advanced Grant) agreement no. 787611 (DeCRyPT). E.S. is supported by grants from the European Research Council and the Israel Science Foundation. H.Y.C. is supported by NIH RM1-HG007735 and R35-CA209919. H.Y.C. is an investigator of the Howard Hughes Medical Institute. J.-S.K. is supported by the Institute for Basic Science (IBS-R021-D1). N.L-B. acknowledges funding from the European Research Council (Consolidator Grant 682398), the Spanish Ministry of Economy and Competitiveness (SAF2015-66084-R, European Regional Development Fund) and the Asociación Española Contra el Cáncer (GC16173697BIGA). S.A.T. is funded by NIH grants R35 GM134957-01 and NIAMS R01AR076241-01A1 and American Diabetes Association Pathway to Stop Diabetes grant #1-19-VSN-02.

Author information

Authors and affiliations.

Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX, USA

Amy L. McGuire

Broad Institute of MIT and Harvard, Cambridge, MA, USA

Stacey Gabriel & Alexander Meissner

Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA

Sarah A. Tishkoff

Department of Biology, University of Pennsylvania, Philadelphia, PA, USA

Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa

Ambroise Wonkam

Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa

Center for Human Genetics and Genomics, New York University Grossman School of Medicine, New York, NY, USA

Aravinda Chakravarti

European Molecular Biology Laboratory, Genome Biology Department, Heidelberg, Germany

Eileen E. M. Furlong

Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland

Barbara Treutlein

Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany

Alexander Meissner

Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA

Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany

Center for Personal Dynamic Regulomes, Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA

Howard Y. Chang

Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain

Núria López-Bigas

Research Program on Biomedical Informatics, Universitat Pompeu Fabra, Barcelona, Spain

Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain

Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel

Center for Genome Engineering, Institute for Basic Science, Daejon, Republic of Korea

Jin-Soo Kim

You can also search for this author in PubMed   Google Scholar

Corresponding authors

Correspondence to Amy L. McGuire , Stacey Gabriel , Sarah A. Tishkoff , Ambroise Wonkam , Aravinda Chakravarti , Eileen E. M. Furlong , Barbara Treutlein , Alexander Meissner , Howard Y. Chang , Núria López-Bigas , Eran Segal or Jin-Soo Kim .

Ethics declarations

Competing interests.

H.Y.C. is a co-founder of Accent Therapeutics and Boundless Bio and an advisor of 10x Genomics, Arsenal Biosciences and Spring Discovery. J.-S.K. is a co-founder of and holds stock in ToolGen Inc. A.C., A.L.M., A.M., A.W., B.T., E.E.M.F., E.S., N.L.-B., S.G. and S.A.T. declare no competing interests.

Additional information

Publisher’s note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Beyond 1 Million Genomes : https://b1mg-project.eu/

Blueprint Epigenome : https://www.blueprint-epigenome.eu/

cBioPortal for Cancer Genomics : https://www.cbioportal.org/

ENCODE : https://www.encodeproject.org/

Global Alliance for Genomics and Health : https://www.ga4gh.org/

gnomAD : https://gnomad.broadinstitute.org/

GTEx : https://www.gtexportal.org/home/

GWAS Catalog : https://www.ebi.ac.uk/gwas

H3Africa : https://h3africa.org

Roadmap Epigenomics Project : http://www.roadmapepigenomics.org/

Rights and permissions

Reprints and permissions

About this article

Cite this article.

McGuire, A.L., Gabriel, S., Tishkoff, S.A. et al. The road ahead in genetics and genomics. Nat Rev Genet 21 , 581–596 (2020). https://doi.org/10.1038/s41576-020-0272-6

Download citation

Accepted : 21 July 2020

Published : 24 August 2020

Issue Date : October 2020

DOI : https://doi.org/10.1038/s41576-020-0272-6

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Overcoming barriers to single-cell rna sequencing adoption in low- and middle-income countries.

  • Tracy Boakye Serebour
  • Adam P. Cribbs
  • Sarah J. B. Snelling

European Journal of Human Genetics (2024)

An overview of artificial intelligence in the field of genomics

  • Khizra Maqsood
  • Hani Hagras
  • Nicolae Radu Zabet

Discover Artificial Intelligence (2024)

IMA Genome – F19

  • Janneke Aylward
  • Andi M. Wilson
  • Brenda D. Wingfield

IMA Fungus (2024)

Current trends, limitations and future research in the fungi?

  • Kevin D. Hyde
  • Petr Baldrian
  • Arttapon Walker

Fungal Diversity (2024)

Improving variant calling using population data and deep learning

  • Nae-Chyun Chen
  • Alexey Kolesnikov
  • Andrew Carroll

BMC Bioinformatics (2023)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

latest genetics research

Scientists find a likely cause of many unexplained cases of intellectual disability: A genetic disorder

Close up of a finger silhouette pointing at the results of a DNA sequence analysis on a computer screen

A newly identified neurodevelopmental disorder may explain tens of thousands of cases of intellectual disability whose cause was previously unknown, according to a new study.

The research, published Friday in the journal Nature Medicine, investigates the effects of mutations in the gene RNU4-2, which is found in all animals, plants and fungi.

The gene plays an important role in gene splicing — the process of cutting out portions of genetic material and stitching others together. Ernest Turro, the new study’s senior author and an associate professor of genetics and genomic science at the Icahn School of Medicine at Mount Sinai, said that in theory, mutations in the RNU4-2 gene could disrupt that splicing process, ultimately leading to abnormal brain development and intellectual disability. 

This type of disability is characterized by significant limitations to a person’s ability to learn, reason, problem-solve, communicate or socialize, and it is often indicated by a low IQ. People with the disorder might also have seizures, motor delays, small heads, short stature or low muscle mass, according to the research.

The researchers hope that genetic tests for intellectual disabilities in children can quickly be updated to screen for the mutations. 

“A considerable number of families will finally be able to have a genetic diagnosis,” Turro said. 

Dr. Hakon Hakonarson, director of the Center for Applied Genomics at Children’s Hospital of Philadelphia, who was not involved in the study, said that because most cases of intellectual disability don’t have a known cause, the findings could “explain a good number of cases that are currently unexplained.”

The study estimates that up to 1 in 20,000 young people might have the condition. Researchers don’t know about the life expectancy associated with the disorder, so they have not estimated its prevalence among older adults, but Turro said some people with the genetic mutation have lived into adulthood.

The estimate suggests that the condition is slightly less common than Rett syndrome, a genetic disorder that causes babies to rapidly lose coordination, speech and mobility and affects about 1 in 10,000 female infants.

But Dr. Jeffrey Gruen, a professor of pediatrics and genetics at Yale School of Medicine who was not part of the research, said mutations in the RNU4-2 gene may turn out to be less common than the study suggests. He also questioned whether everyone with the mutations would have obvious learning or developmental issues.

“There are probably tens of thousands of people around the world that carry this, but does it cause intellectual disability in those tens of thousands? I don’t know,” he said. Gruen added, however, that the discovery is significant.

Hakonarson said the mutations probably cause at least some symptoms.

“The likelihood that this is disease-causing with these variants — which are not seen, by the way, in healthy people — is almost 100%,” he said.

The findings are based on data from the National Genomic Research Library, which contains information about the genomes — the entirety of a person’s genetic code — of people in the U.K. The study looked at the genomes of more than 77,000 participants.

Historically, studies of neurodevelopmental disorders have only looked at a small portion of the genome — specifically, so-called coding genes that are involved in the production of proteins. Of the 1,427 genes linked to intellectual disability, all but nine are coding genes.

Instead, Turro and his research team looked at noncoding genes — which don’t produce proteins — in about 5,500 people with intellectual disabilities. Mutations in the RNU4-2 gene were strongly associated with that group, compared with around 46,000 people who did not have intellectual disabilities. 

“There’s no question this paper is going to provoke a lot of studies now,” Hakonarson said. “People are going to go hunting for additional genes, because there’s a lot of noncoding RNA genes.”

The mutations in the RNU4-2 gene seem to occur at random, so they most likely can’t be passed from parent to child. For that reason, getting a diagnosis could be a comfort to parents who want to have more children, Turro said.

The researchers said it will be quite some time before they figure out whether the disorder can be treated with drugs or gene therapy.

“These are an extremely tough group of disorders to tackle therapeutically,” Andrew Mumford, a co-author of the study and research director of the South West England NHS Genomic Medicine Service, said on a call with reporters.

But even without an available treatment, he added, families often benefit from having a diagnosis.

“It helps them come to terms with the impact,” he said. “Being able to tell someone, ‘Yes, we have found the cause of development disorder in your child’ is incredibly powerful.”

Gruen said the discovery could also help connect families whose children have the same genetic condition so they can share stories and offer support.

“You could get some idea of what the future holds for them,” Gruen said. “Is this something that could be remediated? Can we expect there to be language? Can we expect there to be motor issues? That’s also very, very important to know.”

latest genetics research

Aria Bendix is the breaking health reporter for NBC News Digital.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Portland Press Opt2Pay

Logo of portlandopen

Recent developments in genetic/genomic medicine

Rachel h. horton.

Clinical Ethics and Law, Faculty of Medicine, University of Southampton, Southampton, United Kingdom

Anneke M. Lucassen

Advances in genetic technology are having a major impact in the clinic, and mean that many perceptions of the role and scope of genetic testing are having to change. Genomic testing brings with it a greater opportunity for diagnosis, or predictions of future diagnoses, but also an increased chance of uncertain or unexpected findings, many of which may have impacts for multiple members of a person’s family. In the past, genetic testing was rarely able to provide rapid results, but the increasing speed and availability of genomic testing is changing this, meaning that genomic information is increasingly influencing decisions around patient care in the acute inpatient setting. The landscape of treatment options for genetic conditions is shifting, which has evolving implications for clinical discussions around previously untreatable disorders. Furthermore, the point of access to testing is changing with increasing provision direct to the consumer outside the formal healthcare setting. This review outlines the ways in which genetic medicine is developing in light of technological advances.

Introduction

The past two decades have seen major shifts in our technical ability to sequence genetic information at scale. Historically, genetic testing tended to consist of either highly detailed molecular testing of nominated single genes, or broad genome-wide dosage screening at low resolution, for example karyotyping [ 1 , 2 ]. Genome sequencing was too slow and too expensive to be used in clinical contexts: for example the Human Genome Project, which was 99% complete in 2004, cost three billion dollars and took 13 years to sequence [ 3 ].

More recently, advances in sequencing technology have made it possible to undertake broad genetic testing on an individual patient basis within a clinically useful timeframe, via exome and genome sequencing. Exome tests sequence the entire protein-coding region of the genome, representing less than 2% of the genome but containing approximately 85% of known disease-causing variants [ 4 ]; genome sequencing encompasses the exome but also sequences all the non-protein-coding DNA. Initially implementation of such tests was via clinical research studies such as the Deciphering Developmental Disorders project [ 5 ], but more recently exome sequencing has been utilised as a clinical diagnostic test [ 6 ]. Genome sequencing is also due to transition to being available as a standard NHS test in June 2019, having previously only been available via initiatives such as the 100,000 Genomes Project [ 7 ].

Sequencing technology has improved in depth as well as breadth, and this has been of importance in better understanding cancer. The ability to sequence cancer genomes has led to rapid identification of driver mutations and has helped to work out the complex relationships between different cancer subclones over space and time, demonstrating the enormous heterogeneity of cancers and the difficulty of successfully treating them [ 8 ]. As sequencing techniques have advanced to the level where tiny amounts of tumour or individual cells can be sequenced, it has been possible to identify previously unknown mutational mechanisms, such as chromothripsis 1 [ 9 ] and kataegis 2 [ 10 ].

However, our ability to generate genomic data has substantially outstripped our ability to interpret its significance for an individual, and while improvements in genomic technology are in many cases driving improvements in healthcare, we are also encountering new problems as genomic testing shifts into the clinical setting. The Global Alliance for Genomics and Health (GA4GH) predicts that by 2025, over 60 million people will have had their genome sequenced in a healthcare context [ 11 ], but pathways for managing the output from genome sequencing are still in their infancy. The detailed but unfocused approach of genomic tests gives opportunities to answer questions that go beyond the problems that led to a patient having a test. However, deciding which of the multitude of possible outputs from genomic tests should be considered a ‘result’ at any given time is very challenging, not least because the links between many genetic variants and diseases are often unproven or poorly understood [ 12 ]. Multidisciplinary input and collaboration are increasingly key to interpreting the significance of genomic results. This review discusses the developments in practice that are evolving as a result of increasing use of genomic technologies.

New disease gene discovery and changing concepts of diagnosis

Exome and genome sequencing are powerful diagnostic tools – for example the Deciphering Developmental Disorders project, which recruited patients with severe undiagnosed disorders (who had generally already had any currently available diagnostic genetic testing), achieved a 40% diagnosis rate via trio exome sequencing for the first 1133 family trios in the study [ 13 ]. The search for a diagnosis has often been described as a journey [ 14 ], with parents of children with rare genetic disorders anticipating that a diagnosis may guide treatment, prognosis, acceptance and social support [ 15 ]. However, identification of new rare disease genes may be changing the impact of receiving a diagnosis, and in many cases very little is known about the long-term effects of newly identified genetic conditions.

Historically when making a genetic diagnosis, it has usually been possible to give families some information regarding prognosis, and to provide some parameters as to what to expect for the future, based on previous experience of what has happened for other children affected by the same condition. Now, while in some situations due to strong phenotypic match it is possible to be confident that a child’s rare disease has been caused by pathogenic variants in a recently described rare disease gene, often this provides little information about a child’s future.

We are increasingly in the position of learning about the effects of possible disease-causing variation(s) in a gene through meeting the patients in whom such genetic changes have been discovered. Often these changes will be in a gene newly thought to be linked to developmental disorders and there will be little, if any, published literature to draw on. We then have to speculate whether the genetic change detected is the cause of our patient’s health problems, and whether any additional difficulties that have happened for our patient that have not yet been noted in other patients with changes in the same gene are an extension of the phenotype of the newly described disease gene, or coincidental. In situations like this, we are often unable to give people information about what a new diagnosis might mean for them or their child in the longer term.

This has led to patient support and awareness groups taking on an increasingly important role [ 16 ], as families gather to share their lived experience of newly diagnosed rare genetic conditions, in turn informing clinical services. For example, the charity Unique works with families and professionals to develop specialist information relating to many rare and newly described genetic conditions, and to gather information about their long-term effects, increasing awareness and understanding of what it is like to live with rare genetic conditions. The rapidity with which such information can be gathered is also exemplified by the work of the PURA Syndrome Foundation: in 2014 the first patients with a rare condition called PURA syndrome were described in the medical literature [ 17 ]. Shortly afterwards the PURA Syndrome Foundation was established which has catalysed links between families, clinicians and researchers, greatly improving the speed and quality of research into the condition [ 18 ].

The agnostic approach of exome and genome sequencing is also challenging our previous concepts of existing genetic diagnoses, when apparently pathogenic variants are found in well-described disease genes but the patient’s clinical picture falls outside the boundaries of what we would conventionally expect for a patient affected by that particular genetic condition. For example, loss-of-function variants in SOX2 are known to cause anophthalmia and microphthalmia in addition to other phenotypes such as developmental delay and structural brain anomalies. Eye abnormalities were thought to be a key feature of SOX2 -related disorders, and so SOX2 would only be requested as a genetic test in patients who had absent or small eyes. Recently, via ‘genotype-first’ approaches, loss-of-function SOX2 variants have been found in people with developmental delay but without anophthalmia or microphthalmia, broadening the phenotypic spectrum associated with this gene [ 19 ]. Case Study 1 shows a further example where exome testing has extended previous perceptions of the clinical scope of a genetic condition.

Case Study 1

Redefining our understanding of genetic conditions (fictional case based on eggens et al. [ 20 ]).

An 8-year-old girl was referred to clinical genetics in order to investigate her progressive weakness. She had been floppy as a baby and from the age of 5 years had developed worsening limb weakness with frequent unusual movements, and difficulty in swallowing. Serial brain scans had shown progressive cerebellar atrophy.

Exome testing found that she was homozygous for a variant predicted to disrupt the function of EXOSC3 , a gene associated with pontocerebellar hypoplasia. This diagnosis had never been thought of as she did not have one of the defining characteristics: pontine hypoplasia. Her clinical picture also seemed atypical for this condition – most children with pontocerebellar hypoplasia do not survive infancy.

However, recent research has shown genotype–phenotype correlations in EXOSC3 -mediated pontocerebellar hypoplasia – patients homozygous for p.D132A variants (like this patient had) tend to have a milder clinical course and preservation of the pons. This genetic explanation fitted well in retrospect, but would not have been considered in advance of the exome test.

Key messages

  • Many well-recognised genetic conditions may have a wider spectrum of effects than previously thought.
  • Patients with genetic conditions identified via genomic tests may not conform to the pattern we expect based on experience of patients with the same condition identified via single gene testing. It can be very difficult to be sure whether this reflects an incorrect diagnosis, or a wider disease spectrum than previously recognised.

In many cases, our understanding of why the same genetic condition may be expressed so differently among different people is at an early stage, and this often makes genetic counselling very challenging, particularly in the prenatal setting. For some genetic conditions, it is becoming possible to provide more personalised risk estimates, based on combining knowledge of a person’s genetic diagnosis, with analysis of other factors that may influence their risk. Personalisation of risk in this way has generally been crude and reliant on clinically obvious characteristics: for example, men with pathogenic BRCA variants have a lower risk of developing breast cancer than women with pathogenic BRCA variants. More recently, genetic testing is being developed to complement ‘key’ genetic test results to provide an increasingly refined personal risk. For example, use of a polygenic risk score using breast cancer and ovarian cancer susceptibility SNPs identified via population GWAS showed large differences in absolute cancer risks between women with pathogenic BRCA variants with higher compared with lower polygenic risk score values [ 21 ]. This has yet to translate into routine clinical practice, but has the potential to help women with pathogenic BRCA variants make more informed decisions about how and when to manage their cancer risk.

The downsides of improved sensitivity: increased uncertainty in what tests mean

The prior probability of any one variant identified via genome sequencing being causative for a patient’s rare disease is extremely low. Attempts to catalogue human genetic variation, for example via the 1000 Genomes Project, show that a typical human genome differs from the reference human genome at 4.1–5 million sites [ 22 ]. Most of these variations will be entirely benign, some may subtly impact on risk of various common diseases, and a very small number will have the potential to cause serious disease either in an individual, or in their children (potentially in combination with variants inherited from their partner).

Genome sequencing identifies the majority of these variants, which then need careful filtering to produce a meaningful output. This has required a significant change in mindset from an era when most variants were identified in the context of carefully chosen single gene sequencing, and so had a much higher prior probability of being causative. There is an increasing shift towards a view that variants should be ‘innocent until proven guilty’ [ 23 ], but there is a lack of consensus regarding how to translate this principle into clinical practice.

There is also considerable discrepancy in how different genetics laboratories interpret the same variants. International guidelines for variant interpretation are helpful but insufficient to remove a great deal of noise when attempting to assign significance to particular findings [ 24 ]. This was illustrated in a recent study comparing variant classification among nine genetic laboratories: although they all used the same guidelines, only 34% of variants were given the same classification by all laboratories, and 22% of variants were classified so differently that different medical interventions would be recommended [ 25 ]. At a lower resolution level, even being sure of the relationship between genes and diseases is often difficult. For example, curation of the 21 genes routinely available on Brugada syndrome gene panels using the ClinGen gene curation scoring matrix found that only one of these genes was definitively linked to Brugada syndrome [ 26 ]. Our improving knowledge of variant interpretation leaves us with a difficult legacy, with many patients having been diagnosed incorrectly with genetic conditions. The effects of this can be far-reaching and difficult to undo, as illustrated by Case Study 2 .

Case Study 2

The legacy of incorrect diagnosis (case reported by ackerman et al. [ 27 ]).

A teenage boy died suddenly and genetic testing was then undertaken for his brother, resulting in the finding of a rare variant in KCNQ1 . On the basis of this test, the living brother was diagnosed with long QT syndrome, and the teenage boy’s sudden death was attributed to long QT syndrome. The living brother had an implantable cardioverter defibrillator inserted, and via cascade genetic testing over 24 relatives were diagnosed as having long QT syndrome, despite having normal QT intervals on ECG.

However, subsequent examination of post-mortem samples found that the boy who died had cardiac features inconsistent with long QT syndrome, did not have the KCNQ1 variant found in the wider family, and instead had a clearly disease-causing de novo variant in DES , a gene linked to cardiomyopathy.

  • It is very important to consider whether the clinical picture fits when evaluating variant significance: genetic variants will usually only predict disease well if found in the context of a medical or family history of the relevant disease.
  • Incorrect (or inappropriately deterministic) genetic test interpretation can affect the clinical care of a whole family, not just the person being tested.

Although this suggests that we need to be very cautious in making firm genetic diagnoses, it is difficult to know where the threshold should lie for communicating genetic variation of uncertain significance. There is some evidence that people find receiving a variant of uncertain significance surprising and disturbing, and some people misinterpret it as being definitely pathogenic or definitely benign [ 28 ]. However, there is also evidence that many people have a strong desire to receive a broad range of results from genetic testing, including uncertain results, and are uncomfortable with the idea that decisions about non-disclosure might be made without involving them [ 29 ].

The fear is that disclosure of uncertain variants will lead to over-diagnosis and over-management, with variants inappropriately being treated as if pathogenic. Excessive and inappropriate interventions (not to mention anxiety and distress) might then cascade through families, going against one of the fundamental principles of medicine to ‘first do no harm’. However, we also fear missing something or being accused of ‘hiding information’. The result is that we tend to end up in purgatory, documenting uncertain variants on lab reports (though sometimes not) and having lengthy conservations with patients about them (though sometimes not), then tacking on a caveat that ‘maybe this means nothing’. This nominally shifts the responsibility to the next person in the chain but feels unsatisfactory for all concerned.

Uncertainty when to stop looking and what to communicate

Another issue arising from improved sensitivity is the ability to find genetic variants that are unrelated to the clinical problem that a patient presents with, but that may be relevant for their health in other ways. This may be viewed as positive or negative, but working out how to handle this information raises difficult questions. In 2013, the American College of Medical Genetics and Genomics (ACMG) suggested that laboratories should automatically seek and report pathogenic variants in 56 genes associated with ‘medically actionable’ conditions when performing clinical sequencing [ 30 ]. The main rationale was the potential to benefit patients and families by diagnosing disorders where preventative measures and/or treatments were available, with the aim of improving health. However, these recommendations proved controversial. The main debate at the time centred around whether patients should have a right to choose not to know such information [ 31 ]. Subsequent questions about the role of clinicians in offering additional findings, what constitutes a ‘medically actionable’ finding, and what is the predictive value of such findings in the absence of a phenotype or family history of the relevant disorder, are yet to be fully addressed.

Analysis of data from the 1000 Genomes cohort demonstrated that approximately 1% of ‘healthy’ people will have a ‘medically actionable’ finding in one of the 56 genes [ 32 ]. However, what this might mean on an individual basis is often unclear. Most of our knowledge regarding the effects of variation in any given gene has been gathered by observing people who have been identified as having variants in the gene because they were tested as they had a personal history or family history of disease, biasing the sample from which our conclusions are drawn. It is less clear what it might mean to find, for example, an apparently pathogenic variant in a gene linked to cardiomyopathy in a person with no personal or family history of heart problems. This has important implications for ‘cascade screening’, where relatives of a patient affected by a condition with a known genetic cause are offered testing to see whether they have the disease-causing genetic variant that was found in their clinically affected family member (meaning that they may also be at risk of developing the disease). To what extent should testing and subsequent screening be offered in a family based on an incidental finding of a genetic variant thought to be predictive of a particular condition, if there is no clinical evidence that anyone in the family, including the person in whom the genetic variant in question was first identified, is actually affected by it?

Broad genomic testing also has the potential to detect carrier status for recessive and X-linked conditions. From population studies, we know that being a carrier for a genetic condition is very common. For example, a gene panel testing carrier status for 108 recessive disorders in 23453 people found that 24% were carriers for at least one of the 108 disorders, and 5.2% were carriers for multiple disorders [ 33 ]. On a disorder-by-disorder status, being a carrier for a genetic condition is very rare (with notable exceptions such as haemochromatosis and cystic fibrosis), but when considered collectively, it is ‘normal’ to be a carrier for a genetic condition. For most people, being a carrier will have no impact on their life at all. However, if their partner happens to be a carrier for the same condition then the implications could be very profound, as each of their children would have a one in four chance of being affected by the genetic condition. This is particularly relevant for couples who are known to be biologically related [ 34 ], and couples with common ancestry, as they will have a higher chance of both being carriers for the same recessive condition. Carrier screening for various autosomal recessive diseases has been available in some instances for many years, for example screening for carrier status for Tay–Sachs disease for people of Ashkenazi Jewish ancestry has been offered since the 1970s [ 35 , 36 ]. More recently, advances in technology have led to development of expanded carrier screening tests, which check carrier status for multiple diseases simultaneously and are often less targeted towards particular genetic populations [ 37 ].

The increased scope of carrier screening, combined with the recognition that it is very common to be a carrier for one or more recessive genetic conditions, has led to an increasing move to consider carrier results for recessive genetic conditions on a couple basis, where carrier status is only communicated if it would be relevant in the context of a particular relationship (i.e. if both people in a couple are carriers for the same condition) [ 38 ]. This avoids pathologising the status of ‘being a carrier’, recognising that most of us are carriers for some genetic conditions, and conserves resources for genetics services by not flooding the system with large volumes of individual carrier results, most of which will be meaningless in the context of that individual’s life. Objections to this approach are that by not communicating individual carrier results, a person would not know this information for future relationships, and their family could not access cascade screening to see whether they are also carriers. However, these objections could be obviated by widespread adoption of couple carrier testing – a person (or their close relatives) could find out their carrier status if relevant when they next had a couple carrier test in the context of their new relationship. In some ways, this could be seen as comparable with management of infectious disease – lots of healthy people carry MRSA, but very few die of MRSA infection. People are therefore screened at times when they might be especially vulnerable to becoming unwell from MRSA, or when they might pass it on to others at risk, for example when admitted to hospital, rather than being tested at random points when they are generally well.

The expanding remit and availability of genetic technology

‘acute genetics’.

For many years, clinical genetics input has at times influenced acute care, for example in diagnosing trisomies in the neonatal period, or informing the care of babies born with ambiguous genitalia. However in many circumstances, the key contribution of clinical genetics was in providing a post hoc explanation for serious medical problems, rather than in influencing treatment decisions on a real-time basis. This is changing as the availability of exome and genome sequencing increases, as shown by Case Study 3 . A recent study in a neonatal intensive care unit in Texas studied outcomes for 278 infants who were referred for clinical exome sequencing, and found that 36.7% received a genetic diagnosis, and medical management was affected for 52% of infants with diagnoses [ 39 ]. There is increasing evidence that this approach is cost-effective: for example, a prospective study of exome sequencing for infants with suspected monogenic disorders found that standard care achieved an average cost per diagnosis of AU$ 27050, compared with AU$ 5047 for early singleton exome sequencing [ 40 ]. Similarly, ‘real-time’ genetic and genomic testing is making an impact in cancer treatment, where in many cases testing is available to help guide treatment choices by identifying actionable genetic variants in tumours that may respond to specific therapies [ 41 , 42 ].

Case Study 3

Insights from exome testing transforming a clinical course (case from wessex genomic medicine centre [ 43 ]).

A young woman was referred for exome testing having spent months in a coma. From childhood she had experienced sensory problems, and as a young adult she had gone on to develop seizures which deteriorated into status epilepticus, necessitating ventilation on intensive care.

After 3 years during which all other avenues had been explored, analysis of her exome was proposed. An unexpected diagnosis of pyridoxine-dependent epilepsy was found; this had not previously been considered as classically it causes seizures in the first few months of life. She began treatment with pyridoxine (vitamin B 6 ). From that point on she had no further seizures and her clinical situation transformed. Over a 6-month period she was weaned off all of her anti-epileptic drugs, and was able to return to a normal life.

Key message

  • Exome or genome tests have the potential to make an enormous difference to clinical care and to people’s lives.

Pharmacogenomics

As well as guiding treatment choice, genetic testing will increasingly influence what doses are prescribed, and whether medications are considered unsuitable in view of a high risk of an adverse reaction. Around the time that the Human Genome Project was completed, there was considerable excitement about the possibility of genetic testing guiding use of medication in the clinic [ 44 , 45 ]. The potential of genotype-driven drug dosing has for the most part yet to be realised, in part because the interaction of the genetic factors involved is sometimes complex, and in part because environmental factors may also have a significant impact on how a person responds to a drug. For example, genotype-driven prescription of warfarin, which has notoriously wide inter-individual variation in dosage requirements, largely remains in the realm of research [ 46 ].

However, for some drugs, pharmacogenomics has already had a significant impact in reducing morbidity and mortality. For example, when the antiretroviral drug abacavir was first introduced, approximately 5% of the people treated developed an idiosyncratic hypersensitivity reaction that could be life-threatening on repeated exposure to the drug [ 47 , 48 ]. Research established that immunologically confirmed hypersensitivity reactions to abacavir only occurred in people with the HLA-B*5701 allele, and a clinical trial went on to show that pre-screening patients to check that they did not have HLA-B*5701 prior to starting the drug led to no confirmed hypersensitivity reactions in the pre-screened arm, while 2.4% of the unscreened patients had reactions [ 49 ]. Patients are now screened for HLA-B*5701 as standard before starting abacavir treatment [ 50 ]. Similar screening is likely to become more widespread as we learn more about genetic risk factors for adverse drug reactions. For example, there are increasing suggestions that the mitochondrial variant m.1555A>G should be checked in patients with cystic fibrosis in order to guide antibiotic treatment choices, in view of the evidence that people with this variant may develop hearing loss when exposed to aminoglycosides [ 51 ].

Evolving options in prenatal genetics

Genetic testing is also being used more extensively in the prenatal setting, in part because of developments in non-invasive prenatal testing and diagnosis, which allow genetic screening or testing of a developing pregnancy by doing a blood test for the mother [ 52 ]. This removes the risk of miscarriage associated with conventional prenatal tests (chorionic villus sampling or amniocentesis). While this is in some ways a stride forward, it raises various ethical issues, as the technical test safety may lead to such testing becoming viewed as routine. This raises the concern that couples will give less careful consideration as to whether they really want to know the results before having such tests, and that women may feel that there is an expectation that they should have testing. The worry is that this could potentially lead to people feeling under pressure to terminate pregnancies in response to genetic test results (including in situations where the clinical implications of the results may be far from clear) [ 53 ].

Widening access to genetic testing within healthcare

The expanding options for genetic testing and the escalating expectation for quick results to drive clinical management mean that testing provision is increasingly being pushed out of highly specialised genetics centres into mainstream medicine. For example, many women with ovarian cancer will now be offered BRCA testing via their oncology team, and only referred to genetics if needed based on the test results [ 54 ]. Genetics appointments now frequently focus on interpretation of tests already done, working out if the test outcome seems to match the clinical problem, and arranging testing and surveillance for family members.

The rise of direct-to-consumer genetic testing

As clinical services have increasingly grown to expect and demand genetic answers for patients with complex health problems, on a broader societal level the hunger for genetic information also seems to be increasing. However this is occurring in the context of a public discourse about personalised/precision medicine and genetics that tend to enthusiastically promote it in a very optimistic light, rarely dwelling on potential concerns and limitations, and therefore potentially sculpting inappropriate expectations from technology that is still being developed [ 55 ].

Direct-to-consumer tests currently sit outside much of the regulation that governs clinical genetic testing, but claim to provide insight into issues as diverse as ancestry, nutrition, athletic ability, and child talent [ 56 ]. Many testing providers also claim to help provide insight on health, though the information provided by many direct-to-consumer companies is far from comprehensive. For example, a recent analysis of 15 direct-to-consumer genetic testing companies advertising to U.K. consumers found that none of them complied with all the U.K. Human Genetics Commission principles for good practice regarding consumer information [ 57 ]. There are also examples that might make us reflect sceptically on the value of these tests – for example a case where a family sent a sample from their dog to a direct-to-consumer testing company designed to provide insights on people’s genetic ‘superpowers’ and received a report which did not mention that the sample was not human but conjectured that the client would be talented at basketball [ 58 ].

‘DIY genetics’ has also risen in popularity, with people asking for raw data from direct-to-consumer companies then processing this themselves via third-party interpretation services, as discussed in Case Study 4 . Approximately 40% of genetic changes in direct-to-consumer test raw data sent for clinical confirmation are false positives [ 59 ], but this is often not appreciated by customers or the doctors they may subsequently visit, leading to anxiety and often inappropriate medical interventions [ 60 ]. However, clearly many people see a value in receiving genetic information and are prepared to pay for this. This marks a shift from genetic testing in order to explain health problems or for people at high risk of developing specific genetic conditions, to testing of healthy people with the rationale of facilitating life planning. This idea has been taken to the extreme with initiatives such as the BabySeq project, exploring the medical, behavioural and economic impacts of integrating genome sequencing into the care of healthy newborns [ 61 ].

Case Study 4

Grime on the crystal ball (fictional case based on moscarello et al. [ 60 ]).

A healthy medical student was given a direct-to-consumer genetic test for Christmas, and explored the raw data from this test using an online interpretation programme, finding a variant in MYBPC3 that was predicted to cause hypertrophic cardiomyopathy. He was understandably worried by this result, taking time off university as he came to terms with it, and giving up running, which he used to really enjoy.

He was seen in a hypertrophic cardiomyopathy clinic and had an expert cardiology assessment including ECG, echocardiogram and review of his family history. He was found to have no clinical evidence of hypertrophic cardiomyopathy, and further genetic testing showed that he did not actually have the disease-causing MYBPC3 variant that the online interpretation programme had identified. However, he continued to feel anxious about his risk of heart problems and decided to give up running permanently.

  • Information provided from direct-to-consumer testing may be unreliable, especially where online interpretation programmes are used to further explore the raw data from the test: the level of quality control may be very different from that of accredited genetic laboratories, increasing the likelihood of false positives, false negatives and sample mix-up.
  • Many direct-to-consumer genetic tests involve no meaningful pre-test counselling – people are often totally unprepared for the information that might come out of such testing (and are unaware that it might be wrong).

Genetic information as family information

The familial nature of genetic information has always generated discussion as to how to respect the confidentiality of individual patients while ensuring that their close relatives have access to information that may be relevant for their own health and life choices. Clinical guidance in this area has increasingly taken the stance that genetic information should be confidential to families, not individuals (though the personal consequences of having a genetic change for a given individual should be confidential to them alone) [ 62 ].

The consequences of this shift are still being navigated in the clinical setting – research indicates that patients often see genetic information as belonging to their family rather than exclusively to them [ 63 ], but healthcare professionals are often reticent about taking a familial approach to the confidentiality of genetic information in practice, worrying that this stance could disrupt family dynamics or erode patient trust in the health service [ 64 ]. A recent BMJ poll which asked, ‘Are there situations when sharing a patient’s genetic information with relatives without consent is acceptable?’ demonstrated the current split in opinion, with 51% of respondents answering ‘yes’ and 49% ‘no’ [ 65 ]. The personal versus familial nature of genetic information is currently being tested in the courts via the ABC case, which centres around non-disclosure of genetic risk to the daughter of a patient with Huntington’s disease [ 66 ].

Treatment for genetic disorders

One of the most exciting recent developments in genetics and genomics is the prospect of treatment for an increasing number of genetic conditions. However this topic has to be treated with caution as the practical reality for many patients and families is that though promising research is ongoing, meaningful treatment is not possible in many cases. Even in situations where evidence-based treatments have been developed, the expense of many of these therapies risks making them inaccessible.

Many different approaches have been taken to try to treat genetic conditions. Gene therapy, which involves delivering functional genetic code, is one approach but its success has been widely variable, often due to difficulty in developing vectors that can deliver genetic material into affected tissues at sufficiently high levels without being destroyed by the immune system. In certain situations this approach can be highly effective, for example promising results have been achieved in various eye conditions, likely because eyes are small and easily accessible, and have a privileged relationship with the immune system [ 67 ]. In cases aiming to deliver gene therapy to a wider area, such as the lungs or the muscles, treatment attempts have generally proved more challenging [ 68 , 69 ].

Other approaches include use of small molecules to modify various steps in the pathway from gene to functional product. For example, Eteplirsen aims to treat Duchenne muscular dystrophy in certain patients by influencing splicing machinery to skip exon 51 from mature DMD mRNA, restoring a more functional reading frame so that a shortened version of dystrophin can be successfully translated [ 70 ]. Ivacaftor potentiates the action of CFTR channels in some patients with cystic fibrosis (G551D pathogenic variant) [ 71 ]. Enzyme replacement therapy is being trialled to treat children with mucopolysaccharidoses, for example idursulphase infusions in mucopolysaccharidosis type 2 [ 72 ].

While lots of these therapies are very exciting and show demonstrable changes at the molecular level in clinical trials, these cellular changes do not always clearly translate into improvements in clinically relevant outcomes. The therapies are also often hugely expensive, which raises very difficult ethical questions regarding whether limited resources should be spent on such treatments where there is often only limited proof of clinical efficacy.

However the increasing possibility of future treatments for genetic conditions is influencing clinical decisions around the care of very ill children. For example, recently nusinersen has shown promise as a treatment for some children with spinal muscular atrophy, but this may begin to raise new questions about whether interventions such as intubation and tracheostomy should be offered to infants with severe spinal muscular atrophy, where previously these would have been considered medically inappropriate [ 73 ]. This has consequences for the clinical conversations happening when these diagnoses are made. In the past, breaking news of such a diagnosis might flow naturally into discussions around palliation. The possibility of treatment now creates new options to consider, but also new challenges in considering with parents how best to care for their child [ 74 ]. The clinical impact and accessibility of emerging treatments is often very uncertain, but parents may prefer to explore even extremely long-shot treatments over accepting a palliative care pathway route, and may expect or seek crowd funding for experimental treatments for which there is as yet very little, if any, evidence of benefit.

Improving genetic technology has also had a significant impact on fertility services, ranging from pre-implantation genetic diagnosis to mitochondrial donation, offering new options for families affected by genetic conditions [ 75 , 76 ]. Increasing technological capability is set to extend the theoretically possible range of options – for example last year a group in China used the CRISPR/Cas9 system to correct pathogenic variants in the HBB and G6PD genes in human zygotes [ 77 ], though the efficiency and accuracy of the correction procedure was variable. This emerging possibility raises significant ethical issues which need debate. A recent report of the Nuffield Council on Bioethics on genome editing in the context of human reproduction suggested that there may be certain contexts in which this may be ethically acceptable, provided that such interventions were intended to secure the welfare of a person who may be born as a result, and that any such interventions would uphold principles of social justice and solidarity [ 78 ].

Conclusions

Insights from genomic technology have great potential to improve health, but we are currently going through a teething process in learning how to respond to the nebulous information that genomic tests can provide in the clinical setting. In part, this learning process is being driven by patients and families, with patient support groups coming to the fore in an era where we can now make extremely rare diagnoses that link different families across the world, but often have very little information on what this might mean for the future. Our current response to the outcomes from genomic tests is often reactive and ad hoc, partly because we are still learning how to interpret genomic variation and are often unable to gain a consensus on whether genetic variants are clinically significant or not. This situation is exacerbated by the different routes in which genomic information is now accessible – rapid tests to establish diagnosis or plan treatment for patients are now a reality in the real-life clinical setting, but healthy people also have increasing access to commercial tests that claim to provide genetic information to improve health and life planning. This raises particular challenges in the context of a public discourse about genomics that tends to present it as far more predictive and certain than it actually is. Some of the most exciting recent developments in genomic medicine relate to potential future treatments and reproductive options for people and families affected by rare genetic conditions. However hurdles relating to treatment efficacy and optimal timing of treatment, mean that we need to keep these advances in perspective and consider how to research potential treatments responsibly, avoiding creating hype that undermines the ability of families to make a balanced decision whether or not to participate in this research. It is also important to consider financial sustainability, avoiding situations where useful new treatments are developed that remain inaccessible to the patients who need them on account of their cost. To summarise, the introduction of genomic testing is having a big impact on patient care, but raises various issues that need further study and debate in order to help us maximise the potential benefits of genomic medicine while minimising the possible harms.

Acknowledgments

We thank the patient in Case Study 3 for her help with the Case Study box and for sharing her story.

Abbreviations

1 Complex chromosome rearrangements, thought to occur due to single catastrophic events where chromosomes ‘shatter’ and are repaired by error-prone mechanisms.

2 Clusters of localised mutations.

This work was supported by funding from a Wellcome Trust collaborative award [grant number 208053/Z/17/Z (to A.L.)].

Competing interests

The authors declare that there are no competing interests associated with the manuscript.

  • U.S. Department of Health & Human Services

National Institutes of Health (NIH) - Turning Discovery into Health

  • Virtual Tour
  • Staff Directory
  • En Español

You are here

Nih research matters.

June 4, 2024

Gene variants and breast cancer risk in Black women

At a glance.

  • In the largest study of its kind, researchers identified genetic variants that appear to boost breast cancer risk among females of African ancestry.
  • The findings could help improve risk prediction in this population and lead to more targeted therapies and prevention strategies.

Adult daughter kissing smiling mother.

Breast cancer is the most often diagnosed cancer in many parts of the world, including the U.S. More than 310,000 new cases are expected nationwide this year.

Black women tend to develop breast cancer at a younger age than White women. Black women are also more likely than Whites to die from the disease, and they are twice as likely to develop an aggressive subtype called triple-negative breast cancer. But despite the increased risks faced by women of African descent, most large-scale genetic studies of breast cancer to date have focused on women of European ancestry.

To better understand their unique genetic risks, a research team led by Dr. Wei Zheng of Vanderbilt University analyzed genetic data from over 40,000 females of African descent. About 18,000 had been diagnosed with breast cancer. The data were gathered as part of the NIH-funded African Ancestry Breast Cancer Genetic consortium, which combined data from 26 studies. Most participants (85%) were African Americans. The rest were from Barbados or Africa.

The researchers conducted a genome-wide association study (GWAS) to look for genetic variants that are found more often in participants with breast cancer than in those without. This is believed to be the largest GWAS study to date of breast cancer in this population. Results were reported in Nature Genetics on May 13, 2024.

The analysis pinpointed 12 genetic regions, or loci, associated with breast cancer. Three of these loci were linked to the aggressive triple-negative cancer. About 8% of the women carried two genetic copies of risk variants in all three of these loci. Such women, the researchers found, were 4.2 times more likely to be diagnosed with triple-negative breast cancer than women who had only one or no copies of the variants.

Because this type of cancer lacks specific cell receptors often seen with breast cancer (like estrogen or HER2 receptors), there are fewer targeted options for treatment. These findings may help researchers identify new treatment targets.

The researchers also confirmed many breast cancer risk variants that were found earlier in other populations. And they identified an uncommon risk variant in the gene ARHGEF38 , which had been previously linked to aggressive prostate and lung cancers.

The scientists used their findings to create polygenic risk scores (PRS) for breast cancer risk in females of African descent. PRS use genomic data to gauge the chance that a person will develop a certain medical condition. PRS created previously, using results from other populations, tend to perform poorly at predicting breast cancer risk for Black women. The new PRS, based on genomic data from African descendants, outperformed previous PRS at predicting breast cancer risk in this population.

The findings and data could lead to improved detection of breast cancer in this at-risk population and provide clues for potential treatment targets. Studies with even larger, more diverse populations will be needed to further improve the prediction of breast cancer risk.

“We have worked with researchers from more than 15 institutions in the U.S. and Africa to establish this large genetic consortium,” Zheng says. “Data put together in this consortium have been and will continue to be used by researchers around the world.”

—by Vicki Contie

Related Links

  • Human Pangenome Boosts Accuracy and Reflects Diversity
  • Technique May Improve Detection of Breast Tumors
  • Test Predicts Whether Chemotherapy Will Help Early-Stage Breast Cancer Patients
  • Advances in Breast Cancer: Screening and Treatment Get Personal
  • Breast Cancer
  • Polygenic Risk Scores

References:  Genome-wide association analyses of breast cancer in women of African ancestry identify new susceptibility loci and improve risk prediction. Jia G, Ping J, Guo X, Yang Y, Tao R, Li B, Ambs S, Barnard ME, Chen Y, Garcia-Closas M, Gu J, Hu JJ, Huo D, John EM, Li CI, Li JL, Nathanson KL, Nemesure B, Olopade OI, Pal T, Press MF, Sanderson M, Sandler DP, Shu XO, Troester MA, Yao S, Adejumo PO, Ahearn T, Brewster AM, Hennis AJM, Makumbi T, Ndom P, O'Brien KM, Olshan AF, Oluwasanu MM, Reid S, Butler EN, Huang M, Ntekim A, Qian H, Zhang H, Ambrosone CB, Cai Q, Long J, Palmer JR, Haiman CA, Zheng W. Nat Genet. 2024 May;56(5):819-826. doi: 10.1038/s41588-024-01736-4. Epub 2024 May 13. PMID: 38741014.

Funding:  NIH’s National Cancer Institute (NCI).

Connect with Us

  • More Social Media from NIH
  • Introduction to Genomics
  • Educational Resources
  • Policy Issues in Genomics
  • The Human Genome Project
  • Funding Opportunities
  • Funded Programs & Projects
  • Division and Program Directors
  • Scientific Program Analysts
  • Contact by Research Area
  • News & Events
  • Research Areas
  • Research investigators
  • Research Projects
  • Clinical Research
  • Data Tools & Resources
  • Genomics & Medicine
  • Family Health History
  • For Patients & Families
  • For Health Professionals
  • Jobs at NHGRI
  • Training at NHGRI
  • Funding for Research Training
  • Professional Development Programs
  • NHGRI Culture
  • Social Media
  • Broadcast Media
  • Image Gallery
  • Press Resources
  • Organization
  • NHGRI Director
  • Mission & Vision
  • Policies & Guidance
  • Institute Advisors
  • Strategic Vision
  • Leadership Initiatives
  • Diversity, Equity, and Inclusion
  • Partner with NHGRI
  • Staff Search

Current Clinical Studies

Researchers at the National Human Genome Research Institute (NHGRI) are working with patients and families to better understand of how genes can cause or influence diseases and develop new and more effective diagnostics and treatments.

Value of Clinical Studies

Clinical studies give us a better understanding of how genes can cause or influence diseases. NHGRI researchers are working with patients, and with families with a history of inherited diseases, to learn more about the genetic components of common and rare disorders, and to develop new and more effective tests and treatments.

Deciding whether to participate in a clinical study is an important and personal process. Some reasons people choose to participate include:

  • Participants in clinical studies help current and future generations. Through these studies, researchers develop new diagnostic tests, more effective treatments, and better ways of managing diseases with genetic components.
  • Participants in studies are actively involved in understanding their disorder and current research.
  • Participants in some studies gain access to new tests and treatments before they are widely available.

Featured Clinical Studies

ADHD Genetic Research Study | NHGRI

Metabolism, Infection and Immunity (MINI) Section

The MINI section aims to define the relationship between infection, immunity and clinical decline in individuals with mitochondrial disease.

COVID-19 MINI

Other Clinical Studies

The following are conducted by NHGRI researchers. For eligibility requirements and contact information, visit the study on clinicaltrials.gov.

Last updated: January 12, 2023

Together we are beating cancer

  • Cancer types
  • Breast cancer
  • Bowel cancer
  • Lung cancer
  • Prostate cancer
  • Cancers in general
  • Clinical trials
  • Causes of cancer
  • Coping with cancer
  • Managing symptoms and side effects
  • Mental health and cancer
  • Money and travel
  • Death and dying
  • Cancer Chat forum
  • Health Professionals
  • Cancer Statistics
  • Cancer Screening
  • Learning and Support
  • NICE suspected cancer referral guidelines
  • Make a donation
  • By cancer type
  • Leave a legacy gift
  • Donate in Memory
  • Find an event
  • Race for Life
  • Charity runs
  • Charity walks
  • Search events
  • Relay for Life
  • Volunteer in our shops
  • Help at an event
  • Help us raise money
  • Campaign for us
  • Do your own fundraising
  • Fundraising ideas
  • Get a fundraising pack
  • Return fundraising money
  • Fundraise by cancer type
  • Set up a Cancer Research UK Giving Page
  • Find a shop or superstore
  • Become a partner
  • Cancer Research UK for Children & Young People
  • Our Play Your Part campaign
  • Brain tumours
  • Skin cancer
  • All cancer types
  • By cancer topic
  • New treatments
  • Cancer biology
  • Cancer drugs
  • All cancer subjects
  • All locations
  • By Researcher
  • Professor Duncan Baird
  • Professor Fran Balkwill
  • Professor Andrew Biankin
  • See all researchers
  • Our achievements timeline
  • Our research strategy
  • Involving animals in research
  • Research opportunities
  • For discovery researchers
  • For clinical researchers
  • For population researchers
  • In drug discovery & development
  • In early detection & diagnosis
  • For students & postdocs
  • Our funding schemes
  • Career Development Fellowship
  • Discovery Programme Awards
  • Clinical Trial Award
  • Biology to Prevention Award
  • View all schemes and deadlines
  • Applying for funding
  • Start your application online
  • How to make a successful applicant
  • Funding committees
  • Successful applicant case studies
  • How we deliver research
  • Our research infrastructure
  • Events and conferences
  • Our research partnerships
  • Facts & figures about our funding
  • Develop your research career
  • Recently funded awards
  • Manage your research grant
  • Notify us of new publications
  • Find a shop
  • Volunteer in a shop
  • Donate goods to a shop
  • Our superstores
  • Shop online
  • Wedding favours
  • Cancer Care
  • Flower Shop
  • Our eBay store
  • Shoes and boots
  • Bags and purses
  • We beat cancer
  • We fundraise
  • We develop policy
  • Our global role
  • Our organisation
  • Our strategy
  • Our Trustees
  • CEO and Executive Board
  • How we spend your money
  • Early careers
  • Your development

Cancer News

  • For Researchers
  • For Supporters
  • Press office
  • Publications
  • Update your contact preferences
  • About cancer
  • Get involved
  • Our research
  • Funding for researchers

The latest news, analysis and opinion from Cancer Research UK

  • Science & Technology
  • Health & Medicine
  • Personal Stories
  • Policy & Insight
  • Charity News
  • Science & Technology

At-home saliva test could help diagnose prostate cancer sooner

Tim Gunn

1 June 2024

A new saliva test for identifying men at high risk of prostate cancer could help find more cases of the disease earlier, when doctors have a better chance of treating it successfully.

The researchers behind the BARCODE 1 study, which we helped fund, say their test could help “turn the tide” on prostate cancer.

Although it is the second biggest cancer killer of men in the UK, taking around 12,000 lives a year, there is no national screening programme for prostate cancer . This is because the only current option, the prostate-specific antigen (PSA) blood test , is too inaccurate.

Instead of measuring the amount of PSA in blood, the new spit test sorts people into groups by looking through the DNA in their saliva samples for a range of small genetic changes linked to prostate cancer. The results of the trial suggest that this approach works better. The men the new test puts in its highest risk group are more likely to have prostate cancer than men with raised PSA levels.

The new test also falsely identified prostate cancer fewer times than the PSA test does, and picked up a higher proportion of aggressive cancers.

“With this test, it could be possible to turn the tide on prostate cancer,” said Ros Eeles, a professor at the Institute of Cancer Research and a consultant at the Royal Marsden NHS Foundation Trust, which jointly led the research. ”We have shown that a simple, cheap spit test to identify men at higher risk due to their genetic makeup is an effective tool to catch the cancer early.”

Eeles is presenting the results of BARCODE 1 at the annual American Society of Clinical Oncology meeting in Chicago. She ties her work to a long legacy of research into the genetic markers of prostate cancer.

“Building on decades of research into the genetic markers of prostate cancer, our study shows that the theory does work in practice – we can identify men at risk of aggressive cancers who need further tests, and spare the men who are at lower risk from unnecessary treatments.”

From PSA testing to polygenic risk scoring

In the study, spit samples were used to calculate prostate cancer polygenic risk scores (PRSs) for more than 6,000 European men. All participants were recruited through their GP surgeries when they were between 55 and 69 – an age at which risk of prostate cancer is increased.

Their PRSs were based on 130 genetic variations – many hereditary – shown to be linked with prostate cancer through studies into the DNA of hundreds of thousands of men.

For BARCODE 1, the men with the highest 10% of risk scores were invited to further screening. Following an MRI and a prostate biopsy, 187 of them (40% of the total) were diagnosed with prostate cancer. That’s a significant jump from the 25% of men identified by PSA tests who actually have prostate cancer. Moreover, 147 (78%) of the men diagnosed thanks to the new saliva test had a ‘normal’ PSA level, which would usually indicate that no further screening is required.

The researchers also took a closer look at how the prostate cancers picked up by their new test behaved (which can be assessed by looking at how abnormal they appear, measured by grade ). PSA testing picks up many people who have cancers that grow too slowly to cause any significant health impacts, meaning that men may undergo unnecessary MRI scans, invasive biopsies, and treatments. Importantly, then, the new spit test identified a higher proportion of aggressive cancers – which are fast growing and likely to spread – than the PSA test. Of the 187 cancers detected in BARCODE 1, 55% were aggressive cancers, compared with 36% of those identified by a PSA test in a recent study.

The PRS test is also more accurate than an MRI scan for men who score in the highest 10% for genetic risk.

Naser Turabi, our director of evidence and implementation, put the findings into context.

“Right now, there’s no reliable method to detect aggressive prostate cancer, but this study brings us a step closer to finding the disease sooner in those people who need treatment,” he said. “It’s encouraging to see that genetic testing might help to guide a more targeted approach to screening based on someone’s risk of developing prostate cancer. More research is now needed to confirm if this tool can save lives from the disease so that it can be rolled out to improve diagnosis.”

Since BARCODE 1 started, an international research team has identified more genetic variants associated with prostate cancer risk in men of Asian and African ancestry. The ICR team intend to trial a saliva test for this population to ensure polygenic risk scoring can benefit all men. They are also comparing the saliva test to other potential screening options as part of the ongoing TRANSFORM trial.

This research was funded by the European Research Council, the Bob Willis Fund, Cancer Research UK, The Peacock Trust and the National Institute for Health and Care Research (NIHR) Biomedical Research Centre (BRC) at The Royal Marsden and The Institute of Cancer Research (ICR).

Amazing news, I pray this will be commercially available soon in Ireland, we are years behind in Ireland with genetic testing.

Does that mean no more biopsies? (Breathes sigh of relief) Uses in detection for other cancers?

fantastic reseach

Excellent Idea as I think many men are embarrassed I feel it would stop a lot of worry

This hopefully a game changer and fully on board. Unfortunately men are simply not being heard or seen by there GP , just simply not good enough four week wait, zero to do with covid, many many people young old falling through the inadequacy’s from all cancers.

I would like to thank you so very much for this amazing research you are carrying out. I personally have had prostate cancer but I was treated successfully if this can save more men from dying from this horrible disease it is a truly life changing moment.

If test preliminary test need to be rolled out to a larger test group, I’d be happy to take part. To many men die unnecessarily due to this cancer so this is a great step forward.

The sooner we adopt this method of screening in the UK then the better for all men, regardless of age or ethnicity. I expect the UK won’t be adopting this screening method for at least 5 years because of all the “usual” red tape plus demands for lengthy UK based clinical trial to validate it’s efficacy and worth to the NHS.

Tell us what you think

Leave a reply cancel reply.

Your email address will not be published. Required fields are marked *

Save my name, email, and website in this browser for the next time I comment.

Read our comment policy .

Highlighted content

34,000 cancer deaths could be prevented if we make this election a turning point for cancer, thousands of nhs patients to access trials of personalised cancer 'vaccines', sarah harding's legacy: finding women who may have higher breast cancer risks, asco 2024: data on weight loss drugs and a melanoma vaccine, a test that can predict breast cancer relapse, and more, one to one with rhian: volunteering special 2 - that cancer conversation podcast, general election 2024: what does this mean for the tobacco and vapes bill, skin cancer cases reach all-time high, an animal's guide to staying safe in the sun, related topics.

  • Cancer Research UK-funded research
  • Early detection
  • Diagnosing cancer
  • The Institute of Cancer Research
  • Cancer in the news

Gene Therapy in Both Ears Restores Hearing to Babies Born Deaf

HealthDay June 5, 2024

Gene-Targeted Immunotherapy Boosts Survival for Some Colon Cancer Patients

HealthDay June 3, 2024

Scientists Spot Gene Behind Form of Intellectual Disability Affecting Thousands Worldwide

Early hints at a gene therapy cure for herpes.

HealthDay May 15, 2024

Baby Girl Born Deaf Gains Hearing After Gene Therapy

HealthDay May 10, 2024

Gene Discovery Points to a New Form of Alzheimer's

HealthDay May 7, 2024

Gene Therapy Improves Vision in People With Inherited Blindness

HealthDay May 6, 2024

Living Healthy Counters Effects of 'Life-Shortening' Genes

HealthDay April 30, 2024

Scientists Discover Cause of Rare Movement Disorder

HealthDay April 29, 2024

Patient Gets First-Ever Pig Kidney Transplant Plus Heart Pump

HealthDay April 24, 2024

America 2024

latest genetics research

U.S. News Live

latest genetics research

latest genetics research

Research provides genetic insights into the chrysanthemum's architectural elegance

C hrysanthemums, admired globally for their intricate capitulum structures, have posed a genetic puzzle for scientists and horticulturists. The genetic mechanisms that dictate their development have been largely unknown, limiting the ability to selectively breed chrysanthemums for enhanced traits. This gap in knowledge has underscored the necessity for a focused genetic investigation to facilitate the cultivation of chrysanthemums with desired characteristics.

A collaborative effort by the Beijing Forestry University's School of Landscape Architecture has yielded a study on the genetic architecture of chrysanthemum inflorescence. Their findings, published in Horticulture Research on February 22, 2024, detail the role of the cla-miR164-NO APICAL MERISTEM (ClNAM) gene in shaping the flower's complex structure, marking a step forward in floral genetic research.

The study offers a detailed examination of the ClNAM gene's regulatory influence on the inflorescence of Chrysanthemum lavandulifolium. A key finding was the gene's central role within a broader genetic regulatory network. Overexpression of ClNAM was shown to hasten the initiation of floral primordium, leading to changes in receptacle morphology and a reduction in floret count.

This manipulation also resulted in a decrease in ray floret corolla length and an increase in disk floret elongation, highlighting ClNAM 's impact on floral organ development. Additionally, the study revealed that ClNAM is targeted by the microRNA cla-miR164, indicating a complex post-transcriptional regulation mechanism.

These discoveries shed light on the genetic pathways that govern the chrysanthemum's inflorescence, presenting opportunities for precision breeding and genetic enhancement.

Dr. Silan Dai, the senior author and an expert in plant molecular genetics, says, "Our research harnesses the power of genetic inquiry to demystify and manipulate plant development. The ClNAM gene's regulatory network is a monumental discovery and a versatile tool for pioneering breeding programs designed to amplify the chrysanthemum's innate splendor and variety."

The study's findings are set to transform the horticultural industry, providing a genetic roadmap for developing chrysanthemum varieties with tailored inflorescence features. This breakthrough may enable the cultivation of plants for specific uses, from decorative florals to landscape design.

Additionally, understanding ClNAM 's regulatory role could extend to other plant species, offering broader applications in agriculture and deepening our knowledge of plant development for conservation and scientific advancement.

More information: Junzhuo Li et al, cla-miR164-NO APICAL MERISTEM (ClNAM) regulates the inflorescence architecture development of Chrysanthemum lavandulifolium, Horticulture Research (2024). DOI: 10.1093/hr/uhae039

Provided by TranSpread

Overexpression of ClNAM caused: (A–B) a significant increase in the number of capitulum on top of plants. (C) developmental delayed ray floret corolla and elongated disk corolla. (D) changes in the receptacle morphology of the C. lavandulifolium and a decrease in the area of receptacle. (E) shortened ray floret corolla and elongated disk corolla. (F–G) change in the length of both types of florets with the position. (H–I) change in the number of both types of florets with the position height. Credit: Horticulture Research (2024). DOI: 10.1093/hr/uhae039

Jill Barnholtz-Sloan Named CBIIT’s Acting Director

June 6, 2024 , by NCI CBIIT Staff

latest genetics research

Dr. Jill Barnholtz-Sloan  is now the acting director for  NCI Center for Biomedical Informatics and Information Technology  (CBIIT). She takes this role after Dr. Tony Kerlavage, the previous director for seven years, retired on May 31, 2024.

Dr. Barnholtz-Sloan, who has been with CBIIT for the past three years, shows an extensive involvement in and a profound understanding of data. She has strong relationships with CBIIT and NCI staff, and possesses clear knowledge of NCI research programs. As both an active researcher and administrator, she has insight into how to translate data into real-world solutions to help diagnose, prevent, and treat cancer.

Along with her new acting director responsibilities, Jill will maintain her position as associate director for the  Informatics and Data Science Program  within CBIIT. In this capacity, she spearheads initiatives to formulate informatics and  data science  strategies while supporting collaboration throughout NCI, NIH, and beyond. She’ll also continue her role as senior investigator in the  Division of Cancer Epidemiology and Genetics Trans-Divisional Research Program .

Dr. Barnholtz-Sloan is a sought-out expert in informatics and data science. She finds and addresses new ways for ensuring that CBIIT’s rich data tools are available to investigators globally.

“I am thrilled to have this opportunity to ensure CBIIT’s excellence in data science,  data sharing , and technology; I’ll continue to innovate in these areas that impact the cancer research community,” notes Dr. Barnholtz-Sloan.

  • Fellowships & Training
  • Linkage Newsletter
  • People in the News
  • Research Highlights

IMAGES

  1. Center for Human Genetics & Genomics Research

    latest genetics research

  2. Human genome: 1 million sequences will open the doors of genetics

    latest genetics research

  3. World Ranking of Top Genetics & Molecular Biology Scientists in 2022

    latest genetics research

  4. Overview of Genes, DNA, and Chromosomes

    latest genetics research

  5. Human Genetic Modification

    latest genetics research

  6. Advances in Genetics Research. Volume 21

    latest genetics research

VIDEO

  1. Your Genetics and Longevity: What You Can (and Can't) Control

  2. Complete Genetics

  3. Fundamentals of Genetics Bsc AG.1st semester

  4. Hannah Mitchison PCD Genetic Research 220619

  5. Fermented foods: the real deal?

  6. Writing The Future of Human Genetics with Twist Bioscience

COMMENTS

  1. Genetics

    Genetics is the branch of science concerned with genes, heredity, and variation in living organisms. ... Research now shows that mouse oocytes use an unusual chromatin signature to mark regulatory ...

  2. Genetics News -- ScienceDaily

    Genetics research. Read the latest news on plant and animal genetics from universities and research institutes around the world.

  3. Genetics

    A genetic parasite may explain why humans and other apes lack tails. Around 25 million years ago, a stretch of DNA inserted itself into an ancestral ape's genome, an event that might have taken ...

  4. Scientists develop a rapid gene-editing screen to find effects of

    Caption: Using a variant of CRISPR genome-editing known as prime editing, MIT researchers have developed a method to screen cancer-associated genetic mutations much more easily and quickly than any existing approach. This illustration, by Samuel Gould's brother Owen Gould, is an artistic interpretation of the research and the idea of "rewriting the genome," explains Samuel.

  5. Scientists Finish the Human Genome at Last

    They ended up with 99 scientists working directly on sequencing the human genome, and dozens more pitching in to make sense of the data. The researchers worked remotely through the pandemic ...

  6. Genetics

    S.M. Fullerton and K.B. BrothersN Engl J Med 2024;390:1349-1351. The widening scope and scale of genetic tests are posing new challenges, and the need to address these challenges is becoming ...

  7. The first gene-editing treatment: 10 Breakthrough Technologies 2024

    Now. The first gene-editing cure has arrived. Grateful patients are calling it "life changing.". It was only 11 years ago that scientists first developed the potent DNA-snipping technology ...

  8. Genes News -- ScienceDaily

    Genes and Genetics News. Read today's medical research in genetics including what can damage genes, what can protect them, and more.

  9. CRISPR Gene-Editing Breakthrough Opens Door To Treating Broad Array Of

    Other scientists who are not involved in the research agree. "This is a wonderful day for the future of gene-editing as a medicine," agree Fyodor Urnov , a professor of genetics at the University ...

  10. 275 million new genetic variants identified in NIH precision medicine

    Researchers have discovered more than 275 million previously unreported genetic variants, identified from data shared by nearly 250,000 participants of the National Institutes of Health's All of Us Research Program. Half of the genomic data are from participants of non-European genetic ancestry.

  11. Genetics News

    1. The latest news on genetics, genetic science, genetic research, genetic engineering, genetic studies and genes.

  12. Genetics News

    Genetics is the study of genes, heredity, and genetic variation in living organisms. Our Genetics category provides the latest news, research, and discoveries in this exciting field, including the latest breakthroughs in genetic engineering, gene therapy, and personalized medicine. From the study of genetic diseases to the genetics of complex ...

  13. Genetics study points to potential treatments for ...

    from research organizations. Genetics study points to potential treatments for restless leg syndrome Date: June 5, 2024 ... The team identified over 140 new genetic risk loci, increasing the ...

  14. Opinion

    The Long-Overlooked Molecule That Will Define a Generation of Science. Dr. Cech is a biochemist and the author of the forthcoming book "The Catalyst: RNA and the Quest to Unlock Life's Deepest ...

  15. Genetics

    Genetics coverage from Scientific American, featuring news and articles about advances in the field. ... New research examines the molecular machinery behind a beetle's strange biological cycle ...

  16. The road ahead in genetics and genomics

    In celebration of the 20th anniversary of Nature Reviews Genetics, we asked 12 leading researchers to reflect on the key challenges and opportunities faced by the field of genetics and genomics.

  17. Scientists identify a genetic disorder that may cause intellectual

    Scientists find a likely cause of many unexplained cases of intellectual disability: A genetic disorder. Many neurodevelopmental issues have no known cause. But new research points to mutations in ...

  18. People whose bodies age faster may benefit most from good heart health

    In participants whose bodies were genetically predisposed to aging faster, 39% of the association between LE8 scores and cardiovascular disease or death was due to the effects of cardiovascular health factors on DNA methylation. By comparison, it was 21% in participants with any epigenetic age. Similarly, DNA methylation determined 78% of the ...

  19. Recent developments in genetic/genomic medicine

    Research established that immunologically confirmed hypersensitivity reactions to abacavir only occurred in people with ... et al. (2018) Making new genetic diagnoses with old data: iterative reanalysis and reporting from genome-wide data in 1,133 families with developmental disorders. Genet. Med. 20, 1216-1223 10.1038/gim.2017.246 [PMC free ...

  20. Gene variants and breast cancer risk in Black women

    But despite the increased risks faced by women of African descent, most large-scale genetic studies of breast cancer to date have focused on women of European ancestry. To better understand their unique genetic risks, a research team led by Dr. Wei Zheng of Vanderbilt University analyzed genetic data from over 40,000 females of African descent.

  21. Current Clinical Studies

    Participants in clinical studies help current and future generations. Through these studies, researchers develop new diagnostic tests, more effective treatments, and better ways of managing diseases with genetic components. Participants in studies are actively involved in understanding their disorder and current research.

  22. At-home saliva test could help diagnose prostate cancer sooner

    A new saliva test for identifying men at high risk of prostate cancer could help find more cases of the disease earlier, when doctors have a better chance of treating it successfully. ... "Building on decades of research into the genetic markers of prostate cancer, our study shows that the theory does work in practice - we can identify men ...

  23. Genetics Research

    Genetics Research is a fully open access journal providing a key forum for original research on all aspects of human and animal genetics, reporting key findings on genomes, genes, mutations, developmental, evolutionary, and population genetics as well as ethical, legal and social aspects. ... Latest Articles More articles. Research Article. 30 ...

  24. Genetics

    Rare Genes Can Raise Odds for Obesity 6-Fold. HealthDay April 4, 2024. Load More. Read the latest articles and commentary on genetics at US News.

  25. Research provides genetic insights into the chrysanthemum's

    Their findings, published in Horticulture Research on February 22, 2024, detail the role of the cla-miR164-NO APICAL MERISTEM (ClNAM) gene in shaping the flower's complex structure, marking a step ...

  26. Jill Barnholtz-Sloan Named CBIIT's Acting Director

    Dr. Jill Barnholtz-Sloan is CBIIT's new acting director following Dr. Tony Kerlavage's retirement. In addition to this role, Dr. Barnholtz-Sloan maintains her responsibility as associate director for the Informatics and Data Science Program and senior investigator in the Division of Cancer Epidemiology and Genetics, Trans-Divisional Research Program.