research on complex analysis

Complex Analysis

Complex analysis is the study of complex numbers together with their derivatives , manipulation, and other properties. Complex analysis is an extremely powerful tool with an unexpectedly large number of practical applications to the solution of physical problems. Contour integration , for example, provides a method of computing difficult integrals by investigating the singularities of the function in regions of the complex plane near and between the limits of integration.

The key result in complex analysis is the Cauchy integral theorem , which is the reason that single-variable complex analysis has so many nice results. A single example of the unexpected power of complex analysis is Picard's great theorem , which states that an analytic function assumes every complex number , with possibly one exception, infinitely often in any neighborhood of an essential singularity !

A fundamental result of complex analysis is the Cauchy-Riemann equations , which give the conditions a function must satisfy in order for a complex generalization of the derivative , the so-called complex derivative , to exist. When the complex derivative is defined "everywhere," the function is said to be analytic .

Explore with Wolfram|Alpha

WolframAlpha

More things to try:

  • complex analysis
  • 5th hexagonal number
  • find the area between sinx and cosx from 0 to pi

Referenced on Wolfram|Alpha

Cite this as:.

Weisstein, Eric W. "Complex Analysis." From MathWorld --A Wolfram Web Resource. https://mathworld.wolfram.com/ComplexAnalysis.html

Subject classifications

  • Department of Mathematics
  • Research Groups

Complex Geometry and Complex Analysis

The members of the Complex Geometry and Complex Analysis group conduct research in the areas of  Complex Geometry and Several Complex Variables, Algebraic Geometry, Complex Analysis, Differential Geometry, Diophantine Geometry, and Number Theory etc. The group consists of  faculty members Gordon Heier, Shanyu Ji and Min Ru, as well as seven current graduate students. It runs the Seminar on Complex Geometry and Complex Analysis and regularly offers graduate and undergraduate courses on topics in the above areas.

Members of Complex Geometry and Complex Analysis group

Research Faculty

Graduate Students:

Current Graduate Students:

  • Alvarez, Angelynn (Advisor: Gordon Heier),
  • Chaturvedi, Ananya (Advisor: Gordon Heier)
  • Hussein, Saud (Advisor: Min Ru)
  • Liao, Hungzen (Advisors: Min Ru and Gordon Heier)
  • Mills, Charles (Advisor: Min Ru)
  • Park, Jungim (Advisor: Min Ru)
  • Ugur, Gul (Advisor: Min Ru)

Former Graduate Students (with Ph.D. degree):

  • 2003: Eunjeong Yi (Advisor: Min Ru). Thesis:  Nevanlinanna theory and iteration of rational maps. Current job: Associate Professor, Texas A & M University at Galveston.
  • 2006: Xu, Dekang (Advisor: S. Ji).  First job: CGGVeritas, Houston.
  • 2007: Liu, Y (Advisor: Min  Ru). First job: dGB-USA Earth Science.
  • 2008: Dulock, Matthew (Advisor: Min Ru). Uniqueness theorem for holomorphic curves into Abelian variety. First job: Post-doc at the Fields Institute. Current job: University of North Texas.
  • 2011: Sogome, Suraizou (Advisor: Min Ru). Thesis: Non-Intergrated Defect Relation for Meromorphic Maps of Complete Kahler Manifolds Encountering Divisors.  First job: HHC, Northwest College.
  • 2014: Andrews, Jared (Advisor Shanyu Ji). Thesis: A New Gap Theorem Result For Proper Holomorphic Mappings Between Complex Balls. First job: Data Scientist at Havas Edge, San Diego.
  • 2014: Lee, Brandon (Advisor: Shanyu Ji). Thesis: Pseudoconformal Curvature and the Embeddability of a CR Hypersurface. First job: Clements high school, Sugar Land.

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
  • My Account Login
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Perspective
  • Open access
  • Published: 10 May 2024

Genetics and Genomics

Graph machine learning for integrated multi-omics analysis

  • Nektarios A. Valous   ORCID: orcid.org/0000-0002-4014-2404 1 , 2 ,
  • Ferdinand Popp 1 , 3 ,
  • Inka Zörnig 2 , 4 ,
  • Dirk Jäger 1 , 2 , 4 &
  • Pornpimol Charoentong 2 , 4  

British Journal of Cancer ( 2024 ) Cite this article

97 Accesses

1 Altmetric

Metrics details

  • Computational science
  • Data integration

Multi-omics experiments at bulk or single-cell resolution facilitate the discovery of hypothesis-generating biomarkers for predicting response to therapy, as well as aid in uncovering mechanistic insights into cellular and microenvironmental processes. Many methods for data integration have been developed for the identification of key elements that explain or predict disease risk or other biological outcomes. The heterogeneous graph representation of multi-omics data provides an advantage for discerning patterns suitable for predictive/exploratory analysis, thus permitting the modeling of complex relationships. Graph-based approaches—including graph neural networks—potentially offer a reliable methodological toolset that can provide a tangible alternative to scientists and clinicians that seek ideas and implementation strategies in the integrated analysis of their omics sets for biomedical research. Graph-based workflows continue to push the limits of the technological envelope, and this perspective provides a focused literature review of research articles in which graph machine learning is utilized for integrated multi-omics data analyses, with several examples that demonstrate the effectiveness of graph-based approaches.

Similar content being viewed by others

research on complex analysis

scGPT: toward building a foundation model for single-cell multi-omics using generative AI

research on complex analysis

Genome-wide association studies

research on complex analysis

Best practices for single-cell analysis across modalities

Translational bioinformatics and data-driven biomedical research involving multi-omics profiling studies enable researchers to obtain comprehensive insights into key biological processes in health and disease. These studies are slowly becoming ubiquitous in biomedical research, and typically amalgamate genomics, epigenomics, transcriptomics, proteomics, metabolomics, metagenomics, and other modalities. Single-omic studies provide data and information pertinent to different functional and molecular layers. Single-omic approaches may lack the precision required to establish robust associations between molecular-level changes and phenotypic traits. Many diseases, including cancer, are the result of multistage processes and events that incorporate multiscale information from the genome to the proteome, consequently interactions and synergistic effects are much better explored through multi-omics analysis. Effectively, multi-omics experiments at bulk or single-cell resolution facilitate the discovery of hypothesis-generating biomarkers for predicting response to therapy, as well as aid in uncovering mechanistic insights into cellular and microenvironmental processes.

The primary motivation behind integrated data analysis is to identify key factors that explain or predict disease risk or other biological outcomes [ 1 ]. Integrated data derived from different sources are used for computational analysis through machine learning or biostatistics methods and eventually may lead to more solid results and output [ 2 ]. Many methods for data integration have been developed (concatenation-based, transformation-based, model-based, intermediate, hierarchical), all with strengths and weaknesses, and naturally, no single analysis approach will be optimal for all studies [ 1 , 2 , 3 ]. Furthermore, numerous integration strategies have been established involving graph-free workflows. For instance, these approaches—primarily based on the integration of genomics, epigenomics, transcriptomics, proteomics, and metabolomics data—have been utilized in, e.g., cancer research for the functional identification of driver genomic alterations, tumor classification, etc. [ 4 ]. In this paradigm, the multi-omics datasets are in a tabular format; rows represent samples and columns represent biological variables grouped by omics [ 3 ]. A simple concatenation of features across the omics (early integration) is likely to generate large matrices, outliers, and highly correlated variables [ 2 ]. A mixed integration strategy addresses the shortcomings of early integration by transforming independently each omics set into a simpler representation [ 3 ]. In intermediate integration, features are jointly integrated across the omics without prior omic-specific processing, with the advantage of processing features based on their redundancy or complementarity both within each omic and across the different omics [ 2 ]. Late integration is based on machine learning methods where a model is first trained for each omic to perform the prediction independently, and then the predictions achieved from each omic are combined via averaging or voting [ 2 ]. Machine learning models are commonly employed to analyze complex real-world data. In this context, unsupervised learning (e.g., dimensionality reduction) discovers patterns in multi-omics datasets without mapping input to output data [ 5 , 6 ]. For example, variational autoencoders construct meaningful latent representations of integrated data, in an unsupervised way, by learning a compressed representation of the data and additionally by learning the underlying distribution parameters of the input data [ 7 ]. On the other hand, supervised learning, given input data and output labels, finds a function that maps the input to the label information (phenotypes of interest) [ 6 ]. Namely, Koh et al. developed a supervised learning method for integrating multi-omic profiles over genome-scale biological networks, and extracted network signatures predictive of pre-specified phenotypic groups [ 8 ]. Furthermore, mixed workflow approaches can be utilized for, e.g., modeling patient survival by processing multi-omics data via a combination of autoencoder and supervised machine learning algorithms [ 9 ].

In principle, all approaches aim to provide solutions for enhancing performance in a learning task, while mitigating, as much as possible, an array of challenges pertinent to data and methods. Methodologies commonly engage with incomplete, sparse, high-dimensional data, and obtain optimized representations and/or fuse information from multiple modalities. In this setting, some methods may tend to focus on a subset of modalities that are most helpful during model training while ignoring modalities that could be informative for model implementation, and because different modalities may lead to intricate relational dependencies, modality fusion may not fully leverage multimodal datasets [ 10 ]. In contrast, graph machine learning can model such datasets by connecting different modalities in optimally defined (but more realistically in context-defined) graphs, and by building learning systems for a wide range of tasks [ 10 ]. In this perspective, the authors are discussing the current trend of integrated multi-omics data analysis using graph machine learning approaches in the context of data-driven biomedical research.

Graph modeling and machine learning

It is widely acknowledged that machine learning and especially deep learning systems have been very successful in analyzing complex biomedical datasets from a variety of domains and sources. Commonly, these datasets are defined in the Euclidean domain (modeled in an n -dimensional linear space, e.g., grid data) with existing deep learning methodologies developed to capture hidden patterns in such data, e.g., for large-scale image classification. Deep learning approaches using multi-omics datasets typically transform the high-dimensional features into high-level semantic embeddings, then learn a unified representation from the embeddings, and finally apply the learned representation for downstream tasks [ 11 ]. Hence, conventional deep learning approaches are rather limited in modeling the interrelationships/interactions among different omics, coupled with the incapacity to incorporate graph-based prior knowledge (e.g., protein-protein interaction networks) as input.

A different strategy for omics datasets would be to model them mathematically as graph-structured data, so that the relevant entities can be connected based on their intrinsic relationships, biological properties/significance, and empirical biomedical knowledge. All interactions within and across different omics sets form an interlinked graph (network) composed of vertices (nodes or entities) and edges (links or relationships). Effectively, omics information is no longer embodied as elements in data tables but rather as entities that are linked to one another by edges with properties/attributes that define the associations between the nodes. This heterogeneous graph representation of multi-omics (multiple types of nodes with diverse types of edges among them) provides an advantage for identifying patterns suitable for predictive or exploratory analysis, thus permitting the modeling of complex relationships and interactions.

Geometric deep learning encompasses emerging techniques that attempt to generalize structured deep neural models to graphs and manifolds [ 12 ]. Especially, graph machine learning methods have been developed to process data represented in the form of graphs, i.e., with an underlying structure that is a non-Euclidean space [ 13 ]. Graph neural networks (convolutional, attentional, message-passing) are performing inference over data embedded in a graph structure, consequently allowing for the learning process to consider the explicit relations of the data within and across different omics. Over the past few years, graph neural networks have become powerful and functional tools for machine learning tasks in the graph domain; this progress owes to advances in expressive power, model flexibility, and training algorithms [ 14 ]. On a practical note, there are several software libraries and tools that are regularly utilized for graph machine learning tasks; some of the more popular ones include: PyTorch Geometric (PyG) [ 15 ], Deep Graph Library (DGL) [ 16 ], Graph Nets [ 17 ], and Spektral [ 18 ]. Table  1 presents a broad categorization of graph machine learning techniques for multi-omics data. The table was adapted—from the general grouping of graph machine learning methods found in Xia et al. [ 19 ]—to reflect the multi-omics setting.

Pertaining to the workings of a graph learning approach, a brief outline of the general framework of graph neural networks for node classification (supervised) is presented [ 20 ]. Let \(G=(V,E)\) denote a graph where \(V\) is the set of vertices or nodes and \(E\) the set of the edges connecting the nodes [ 20 ]. Then, \(A\in {{\mathbb{R}}}^{N\times N}\) represents the adjacency matrix where \(N\) is the total number of nodes and \(X\in {{\mathbb{R}}}^{N\times C}\) represents the node attribute matrix ( \(C\) is the number of features for each node) [ 20 ]. The objective is to learn effective node representations (denoted by \(H\in {{\mathbb{R}}}^{N\times F}\) where \(F\) is the dimension of node representations) by combining the graph structure information and the node attributes which are further used for node classification [ 20 ]. The essential idea of graph neural networks is to iteratively update the node representations by combining the representations of their neighbors and their own representations [ 20 ]. Starting from the initial node representation, \({H}^{0}=X\) , in each layer there are two main functions: (1) AGGREGATE which aggregates information from the neighbors of each node, and (2) COMBINE which updates the node representations by combining the aggregated information from neighbors with the current node representations [ 20 ]. Therefore, the general framework of graph neural networks is defined by: Initialize: \({H}^{0}=X\) ; For \(k={{{{\mathrm{1,2}}}}},\ldots ,K\) ; \({a}_{v}^{k}={{{{{{{\rm{AGGREGATE}}}}}}}}^{k}\left\{{H}_{u}^{k-1}:u\in N\left(v\right)\right\}\) ; \({H}_{v}^{k}={{{{{{{\rm{COMBINE}}}}}}}}^{k}\left\{{H}_{u}^{k-1},{a}_{v}^{k}\right\}\) , with \(N\left(v\right)\) being the set of neighbors for the \(v\) -th node [ 20 ]. The node representations \({H}^{K}\) in the last layer can be treated as the final node representations [ 20 ]. The computed node representations can be utilized for downstream tasks, e.g., node classification in which the label of node \(v\) (denoted by \({\hat{y}}_{v}\) ) can be predicted through the Softmax function: \({\hat{y}}_{v}={{{{{{\rm{Softmax}}}}}}}\left(W{H}_{v}^{T}\right)\) where \({H}_{v}^{T}\) is the transpose of \({H}_{v}\) and \(W\in {{\mathbb{R}}}^{\left|{\mathfrak{L}}\right|\times F}\) with \(\left|{\mathfrak{L}}\right|\) being the number of labels in the output space [ 20 ]. Given a set of labeled nodes, the model can be trained by minimizing the loss function: \(O=\left(1/{n}_{l}\right){\sum }_{i=1}^{{n}_{l}}{{{{{{\rm{loss}}}}}}}\left(\hat{{y}_{i}},{y}_{i}\right)\) where \({y}_{i}\) is the ground truth label of node \(i\) , \({n}_{l}\) is the number of labeled nodes, and \({{{{{{\rm{loss}}}}}}}\left(\cdot ,\cdot \right)\) is a loss function such as cross-entropy [ 20 ]. The whole model can be optimized by minimizing the objective function \(O\) with backpropagation [ 20 ].

Integrative analysis methodologies

Multi-omics profiling technologies dive deeply into molecular landscapes and reveal multiple facets of complex research problems, e.g., shedding light on exciting novel aspects of cancer biology; these cutting-edge technologies produce large and intricate datasets, presenting researchers and clinicians with the considerable task of distilling complex information into clinical insights [ 21 ]. For instance, pan-cancer multi-omics analysis has revealed driver gene regulation via DNA methylation, offering insights into methylation-based stratification of cancer patients [ 22 ]. Accordingly, it is broadly acknowledged that there is a need for robust integrative analysis methodologies—for advancing precision medicine—that combine multiple data modalities effectively, hence taking into consideration the multilayered characteristics and interaction information of multi-omics datasets. Graph-based approaches—including graph neural networks—potentially offer a reliable methodological toolset that can provide a tangible alternative to scientists and clinicians that seek ideas and implementation strategies in the integrated analysis of their omics datasets for biomedical research. For example, graph convolutional networks can classify unlabeled nodes in a graph based on both their associated feature vectors as well as the network’s topology, making it possible to integrate graph-based data with feature vectors in a natural way [ 23 ]. Figure  1 shows a conceptual workflow for integrated multi-omics analysis using graph machine learning in the context of precision medicine, i.e., translating the output of these approaches into biomedical outcome.

figure 1

Multiple modalities ( a ) such as genomics (somatic mutations, copy number variants, rare variants, genomic rearrangements, etc.), epigenomics (DNA methylation, chromatin accessibility, histone modifications, etc.), transcriptomics (mRNA expression, non-coding RNAs, etc.), proteomics (abundances and post-translational modifications), metabolomics (amino acids, organic acids, sugars, lipids, nucleotides, drugs, steroids, etc.), metagenomics (microbial enrichment, phylogeny, evolutionary profiles, etc.) and others are modeled as graph-structured data [ 2 ] along with prior knowledge such as, e.g., protein-protein interaction (PPI) networks, gene regulatory networks (GRN), and metabolic networks (MN). Graph machine learning methods [ 13 ] ( b ) are developed/applied for unsupervised, semi-supervised, and supervised learning [ 5 , 6 , 24 ] at the node, edge, or graph level for integrated analysis within and across different omics sets; these methods may include graph autoencoders (AE, upper left part), graph variational autoencoders (VAE, upper right part), and graph convolutional neural networks (CNN, lower part). The diagrams in ( b ) are generic architectural representations of the aforementioned neural networks. The overall objective is to translate the output into biomedical outcome ( c ): perform classification (e.g., tumor grade and subtype), form groups (patient clustering; CL), predict patient survival (KM), and identify potential biomarker (BM) candidates. The augmented information below each conceptual step of the workflow provides complementary details that correspond to general descriptions or actions that may fit different approaches. [Attribution: DNA/chemical formula vectors were adapted from vecteezy.com].

A common approach when investigating graph-based methodologies is to model each omics dataset into a separate graph before analysis. Combining the separate graphs into a single homogeneous graph (through fusion) as the input of machine learning models allows for carrying out clustering, subtype discovery/classification, or survival prediction [ 3 ]. Furthermore, building a multilayered network with inter-layer connections (where each layer represents an omics set and interactions between omics sets are either inferred or retrieved from databases) allows for several methods to explore the network’s topology including shortest paths and random walks [ 3 ]. Graph embedding methods learn low-dimensional representations of nodes and their surroundings from each graph; the new graph-based features are then fed to machine learning models for prediction or classification [ 3 ]. This paradigm—graph representation learning—has emerged as a prominent machine-learning strategy for graphs, where the learned embeddings of graph elements are generated such that they capture the structure and semantics of the graph along with any downstream supervised task [ 24 ]. Graph representation learning includes methods for shallow graph embeddings that are utilized for node- and edge-property prediction, as well as graph neural networks that can generate representations for any graph element by capturing structure, attributes, and node metadata, thus utilized for node-, edge-, and graph-property prediction [ 24 ].

Graph-based integrated multi-omics analysis

Graph-based workflows—including graph machine learning pipelines—continue to push the limits of the technological envelope, facilitating new investigations by combining patient information and biomedical knowledge. Several research articles showcase the potential of graph-based methodologies for integrated multi-omics analysis, hence offering scientists and clinicians inspiration and hints for tackling their elaborate research problems.

Multi-omics integration with no network-based prior knowledge

Pai et al. presented a patient similarity graph-based approach for supervised patient classification using data (clinical; mRNA, miRNA, and protein expression; DNA methylation; and somatic copy number alteration) [ 25 ] from The Cancer Genome Atlas (TCGA) [ 26 ]. The authors demonstrated parity or superiority, comparing to other machine learning approaches (e.g., diagonal discriminant analysis, k -nearest neighbors, logistic regression, nearest centroid, partial least squares, random forests, and support vector machines) in predicting survival across four different tumor types, while visualizing the decision boundary in the context of patient similarity space thus making the results more interpretable [ 25 ]. As a further example, the authors compared their approach to another multi-omic patient classifier (e.g., DIABLO) showing that both tools provide complementary views of predictive multi-omic features that could be useful when applied in tandem [ 25 ]. A comparable approach showed that integrative modeling using genomics and electronic health record data has clinical utility [ 27 ]. Fang et al. showcased the effectiveness of a marginalized graph autoencoder for learning patient similarity feature representations followed by graph spectral clustering, in order to stratify non-small cell lung cancer patients into subgroups with distinct immunotherapy outcomes [ 27 ]. The authors explored differences in biological insight comparing their approach to the conventional log-rank test using clinico-genomic features, and indicated the potential of their method to inform insight on patient stratification as a complement to the traditional approach [ 27 ]. Wang et al. utilized labeled omics datasets (TCGA) and proposed a supervised multi-omics (mRNA and miRNA expression; and DNA methylation) integration approach based on deep multi-view learning (each omics data type as a particular view of the samples) [ 28 ]. The authors utilized graph convolutional networks for omics-specific learning, and a view-correlation discovery network to explore cross-omics correlations at the label space for effective multi-omics integration [ 28 ]. The authors compared the classification performance of their approach with other supervised multi-omics integration algorithms (e.g., k -nearest neighbors, support vector machines, lasso, random forests, gradient boosted trees, shallow and deep fully connected neural networks, adaptive group-regularized ridge regression, and two partial least squares discriminant analysis variants); their method outperformed the other methods in most classification tasks. Further ablation studies showed that their approach outperformed its variations in various classification tasks, and comparisons using their method on different omics sets showed that models trained with multi-omics data achieved better performance compared to single-omics models [ 28 ]. Overall, the authors successfully demonstrated their approach on tumor grade classification in low-grade glioma, kidney cancer type classification, and breast invasive carcinoma subtype classification, as well as biomarker identification related to breast cancer [ 28 ]. Likewise, Li et al. also developed a multi-omics integration model based on graph convolutional networks using copy number variation (exome-seq), transcriptomics (RNA-seq), proteomics (reverse-phase protein array), and clinical data from patients (TCGA) for cancer subtype analysis [ 29 ]. The authors applied a multi-modal autoencoder model to extract features and employed a similarity network fusion model to construct a patient similarity network; they compared their autoencoder with conventional methods such as principal component analysis, factor analysis, independent component analysis, and singular value decomposition [ 29 ]. Next, the authors utilized a graph convolutional network to integrate these two types of heterogeneous features and train the subtype classification model; they compared their graph convolutional network with methods such as decision trees, k -nearest neighbors, Gaussian naïve Bayes, random forests, support vector machines, a deep neural network with four layers, Grassmann clustering, and high-order path elucidated similarity [ 29 ]. Their method performed well for heterogeneous data integration, while addressing the issue of clinical interpretability [ 29 ]. Focusing on the interpretability aspect of graph convolutional neural networks explaining individualized predictions, Chereda et al. generated explanations in the form of relevant subgraphs for each data point, consequently providing interpretable molecular sub-networks that were individual for each patient [ 30 ].

Multi-omics integration with network-based prior knowledge

In the context of integrating different multi-omics data and network-based prior knowledge [ 31 ], Kim et al. presented a graph-based semi-supervised framework for integrating multi-omics TCGA data (mRNA and miRNA expression; DNA methylation; and somatic copy number alteration) and genomic knowledge (pathway, gene ontology, motif, and chromosomal position gene sets), in an intermediate fashion, to predict outcomes according to survival, stage, and grade [ 32 ]. Data-driven graphs were generated from the multi-omics data and knowledge-driven graphs were generated from the genomic knowledge sets [ 32 ]. Their results suggested that the use of genomic knowledge improved the predictive power in explaining cancer phenotypes due to the synergies between genomic processes in the pathways involved in cancer [ 32 ]. The strengths of graph-based integration include its high computational efficiency (due to its sparseness properties) combined with an accuracy that is comparable to those of other methods such as kernel-based integration [ 32 ]. In a more recent approach [ 33 ], Ma and Zhang employed a multi-view factorization autoencoder to integrate multi-omics data (mRNA, miRNA, and protein expression; and DNA methylation) and protein-protein interaction (PPI) networks (STRING database [ 34 ]), learning feature and patient embeddings simultaneously. Their model can be used for unsupervised learning, but with available labeled data then supervised learning is possible by modifying the objective function [ 33 ]. The authors performed experiments on TCGA data for predicting the progression-free interval, and compared their model with other methods such as support vector machines, decision trees, naïve Bayes, random forests, AdaBoost, a variational autoencoder, and an adversarial autoencoder [ 33 ]. Inherently, the authors demonstrated that multi-omics data significantly outperformed single-omics, and additionally they showed that incorporating domain knowledge (e.g., biological interaction networks) in their model improves its generalizability and reduces the risk of overfitting [ 33 ]. In an analogous work, Schulte-Sasse et al. developed an interpretable graph deep learning approach to predict cancer genes from large datasets (pan-cancer data from the TCGA) by combining multi-omics data (mutations, copy number changes, DNA methylation, and mRNA expression) together with protein-protein interaction networks [ 23 ]. Identification of cancer genes plays a crucial role in the development of precision oncology and cancer therapeutics [ 23 ]. Furthermore, interpretability is valuable for assessing the molecular origin of a gene to be associated with cancer, detecting potential artifacts, and increasing trust in the modeling approach [ 23 ]. Their methodology used multi-dimensional multi-omics node features as well as topological features of the protein-protein interaction network in the learning process [ 23 ]. The authors compared their approach with methods grouped into different categories: omics only (methods that use only omics features for training, e.g., random forests), network only (methods that use only the PPI network, e.g., DeepWalk with support vector machines, graph convolutional network, and PageRank), network and omics (methods that use both data types, e.g., DeepWalk with random forest features, and HotNet2 diffusion), and cancer specific (methods specifically tailored to the prediction of cancer genes, e.g., MutSigCV, and 20/20+) [ 23 ]. The authors successfully recognized highly mutated cancer genes and genes harboring other kinds of alterations (aberrant DNA methylation, differential expression), consistently outperforming previous methods [ 23 ].

Single-cell multi-omics integration

Single-cell multi-omics permits the quantification of multiple modalities for fully capturing the perplexity of complex molecular mechanisms and cellular heterogeneity [ 35 ]. Current methods for integrating single-cell multi-omics data typically consider the cells relationship between the reference and query datasets but ignore the relationship among cells within each dataset [ 36 ]. In addition, multiple datasets of the same or different omics often have unpaired cells (due to single-cell sequencing techniques being still cell destructive) [ 36 ]. In this setting, Cao and Gao developed a method for triple-omics integration, integrative regulatory inference, and multi-omics human cell atlas construction over millions of cells [ 37 ]. The authors utilized public datasets (transcriptome through single-cell RNA sequencing; chromatin accessibility through single-cell ATAC sequencing; and DNA methylation through single-nucleus methylome sequencing and single-cell combinatorial indexing for methylation analysis assay), and they systematically benchmarked their approach with multiple popular unpaired multi-omics integration methods, e.g., Online iNMF, LIGER, Harmony, bindSC, Seurat, UnionCom, Pamona, and MMD-MA [ 37 ]. By combining omics-specific autoencoders with graph-based coupling and adversarial alignment, the authors presented a modular framework (graph-linked unified embedding) for integrating unpaired heterogeneous single-cell multi-omics data and inferring regulatory interactions simultaneously; benchmarks showed that their approach was robust, scalable, and extendable [ 37 ]. Gao et al. presented a model, using public datasets, for integrating single-cell multi-omics data (transcriptome through single-cell RNA sequencing; chromatin accessibility through single-cell ATAC sequencing; and protein expression through CITE-seq–cellular indexing of transcriptomes and epitopes by sequencing) based on graph convolutional networks [ 36 ]. The authors compared their approach with four integration algorithms, e.g., Seurat, LIGER, GLUER, and Pamona [ 36 ]. Their results, by applying the method on six datasets, showed that data can be integrated from multiple single-cell sequencing technologies, species, or different omics, outperforming other methods [ 36 ]. Ma et al. developed a heterogeneous graph transformer model, using public datasets, for cell-type-specific biological network inference from single-cell multi-omics data (modalities: single-cell RNA sequencing, CITE-seq, and single-cell ATAC sequencing) [ 35 ]; their model was hypothesis-free and did not rely on the constraints of gene co-expressions. The authors compared their approach to other tools such as the graph-based method of Cao and Gao [ 37 ], Seurat, MOFA+, Harmony, and TotalVI [ 35 ]. For each benchmarking tool, grid-search tests were applied to a combination of parameters such as the number of dimensions for cell clustering and clustering resolution [ 35 ]. Their approach learnt relations among cells and genes within both local and global contexts, and performed better than existing tools in cell clustering and biological network construction [ 35 ].

Challenges and opportunities

Graph-based multi-omics data integration may enable the formation of context-relevant networks that can capture the relations and interactions between different entities, e.g., genes, proteins, metabolites, etc., hence potentially offering a systems-level understanding of cellular and microenvironmental processes. Integration may aid in comprehending the function of genes and proteins more thoroughly (functional setting) and as a result deliver useful insights into biological processes. There might be possibilities in unraveling the mechanisms underlying diseases by studying the relationships between different biological components. In this context, subtyping diseases based on multi-omics profiles may offer perspectives into etiology and progression. Integration may allow for a more personalized approach to biomedicine by taking into account individual variations in omics profiles, as well as contributing to the identification of predictive biomarker candidates pertinent to the effect of a therapeutic intervention, thus aiding in the optimization of targeted therapies. On the other hand, integrating multi-omics data involves the coalescence of information from different molecular levels and this may pose challenges. In general, assembling omics data together into a more complete story is challenging mainly due to the diversity in dataset size, the patterns of missing data and noise across different data types, and the correspondences among measurements from different technologies [ 1 ]. More specifically, challenges may include: an imbalance at the class or feature level, missing values during data acquisition resulting in datasets with partial information, a larger number of features compared to a smaller number of patients, data with different distributions or types due to utilizing different technologies, and noise manifested as mislabeled samples.

Graph machine learning may offer a robust framework for integrating and analyzing multi-omics data by: (1) incorporating omics sets into a unified model, (2) scaling relatively well (up to a limit) in relation to growing data complexity, (3) providing more interpretable model predictions, (4) taking into account the complex interrelationships among the different molecular entities, (5) allowing for prior network-based knowledge integration, (6) leveraging the topology of the graph thus inferring potential associations that may not be apparent with conventional methods, (7) handling data heterogeneity related to scale and distribution quite well, (8) learning embeddings that describe the structure and relationships of the data, and (9) analyzing patterns of connectivity among multi-omics sets. The challenges in utilizing graph machine learning models on omics sets need to be acknowledged as well when developing such algorithms for analyzing biological data. A common challenge is the effort and domain expertise required for the construction of the graph which needs to be adapted to the problem at hand, but also relevant so that it reflects the inherent biological relationships. Data related problems such as increased heterogeneity (formats and scales), noise, and incomplete information can complexify the development of graph learning models. Often, the size of multi-omics datasets can be very large thus developing scalable models can be quite challenging as well. In this context, the large omics sets can make model training computationally expensive, ergo efficiency is paramount given that computational resources are finite. In this regard, Table  2 shows the general advantages and disadvantages of graph neural networks.

The way forward

Integrated multi-omics analysis possesses considerable promise for resolving the inherent complexities of biological systems. New toolsets are supporting the research community [ 38 ] to, e.g., represent biomedical knowledge in a user-friendly manner by building task-specific knowledge graphs that facilitate the navigation and analysis of complex information [ 39 ]. Further research in the field of graph machine learning for integrated multi-omics analysis may assist in unraveling the intricate molecular interactions across diverse biological systems, which can pave the way for a more comprehensive understanding of disease and personalized therapeutic interventions. An increasingly popular and powerful self-supervised learning approach, for alleviating the reliance on labeled data, is contrastive learning [ 40 ]. This technique aims to learn salient features using raw input as the learning signal and usually leverages multiple positive and negative pairs of input samples in one batch, while substantial data augmentation is normally required for learning good and generalizable embedding features [ 40 ]. Extending contrastive learning to graph-structured data may improve performance for downstream analysis by, e.g., utilizing it for the pre-training of multi-omic graphical models. Furthermore, the spatial context in biological studies has profound biomedical/clinical relevance and implications. For instance, Hu et al. presented a graph convolutional network approach that integrated gene expression, spatial location, and histology to model the spatial dependency of gene expression for the identification of spatial domains and domain enriched spatially variable genes [ 41 ]. Going further, in order to develop a basic understanding of the molecular hierarchy from genome to phenome in individual cells, single-cell and spatial multi-omics methodologies (multimodal omics) are required [ 42 ]. In this paradigm, by leveraging the spatial information linking a cellular state to its respective micro- and macro-environments, through the use of graph neural networks, more fine-grained multimodal representations of cellular state should be obtainable [ 42 ].

Conclusions

This perspective is focused on highlighting the significance of data-driven biomedical research, particularly within the context of integrating multiple omics. Integrated multi-omics analyses are crucial for exploring complex diseases, e.g., cancer, where multiple factors contribute to the disease’s development. Here, the use of machine learning is particularly emphasized for integrating and analyzing multi-omics datasets. Graph machine learning on heterogeneous omics sets has proven quite powerful as evidenced by previously published research in precision medicine, cancer biology, and other biomedical applications. Essentially, computational graph-based frameworks for bulk and single-cell integrated multi-omics analysis have indicated their capacity to clarify complex interrelationships and derive valuable insights from highly connected data. The different models presented have demonstrated that the enhanced capacity for analyzing omics interactions and the data integration with network-based prior knowledge are major advantages of graph-based approaches, with additional improvements in model generalizability. Certainly, there are issues on the data level as well as on the methodological level making integration a complicated endeavor when both are coupled for analyzing and in the end understanding complex biological systems. Nevertheless, factoring in the opportunities as well as the challenges of graph machine learning approaches on multi-omics data ensures for more sophisticated, adaptable, and refined models. These approaches will continue to provide demonstrable benefits to scientists and clinicians in terms of a more coherent and quantitative understanding of cell biology, but also more practically for improving the prediction of clinical outcome as well as assisting in the discovery of potential disease-related biomarker candidates.

Ritchie MD, Holzinger ER, Li R, Pendergrass SA, Kim D. Methods of integrating data to uncover genotype–phenotype interactions. Nat Rev Genet. 2015;16:85–97.

Article   CAS   PubMed   Google Scholar  

Chicco D, Cumbo F, Angione C. Ten quick tips for avoiding pitfalls in multiomics data integration analyses. PLoS Comput Biol. 2023;19:e1011224.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Picard M, Scott-Boyer M-P, Bodein A, Périn O, Droit A. Integration strategies of multi-omics data for machine learning analysis. Comput Struct Biotechnol J. 2021;19:3735–46.

Menyhárt O, Győrffy B. Multi-omics approaches in cancer research with applications in tumor subtyping, prognosis, and diagnosis. Comput Struct Biotechnol J. 2021;19:949–60.

Article   PubMed   PubMed Central   Google Scholar  

Vahabi N, Michailidis G. Unsupervised multi-omics data integration methods: a comprehensive review. Front Genet. 2022;13:854752.

Cai Z, Poulos RC, Liu J, Zhong Q. Machine learning for multi-omics data integration in cancer. iScience. 2022;25:103798.

Simidjievski N, Bodnar C, Tariq I, Scherer P, Terre HA, Shams Z, et al. Variational autoencoders for cancer data integration: design principles and computational practice. Front Genet. 2019;10:1205.

Koh HWL, Fermin D, Vogel C, Choi KP, Ewing RM, Choi H. iOmicsPASS: network-based integration of multiomics data for predictive subnetwork discovery. npj Syst Biol Appl. 2019;5:22.

Poirion OB, Jing Z, Chaudhary K, Huang S, Garmire LX. DeepProg: an ensemble of deep-learning and machine-learning models for prognosis prediction using multi-omics data. Genome Med. 2021;13:112.

Ektefaie Y, Dasoulas G, Noori A, Farhat M, Zitnik M. Multimodal learning with graphs. Nat Mach Intell. 2023;5:340–50.

Xiao S, Lin H, Wang C, Wang S, Rajapakse JC. Graph neural networks with multiple prior knowledge for multi-omics data analysis. IEEE J Biomed Health Inf. 2023;27:4591–600.

Article   Google Scholar  

Bronstein MM, Bruna J, LeCun Y, Szlam A, Vandergheynst P. Geometric deep learning: going beyond Euclidean data. IEEE Signal Process Mag. 2017;34:18–42.

Wu Z, Pan S, Chen F, Long G, Zhang C, Yu PS. A comprehensive survey on graph neural networks. IEEE Trans Neural Netw Learn Syst. 2021;32:4–24.

Article   PubMed   Google Scholar  

Zhou J, Cui G, Hu S, Zhang Z, Yang C, Liu Z, et al. Graph neural networks: a review of methods and applications. AI Open. 2020;1:57–81.

Fey M, Lenssen JE. Fast graph representation learning with PyTorch Geometric. arXiv. 2019. https://doi.org/10.48550/arXiv.1903.02428 .

Wang M, Zheng D, Ye Z, Gan Q, Li M, Song X, et al. Deep Graph Library: a graph-centric, highly-performant package for graph neural networks. arXiv. 2020. https://doi.org/10.48550/arXiv.1909.01315 .

Battaglia PW, Hamrick JB, Bapst V, Sanchez-Gonzalez A, Zambaldi V, Malinowski M, et al. Relational inductive biases, deep learning, and graph networks. arXiv. 2018. https://doi.org/10.48550/arXiv.1806.01261 .

Grattarola D, Alippi C. Graph neural networks in TensorFlow and Keras with Spektral. arXiv. 2020. https://doi.org/10.48550/arXiv.2006.12138 .

Xia F, Sun K, Yu S, Aziz A, Wan L, Pan S, et al. Graph learning: a survey. IEEE Trans Artif Intell. 2021;2:109–27.

Tang J, Liao R. Graph neural networks for node classification. In: Wu L, Cui P, Pei J, Zhao L, editors. Graph neural networks: foundations, frontiers, and applications. Singapore: Springer Nature; 2022. pp. 41–61.

Akhoundova D, Rubin MA. Clinical application of advanced multi-omics tumor profiling: shaping precision oncology of the future. Cancer Cell. 2022;40:920–38.

Liang W-W, Lu RJ-H, Jayasinghe RG, Foltz SM, Porta-Pardo E, Geffen Y, et al. Integrative multi-omic cancer profiling reveals DNA methylation patterns associated with therapeutic vulnerability and cell-of-origin. Cancer Cell. 2023;41:1567–85.e7.

Schulte-Sasse R, Budach S, Hnisz D, Marsico A. Integration of multiomics data with graph convolutional networks to identify new cancer genes and their associated molecular mechanisms. Nat Mach Intell. 2021;3:513–26.

Li MM, Huang K, Zitnik M. Graph representation learning in biomedicine and healthcare. Nat Biomed Eng. 2022;6:1353–69.

Pai S, Hui S, Isserlin R, Shah MA, Kaka H, Bader GD. netDx: interpretable patient classification using integrated patient similarity networks. Mol Syst Biol. 2019;15:e8497.

The Cancer Genome Atlas Research Network, Weinstein JN, Collisson EA, Mills GB, Mills Shaw KR, Ozenberger BA, et al. The cancer genome atlas pan-cancer analysis project. Nat Genet. 2013;45:1113–20.

Fang C, Xu D, Su J, Dry JR, Linghu B. DeePaN: deep patient graph convolutional network integrating clinico-genomic evidence to stratify lung cancers for immunotherapy. npj Digital Med. 2021;4:14.

Wang T, Shao W, Huang Z, Tang H, Zhang J, Ding Z, et al. MOGONET integrates multi-omics data using graph convolutional networks allowing patient classification and biomarker identification. Nat Commun. 2021;12:3445.

Li X, Ma J, Leng L, Han M, Li M, He F, et al. MoGCN: a multi-omics integration method based on graph convolutional network for cancer subtype analysis. Front Genet. 2022;13:806842.

Chereda H, Bleckmann A, Menck K, Perera-Bel J, Stegmaier P, Auer P, et al. Explaining decisions of graph convolutional neural networks: patient specific molecular subnetworks responsible for metastasis prediction in breast cancer. Genome Med. 2021;13:42.

Zhu J, Oh JH, Simhal AK, Elkin R, Norton L, Deasy JO, et al. Geometric graph neural networks on multi-omics data to predict cancer survival outcomes. Comput Biol Med. 2023;163:107117.

Kim D, Joung J-G, Sohn K-A, Shin H, Park YR, Ritchie MD, et al. Knowledge boosting: a graph-based integration approach with multi-omics data and genomic knowledge for cancer clinical outcome prediction. J Am Med Inf Assoc. 2015;22:109–20.

Ma T, Zhang A. Integrate multi-omics data with biological interaction networks using multi-view factorization autoencoder (MAE). BMC Genom. 2019;20:944.

Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49:D605–12.

Ma A, Wang X, Li J, Wang C, Xiao T, Liu Y, et al. Single-cell biological network inference using a heterogeneous graph transformer. Nat Commun. 2023;14:964.

Gao HG, Zhang B, Liu L, Li S, Gao X, Yu B. A universal framework for single-cell multi-omics data integration with graph convolutional networks. Brief Bioinforma. 2023;24:bbad081.

Cao ZJ, Gao G. Multi-omics single-cell data integration and regulatory inference with graph-linked embedding. Nat Biotechnol. 2022;40:1458–66.

Santos A, Colaço AR, Nielsen AB, Niu L, Strauss M, Geyer PE, et al. A knowledge graph to interpret clinical proteomics data. Nat Biotechnol. 2022;40:692–702.

Lobentanzer S, Aloy P, Baumbach J, Bohar B, Carey VJ, Charoentong P, et al. Democratizing knowledge representation with BioCypher. Nat Biotechnol. 2023;41:1056–9.

Loh C, Christensen T, Dangovski R, Kim S, Soljačić M. Surrogate- and invariance-boosted contrastive learning for data-scarce applications in science. Nat Commun. 2022;13:4223.

Hu J, Li X, Coleman K, Schroeder A, Ma N, Irwin DJ, et al. SpaGCN: integrating gene expression, spatial location and histology to identify spatial domains and spatially variable genes by graph convolutional network. Nat Methods. 2021;18:1342–51.

Vandereyken K, Sifrim A, Thienpont B, Voet T. Methods and applications for single-cell and spatial multi-omics. Nat Rev Genet. 2023;24:494–515.

Kim SY, Choe EK, Shivakumar M, Kim D, Sohn K-A. Multi-layered network-based pathway activity inference using directed random walks: application to predicting clinical outcomes in urologic cancer. Bioinformatics. 2021;37:2405–13.

Gao Y-L, Hou M-X, Liu J-X, Kong X-Z. An integrated graph regularized non-negative matrix factorization model for gene co-expression network analysis. IEEE Access. 2019;7:126594–602.

Gao M, Liu S, Qi Y, Guo X, Shang X. GAE-LGA: integration of multi-omics data with graph autoencoders to identify lncRNA–PCG associations. Brief Bioinforma. 2022;23:bbac452.

Kesimoglu ZN, Bozdag S. SUPREME: multiomics data integration using graph convolutional networks. NAR Genom Bioinform. 2023;5:lqad063.

Zhao W, Gu X, Chen S, Wu J, Zhou Z. MODIG: integrating multi-omics and multi-dimensional gene network for cancer driver gene identification based on graph attention network model. Bioinformatics. 2022;38:4901–7.

Jing X, Zhou Y, Shi M. Dynamic graph neural network learning for temporal omics data prediction. IEEE Access. 2022;10:116241–52.

Download references

Open Access funding enabled and organized by Projekt DEAL.

Author information

Authors and affiliations.

Applied Tumor Immunity Clinical Cooperation Unit, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany

Nektarios A. Valous, Ferdinand Popp & Dirk Jäger

Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany

Nektarios A. Valous, Inka Zörnig, Dirk Jäger & Pornpimol Charoentong

Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany

Ferdinand Popp

Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany

Inka Zörnig, Dirk Jäger & Pornpimol Charoentong

You can also search for this author in PubMed   Google Scholar

Contributions

Conceptualization and writing—original draft: NAV. Writing—review & editing: NAV, FP, IZ, DJ, and PC. Funding acquisition: IZ and DJ. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Nektarios A. Valous .

Ethics declarations

Competing interests.

The authors declare no competing interests.

Additional information

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ .

Reprints and permissions

About this article

Cite this article.

Valous, N.A., Popp, F., Zörnig, I. et al. Graph machine learning for integrated multi-omics analysis. Br J Cancer (2024). https://doi.org/10.1038/s41416-024-02706-7

Download citation

Received : 15 November 2023

Revised : 25 April 2024

Accepted : 26 April 2024

Published : 10 May 2024

DOI : https://doi.org/10.1038/s41416-024-02706-7

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

Quick links

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

research on complex analysis

Scientists document remarkable sperm whale 'phonetic alphabet'

The various  species of whales  inhabiting Earth’s oceans employ different types of vocalizations to communicate. Sperm whales, the largest of the  toothed whales , communicate using bursts of clicking noises — called codas — sounding a bit like Morse code.

A new analysis of years of vocalizations by sperm whales in the eastern Caribbean has found that their system of communication is more sophisticated than previously known, exhibiting a complex internal structure replete with a “phonetic alphabet.” The researchers identified similarities to aspects of other animal communication systems — and even human language.

Like all marine mammals, sperm whales are very  social animals , with their calls an integral part of this. The new study has provided a fuller understanding of how these whales communicate.

“The research shows that the expressivity of sperm whale calls is much larger than previously thought,” said Pratyusha Sharma, a Massachusetts Institute of Technology doctoral student in robotics and machine learning and lead author of the study published on Tuesday in the journal  Nature Communications .

“We do not know yet what they are saying. We are studying the calls in their behavioral contexts next to understand what sperm whales might be communicating about,” said Sharma.

Sperm whales, which can reach about 60 feet (18 meters) long, have the largest brain of any animal. They are deep divers, feeding on giant squid and other prey.

The researchers are part of the Project CETI (Cetacean Translation Initiative) Machine Learning Team. Using traditional statistical analysis and artificial intelligence, they examined calls made by about 60 whales recorded by the Dominica Sperm Whale Project, a research program that has assembled a large dataset on the species.

“Why are they exchanging these codas? What information might they be sharing?” asked study co-author Shane Gero, Project CETI’s lead biologist and Dominica Sperm Whale Project founder, also affiliated with Carleton University in Canada.

“I think it’s likely that they use codas to coordinate as a family, organize babysitting, foraging and defense,” Gero said.

Variations in the number, rhythm and tempo of the clicks produced different types of codas, the researchers found. The whales, among other things, altered the duration of the codas and sometimes added an extra click at the end, like a suffix in human language.

“All of these different codas that we see are actually built by combining a comparatively simple set of smaller pieces,” said study co-author Jacob Andreas, an MIT computer science professor and Project CETI member.

People combine sounds — often corresponding to letters of the alphabet — to produce words that carry meaning, then produce sequences of words to create sentences to convey more complex meanings.

For people, Sharma said, “There are two levels of combination.” The lower level is sounds to words. The higher level is words to sentences.

Sperm whales, Sharma said, also use a two-level combination of features to form codas, and codas are then sequenced together as the whales communicate. The lower level has similarities to letters in an alphabet, Sharma said.

“Every communication system is tailored to the environment and animal society in which it has evolved,” Sharma added.

The communication system used by sperm whales differs, for example, from the  “songs” of humpback whales  — and, for that matter, from the whistles, chirps, croaks and assorted other vocalizations by various animals.

“Human language is unique in many ways, yes,” Gero said. “But I suspect we will find many patterns, structures and aspects thought to be unique to humans in other species, including whales, as science progresses — and perhaps also features and aspects of animal communications which humans do not possess.”

If scientists can decipher the meaning of what the sperm whales are “saying,” should people try to communicate with them?

“I think there’s a lot more research that we have to do before we know whether it’s a good idea to try to communicate with them, or really even to have a sense of whether that will be possible,” Andreas said.

“At the same time, I’m optimistic that we’re going to be able to learn a lot more about what information is actually encoded in these vocalizations that we’re listening to, what sort of information is contained in these clicks and these codas, as we start to understand the behavioral context in which this occurs,” Andreas added.

Controlled morphology of a new 3D Co(II) metal–organic framework (Co-MOF) via green sonochemical synthesis: crystallography, Hirshfeld surface analysis

  • Published: 09 May 2024
  • Volume 26 , article number  100 , ( 2024 )

Cite this article

research on complex analysis

  • Seyedeh Elahe Hosseini 1 ,
  • Mohammad Kazem Mohammadi 1 ,
  • Payam Hayati 2 ,
  • Haman Tavakkoli 1 &
  • Ayeh Rayatzadeh 1  

Explore all metrics

Nanostructures of a cobalt(II) metal–organic framework (MOF), denoted as 4,4′,4″-s-triazin-1,3,5-triyltri-p-aminobenzoate (TATAB) [[Co 2 (TATAB)(OH)(H 2 O) 2 ].H 2 O.0.6O] n {1 } , were successfully synthesized using two different experimental techniques: solvothermal and sonochemical strategies. Remarkably, both methods yielded an identical crystal structure. Various characterization techniques, including powder X-ray diffraction (PXRD), scanning electron microscopy (SEM), and Fourier transform infrared spectroscopy (FT-IR), were employed to analyze all the compounds. Compound contains cobalt ions (Co 2+ ) that were determined to be six-coordinated through the analysis of single-crystal X-ray diffraction (SCXRD). The effect of various factors such as temperature, reaction time, reactant concentration, and ultrasonic energy on the synthesis and final morphology of the compounds obtained by sonochemical method was investigated. Finally, Hirshfeld surface analysis (HAS) of compound was conducted. The molecular descriptors obtained at the BLYP/6–311 +  + g (d, p) level of theory framework indicate a unique electronic structure for this complex, characterized by low chemical hardness ( η  = 1.702 eV), high electrophilicity ( ω  = 3.637 eV), and a narrow HOMO–LUMO gap (1.55 eV). These descriptors suggest that this complex can be considered a favorable nucleophile in interactions with proteins.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price includes VAT (Russian Federation)

Instant access to the full article PDF.

Rent this article via DeepDyve

Institutional subscriptions

research on complex analysis

Marandi F et al (2017) Synthesis, spectral and X-ray diffraction of two new 2D lead (II) coordination polymers formed by nicotinic acid N-oxide linkers. J Mol Struct 1149:92–98

Article   CAS   Google Scholar  

Yang Y et al (2009) Supramolecular networks of hexanuclear cadmium (II): synthesis, crystal structure and emission property. Inorg Chim Acta 362(9):3065–3068

Moon D et al (2006) Face-driven corner-linked octahedral nanocages: M6L8 cages formed by C 3-symmetric triangular facial ligands linked via C 4-symmetric square tetratopic PdII Ions at truncated octahedron corners. J Am Chem Soc 128(11):3530–3531

Article   CAS   PubMed   Google Scholar  

Norjmaa G et al (2021) Modeling kinetics and thermodynamics of guest encapsulation into the [M4L6] 12–supramolecular organometallic cage. J Chem Inf Model 61(9):4370–4381

Article   CAS   PubMed Central   PubMed   Google Scholar  

Evans OR, Lin W (2002) Crystal engineering of NLO materials based on metal− organic coordination networks. Acc Chem Res 35(7):511–522

Rashidi N et al (2021) Antibacterial and cytotoxicity assay of two new Zn (ii) complexes: synthesis, characterization, X-ray structure, topology, Hirshfeld surface and thermal analysis. J Mol Struct 1231:129947

Hanifehpour Y et al (2015) Sonochemical syntheses of two new flower-like nano-scale high coordinated lead (II) supramolecular coordination polymers. Ultrason Sonochem 23:282–288

Sharifzadeh Z, Morsali A (2022) Amine-functionalized metal-organic frameworks: from synthetic design to scrutiny in application. Coord Chem Rev 459:214445

Caneschi A et al (2001) Cobalt (II)-nitronyl nitroxide chains as molecular magnetic nanowires. Angew Chem Int Ed 40(9):1760–1763

Tanatani A, Mio MJ, Moore JS (2001) Chain length-dependent affinity of helical foldamers for a rodlike guest. J Am Chem Soc 123(8):1792–1793

Zhang Z et al (2023) Coordination-driven self-assembly of dibenzo-18-crown-6 functionalized Pt (II) metallacycles. Chin Chem Lett 34(2):107521

Roesky HW, Andruh M (2003) The interplay of coordinative, hydrogen bonding and π–π stacking interactions in sustaining supramolecular solid-state architectures: a study case of bis (4-pyridyl)-and bis (4-pyridyl-N-oxide) tectons. Coord Chem Rev 236(1–2):91–119

Das D, Roy S, Biradha K (2018) Crystal engineering with isosteric triether and triamine linked aromatic tri-carboxylic acids: iso-structurality and synthon interplay in their co-crystals and salts with bis (pyridyl) derivatives. New J Chem 42(24):19953–19962

Chen XM, Liu GF (2002) Double-stranded helices and molecular zippers assembled from single-stranded coordination polymers directed by supramolecular interactions. Chem A Eur J 8(20):4811–4817

Emerson AJ et al (2018) High-connectivity approach to a hydrolytically stable metal–organic framework for CO2 capture from flue gas. Chem Mater 30(19):6614–6618

Nabipour H et al (2020) Metal-organic frameworks for flame retardant polymers application: a critical review. Compos A Appl Sci Manuf 139:106113

Karimi M et al (2021) Metal–organic framework. Interface Science and Technology. Elsevier, pp 279–387

Google Scholar  

Liu W et al (2018) Cobalt complexes as an emerging class of catalysts for homogeneous hydrogenations. Acc Chem Res 51(8):1858–1869

Rahpeyma M, MJ Soltanian Fard, and P Hayati, Green chemistry syntheses of different morphology novel nano-sized cobalt (II) supramolecular: as a precursor for the synthesis of cobalt (II) oxide nanoparticles, thermal, Hirshfeld surface analysis, and biological activities . Iran J Chem Chem Eng 2023

Lin W, Rieter WJ, Taylor KM (2009) Modular synthesis of functional nanoscale coordination polymers. Angew Chem Int Ed 48(4):650–658

Spokoyny AM et al (2009) Infinite coordination polymer nano-and microparticle structures. Chem Soc Rev 38(5):1218–1227

Li H et al (2015) Multi-component coordination-driven self-assembly toward heterometallic macrocycles and cages. Coord Chem Rev 293:139–157

Article   Google Scholar  

Zheng S-L et al (2001) Toward designed assembly of microporous coordination networks constructed from silver (I)− hexamethylenetetramine layers. Inorg Chem 40(14):3562–3569

Kirillov AM (2011) Hexamethylenetetramine: an old new building block for design of coordination polymers. Coord Chem Rev 255(15–16):1603–1622

Barcikowski S et al (2019) Materials synthesis in a bubble. MRS Bull 44(5):382–391

Qiu L-G et al (2008) Facile synthesis of nanocrystals of a microporous metal–organic framework by an ultrasonic method and selective sensing of organoamines. Chem Commun 31:3642–3644

Sabouni R, Kazemian H, Rohani S (2010) A novel combined manufacturing technique for rapid production of IRMOF-1 using ultrasound and microwave energies. Chem Eng J 165(3):966–973

Safarifard V, Morsali A, Joo SW (2013) Sonochemical synthesis and characterization of nano-sized lead (II) 3D coordination polymer: precursor for the synthesis of lead (II) oxybromide nanoparticles. Ultrason Sonochem 20(5):1254–1260

Paulusse JM, Huijbers JP, Sijbesma RP (2006) Quantification of ultrasound-induced chain scission in PdII–phosphine coordination polymers. Chem Eur J 12(18):4928–4934

Suslick KS (1991) The sonochemical hot spot. J Acoust Soc Am 89:1885–1886

Haque E et al (2010) Synthesis of a metal–organic framework material, iron terephthalate, by ultrasound, microwave, and conventional electric heating: a kinetic study. Chem Eur J 16(3):1046–1052

Jung D-W et al (2010) Facile synthesis of MOF-177 by a sonochemical method using 1-methyl-2-pyrrolidinone as a solvent. Dalton Trans 39(11):2883–2887

Alavi MA, Morsali A (2010) Syntheses and characterization of Sr (OH) 2 and SrCO3 nanostructures by ultrasonic method. Ultrason Sonochem 17(1):132–138

Rigaku, C.-S., Expert 2.1 b43. The Woodlands, Texas, USA, and Rigaku Corporation, Tokyo, Japan, 2014

Sheldrick GM (2015) SHELXT–integrated space-group and crystal-structure determination. Acta Crystallogr A Found Adv 71(1):3–8

Article   PubMed Central   PubMed   Google Scholar  

Spek AL (2009) Structure validation in chemical crystallography. Acta Crystallogr D Biol Crystallogr 65(2):148–155

Nonius, B., SADABS. Bruker Nonius, Delft, The Netherlands, 2002

Altomare A et al (1999) SIR97: a new tool for crystal structure determination and refinement. J Appl Crystallogr 32(1):115–119

Turner, M., et al., CrystalExplorer17; University of Western Australia, 2017. (b) Spackman, MA; Jayatilaka, D. CrystEngComm, 2009 11 19

Blatov VA, Shevchenko AP, Proserpio DM (2014) Applied topological analysis of crystal structures with the program package ToposPro. Cryst Growth Des 14(7):3576–3586

Farrugia LJ (2012) WinGX and ORTEP for Windows: an update. J Appl Crystallogr 45(4):849–854

Abbasi A, Moradpour T, Van Hecke K (2015) A new 3D cobalt (II) metal–organic framework nanostructure for heavy metal adsorption. Inorg Chim Acta 430:261–267

Pakiari AH, Eshghi F (2017) Geometric and electronic structures of vanadium sub-nano clusters, Vn (n= 2–5), and their adsorption complexes with CO and O2 ligands: a DFT-NBO study. Phys Chem Res 5(3):601–615

CAS   Google Scholar  

Frisch M et al., Gaussian 09, Revision D. 01, Gaussian, Inc., Wallingford CT . See also: URL: http://www.gaussian.com , 2009

Wang YA, Liu S, Parr RG (1997) Laurent series expansions in density functional theory. Chem Phys Lett 267(1–2):14–22

Molegro A, MVD Molegro Virtual Docker 5.0. Molegro: Aarhus C, Denmark, 2011

Thomsen R, Christensen MH (2006) MolDock: a new technique for high-accuracy molecular docking. J Med Chem 49(11):3315–3321

Li CG et al (2022) Novel zinc (II) and nickel (II) complexes of a quinazoline-based ligand with an imidazole ring: Synthesis, spectroscopic property, antibacterial activities, time-dependent density functional theory calculations and Hirshfeld surface analysis. Appl Organomet Chem 36(5):e6622

Tyula YA et al (2018) A new supramolecular zinc (II) complex containing 4-biphenylcarbaldehyde isonicotinoylhydrazone ligand: nanostructure synthesis, catalytic activities and Hirshfeld surface analysis. Appl Organomet Chem 32(3):e4141

Chai YM et al (2022) Antimicrobial activities of two 1-D, 2-D, and 3-D mononuclear Mn (II) and dinuclear Bi (III) complexes: X-ray structures, spectroscopic, electrostatic potential, Hirshfeld surface analysis, and time-dependent/density functional theory studies. Appl Organomet Chem 36(6):e6682

Tasi G et al (1993) Calculation of electrostatic potential maps and atomic charges for large molecules. J Chem Inf Comput Sci 33(3):296–299

Download references

Acknowledgements

Support of this investigation by Islamic Azad University, Ahvaz Branch, is gratefully acknowledged.

Author information

Authors and affiliations.

Advanced Surface Engineering and Nano Materials Research Center, Department of Chemistry, Ahvaz Branch, Islamic Azad University, Ahvaz, Iran

Seyedeh Elahe Hosseini, Mohammad Kazem Mohammadi, Haman Tavakkoli & Ayeh Rayatzadeh

Organic and Nano Group, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran

Payam Hayati

You can also search for this author in PubMed   Google Scholar

Contributions

Elahe Hosseini worked on the synthesis of nanocomposite. Mohammad Kazem Mohammadi, Haman Tavakkoli, and Ayeh Rayatzadeh characterized the synthesized nanostructure and theory.

Corresponding author

Correspondence to Mohammad Kazem Mohammadi .

Ethics declarations

Competing interests.

The author(s) declare no competing interests.

Additional information

Publisher's note.

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

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 260 KB)

Rights and permissions.

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Hosseini, S.E., Mohammadi, M.K., Hayati, P. et al. Controlled morphology of a new 3D Co(II) metal–organic framework (Co-MOF) via green sonochemical synthesis: crystallography, Hirshfeld surface analysis. J Nanopart Res 26 , 100 (2024). https://doi.org/10.1007/s11051-024-05991-8

Download citation

Received : 09 February 2024

Accepted : 12 April 2024

Published : 09 May 2024

DOI : https://doi.org/10.1007/s11051-024-05991-8

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

  • Metal–organic framework
  • Sonochemical Synthesis
  • Crystallography
  • Hirshfeld Surface Analysis
  • Find a journal
  • Publish with us
  • Track your research

IMAGES

  1. (PDF) Complex Analysis (Maximum Principle and its Applications)

    research on complex analysis

  2. Complex Analysis, 2nd Edition (PDF)

    research on complex analysis

  3. (PDF) Lecture Course on Complex Analysis

    research on complex analysis

  4. Introduction to Complex Analysis

    research on complex analysis

  5. Introduction to Complex Analysis

    research on complex analysis

  6. Chapter 13 Review on Complex Analysis II (PDF)

    research on complex analysis

VIDEO

  1. Complex Analysis, Lec.- 15(More problems on Analytic function)

  2. complex analysis ex 1.4 questions 5-8

  3. complex analysis lec-10

  4. Complex Analysis: Lecture 24: review for Test 2

  5. complex analysis lec-06

  6. Complex Analysis: Lecture 1

COMMENTS

  1. 97715 PDFs

    Explore the latest full-text research PDFs, articles, conference papers, preprints and more on COMPLEX ANALYSIS. Find methods information, sources, references or conduct a literature review on ...

  2. Articles

    On the Toeplitz Algebra in the Case of All Entire Functions and All Functions Holomorphic in the Unit Disc. M. Jasiczak. OriginalPaper 02 March 2024 Article: 44. 1. …. Complex Analysis and Operator Theory (CAOT) is devoted to the publication of current research developments in the closely related fields of complex analysis ...

  3. Complex Analysis

    Complex analysis has become a basic course in mathematics, physics, and select engineering departments. This concise textbook provides a thorough introduction to the function theory of one complex variable. It presents the fundamental concepts with clarity and rigor, offering concise proofs that avoid lengthy and tedious arguments commonly ...

  4. Advancements in Complex Analysis

    About this book. The contributions to this volume are devoted to a discussion of state-of-the-art research and treatment of problems of a wide spectrum of areas in complex analysis ranging from pure to applied and interdisciplinary mathematical research. Topics covered include: holomorphic approximation, hypercomplex analysis, special functions ...

  5. Mathematics

    The Special Issue on "Complex Analysis and Its Applications", an Open Access Volume, will be devoted exclusively to the publication of high-quality research papers in complex analysis and its applications. Though complex analysis was created when complex numbers were invented by Gauss in 1831; but it opened entirely new doors in the 20th ...

  6. Special Issue : New Trends in Complex Analysis Researches

    Dear Colleagues, The new Special Issue aims to bring the newest results of the study on complex valued functions of one or several complex variables. The topic related to several variables was added as a tribute to the memory of the great researcher, Professor Gabriela Kohr from Babeș-Bolyai University, Cluj-Napoca, Romania.

  7. Complex analysis

    Complex analysis, traditionally known as the theory of functions of a complex variable, is the branch of mathematical analysis that investigates functions of complex numbers.It is helpful in many branches of mathematics, including algebraic geometry, number theory, analytic combinatorics, and applied mathematics, as well as in physics, including the branches of hydrodynamics, thermodynamics ...

  8. New Trends in Complex Analysis Research, 2nd Edition

    New Trends in Complex Analysis Research, 2nd Edition. A special issue of Mathematics (ISSN 2227-7390). This special issue belongs to the section "Algebra, Geometry and Topology". Deadline for manuscript submissions: closed (29 February 2024) | Viewed by 7040.

  9. PDF Second edition Complex Analysis

    complex analysis has developed, and continues to evolve into the future. With numerous worked examples and exercises, clear and direct proofs, and a view ... is author or coauthor of over 190 research papers and is the bestselling author of over 120 books, from research monographs and textbooks to popular science and science c-tion. His awards ...

  10. PDF Spring 2019 lecture notes

    It revolves around complex analytic functions. These are functions that have a complex derivative. Unlike calculus using real variables, the mere existence of a complex derivative has strong implications for the properties of the function. Complex analysis is a basic tool in many mathematical theories. By itself and through

  11. PDF Complex Analysis Lecture Notes

    ematics of complex analysis. •Complex dynamics, e.g., the iconic Mandelbrot set. See Fig. 2. There are many other applications and beautiful connections of complex analysis to other areas of mathematics. (If you run across some interesting ones, please let me know!) In the next section I will begin our journey into the subject by illustrating

  12. PDF Complex Analysis

    do undergraduate research or a senior capstone may also find a course in Complex Analysis as a useful stepping stone to projects in complex dynamics, differential geometry, or analytic number theory. Complex analysis is a subject that can serve many roles for different majors and types of students. The material and theorems reach into many ...

  13. Complex Analysis -- from Wolfram MathWorld

    Complex analysis is the study of complex numbers together with their derivatives, manipulation, and other properties. Complex analysis is an extremely powerful tool with an unexpectedly large number of practical applications to the solution of physical problems. Contour integration, for example, provides a method of computing difficult integrals by investigating the singularities of the ...

  14. Home

    Complex Analysis and its Synergies is a unique forum dedicated to the cross-fertilization of Complex Analysis with various other significant fields of mathematics and the physical sciences.. Showcases research and exposition surrounding major developments in modern mathematical analysis. Encourages high-quality exposition and incisive research.

  15. PDF An Introduction to Complex Analysis and Geometry

    Chapter 2 develops the basic properties of complex numbers, with a special em-phasis on the role of complex conjugation. The author's own research in complex analysis and geometry has often used polarization; this technique makes precise the sense in which we may treat zand zas independent variables. We will view complex

  16. Complex Geometry and Complex Analysis

    The members of the Complex Geometry and Complex Analysis group conduct research in the areas of Complex Geometry and Several Complex Variables, Algebraic Geometry, Complex Analysis, Differential Geometry, Diophantine Geometry, and Number Theory etc. The group consists of faculty members Gordon Heier, Shanyu Ji and Min Ru, as well as seven ...

  17. Axioms

    Feature papers represent the most advanced research with significant potential for high impact in the field. A Feature Paper should be a substantial original Article that involves several techniques or approaches, provides an outlook for future research directions and describes possible research applications. ... Modern complex analysis is ...

  18. Advancing Complexity Theory as a Qualitative Research Methodology

    Complexity theory provides the means to conceptualize the research problem as a complex adaptive system (CAS), focusing on the patterns of interactions between various system elements at different levels and times. ... Document analysis as a qualitative research method. Qualitative Research Journal (RMIT Training Pty Ltd trading as RMIT ...

  19. Graph machine learning for integrated multi-omics analysis

    Multi-omics profiling technologies dive deeply into molecular landscapes and reveal multiple facets of complex research problems, e.g., shedding light on exciting novel aspects of cancer biology ...

  20. Articles

    Pekka Pankka. Research 22 July 2021 Article: 26. 1. 2. 3. Next. Complex Analysis and its Synergies is a unique forum dedicated to the cross-fertilization of Complex Analysis with various other significant fields of ...

  21. Complex Analysis

    $\begingroup$ No one is saying that you can't do research in the field, but complex analysis is a vast subject and it's hard for someone with just an intro to do substantial research and produce results. Especially if the deadline is in 11 days - that's not at all realistic to expect to produce a research paper from scratch in 11 days, in any field. My advice is, if you want to do publishable ...

  22. Scientists document remarkable sperm whale 'phonetic alphabet'

    A new analysis of years of vocalizations by sperm whales in the eastern Caribbean has found that their system of communication is more sophisticated than previously known, exhibiting a complex ...

  23. Fundamentals of Real and Complex Analysis

    Chapter 2 focuses on topics in real analysis such as p -adic completion, Banach Contraction Mapping Theorem and its applications, Fourier series, Lebesgue measure and integration. One of this chapter's unique features is its treatment of functional equations. Chapter 3 covers the essential topics in complex analysis: it begins with a ...

  24. Geophysical Research Letters

    The last eruption occurred in the Cumbre Vieja volcanic complex on La Palma Island (2021) and had an unexpected magnitude in terms of lava volume, explosivity, and a significant impact on the economy and society of the island. This study used receiver function analysis to image the lithosphere up to a depth of 50 km.

  25. Controlled morphology of a new 3D Co(II) metal-organic ...

    As shown in Figs. 4 and 5, hydrogen bonds between hydrogen atoms of water molecules and oxygen atoms of carboxylate groups in the ligand 4,4′,4″-s-triazin-1,3,5-triyltri-p-aminobenzoate (TATAB) is formed, and the distance between two aromatic rings is 4.092 Å.Figure 6 also shows that this three-dimensional coordination complex is connected through intramolecular interactions and expands ...