1
|
Witte H, Künstner A, Hahn T, Bernard V, Stölting S, Kusch K, Nagarathinam K, Khandanpour C, von Bubnoff N, Bauer A, Grunert M, Hartung S, Arndt A, Steinestel K, Merz H, Busch H, Feller AC, Gebauer N. The mutational landscape and its longitudinal dynamics in relapsed and refractory classic Hodgkin lymphoma. Ann Hematol 2025; 104:1721-1733. [PMID: 39992429 PMCID: PMC12031843 DOI: 10.1007/s00277-025-06274-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
In classic Hodgkin-lymphoma (cHL), only a few cases recur, and only a limited fraction of patients is primary-refractory to standard-polychemotherapy. Underlying genomic features of unfavorable clinical courses remain sparsely characterized. Here, we investigated the genomic characteristics of primary-refractory/relapsed cHL in contrast with responders. Therefore, ultra-deep next-generation panel-sequencing was performed on a total of 59 FFPE-samples (20 responders, 26 relapsed (rHL: 11 initial-diagnosis, 15 relapse) and 13 primary-refractory (prHL: 8 initial-diagnosis, 5 progression) from 44 cHL-patients applying a hybrid-capture approach. We compared samples associated with distinct disease courses concerning their oncogenic drivers, mutational signatures, and perturbed pathways. Compared to responders, mutations in genes such as PMS2, PDGFRB, KAT6A, EPHB1, and HGF were detected more frequently in prHL/rHL. Additionally, we observed that in rHL or prHL, BARD1-mutations occur, whereas ETV1, NF1, and MET-mutations were eliminated through clonal selection. A significant enrichment of non-synonymous variants was detected in prHL compared to responders and a significant selection process in favor of NOTCH-pathway mutations driving rHL or prHL was observed. However, our analysis revealed a negative selection process for non-synonymous variants affecting the hippo-pathway. This study delineates distinct mutational signatures between responders and rHL/prHL, whilst illustrating longitudinal dynamics in mutational profiles using paired samples. Further, several exploitable therapeutic vulnerabilities for rHL and prHL were identified.
Collapse
Affiliation(s)
- Hanno Witte
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany.
- Department of Hematology and Oncology, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany.
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| | - Axel Künstner
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
- Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Thomas Hahn
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Veronica Bernard
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Stephanie Stölting
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Kathrin Kusch
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Kumar Nagarathinam
- Institute of Biochemistry, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Cyrus Khandanpour
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Nikolas von Bubnoff
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Arthur Bauer
- Department of Hematology and Oncology, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany
| | - Michael Grunert
- Department of Nuclear Medicine, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany
| | - Svenja Hartung
- Institute of Pathology, University Ulm, Albert-Einstein Allee 23, 89081, Ulm, Germany
| | - Annette Arndt
- Institute of Pathology and Molecularpathology, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany
| | - Konrad Steinestel
- Institute of Pathology and Molecularpathology, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany
| | - Hartmut Merz
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Hauke Busch
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
- Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Alfred C Feller
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Niklas Gebauer
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| |
Collapse
|
2
|
Mascia E, Nale V, Ferrè L, Sorosina M, Clarelli F, Chiodi A, Santoro S, Giordano A, Misra K, Cannizzaro M, Moiola L, Martinelli V, Milanesi L, Filippi M, Mosca E, Esposito F. Genetic Contribution to Medium-Term Disease Activity in Multiple Sclerosis. Mol Neurobiol 2025; 62:322-334. [PMID: 38850349 DOI: 10.1007/s12035-024-04264-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
Multiple sclerosis (MS) is a complex disorder characterized by high heterogeneity in terms of phenotypic expression, prognosis and treatment response. In the present study, we aimed to explore the genetic contribution to MS disease activity at different levels: genes, pathways and tissue-specific networks. Two cohorts of relapsing-remitting MS patients who started a first-line treatment (n = 1294) were enrolled to evaluate the genetic association with disease activity after 4 years of follow-up. The analyses were performed at whole-genome SNP and gene level, followed by the construction of gene-gene interaction networks specific for brain and lymphocytes. The resulting gene modules were evaluated to highlight key players from a topological and functional perspective. We identified 23 variants and 223 genes with suggestive association to 4-years disease activity, highlighting genes like PON2 involved in oxidative stress and in mitochondria functions and other genes, like ILRUN, involved in the modulation of the immune system. Network analyses led to the identification of a brain module composed of 228 genes and a lymphocytes module composed of 287 genes. The network analysis allowed us to prioritize genes relevant for their topological properties; among them, there are MPHOSPH9 (connector hub in both brain and lymphocyte module) and OPA1 (in brain module), two genes already implicated in MS. Modules showed the enrichment of both shared and tissue-specific pathways, mainly implicated in inflammation. In conclusion, our results suggest that the processes underlying disease activity act on shared mechanisms across brain and lymphocyte tissues.
Collapse
Affiliation(s)
- Elisabetta Mascia
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Nale
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Laura Ferrè
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology and Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Melissa Sorosina
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ferdinando Clarelli
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alice Chiodi
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Silvia Santoro
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonino Giordano
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology and Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Kaalindi Misra
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Miryam Cannizzaro
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology and Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Moiola
- Neurology and Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vittorio Martinelli
- Neurology and Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Massimo Filippi
- Neurology and Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ettore Mosca
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Federica Esposito
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurology and Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
3
|
Janyasupab P, Singhanat K, Warnnissorn M, Thuwajit P, Suratanee A, Plaimas K, Thuwajit C. Identification of Tumor Budding-Associated Genes in Breast Cancer through Transcriptomic Profiling and Network Diffusion Analysis. Biomolecules 2024; 14:896. [PMID: 39199284 PMCID: PMC11352152 DOI: 10.3390/biom14080896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Breast cancer has the highest diagnosis rate among all cancers. Tumor budding (TB) is recognized as a recent prognostic marker. Identifying genes specific to high-TB samples is crucial for hindering tumor progression and metastasis. In this study, we utilized an RNA sequencing technique, called TempO-Seq, to profile transcriptomic data from breast cancer samples, aiming to identify biomarkers for high-TB cases. Through differential expression analysis and mutual information, we identified seven genes (NOL4, STAR, C8G, NEIL1, SLC46A3, FRMD6, and SCARF2) that are potential biomarkers in breast cancer. To gain more relevant proteins, further investigation based on a protein-protein interaction network and the network diffusion technique revealed enrichment in the Hippo signaling and Wnt signaling pathways, promoting tumor initiation, invasion, and metastasis in several cancer types. In conclusion, these novel genes, recognized as overexpressed in high-TB samples, along with their associated pathways, offer promising therapeutic targets, thus advancing treatment and diagnosis for breast cancer.
Collapse
Affiliation(s)
- Panisa Janyasupab
- Advance Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Kodchanan Singhanat
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (K.S.); (P.T.)
| | - Malee Warnnissorn
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (K.S.); (P.T.)
| | - Apichat Suratanee
- Department of Mathematics, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand;
- Intelligent and Nonlinear Dynamics Innovations Research Center, Science and Technology Research Institute, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand
| | - Kitiporn Plaimas
- Advance Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (K.S.); (P.T.)
| |
Collapse
|
4
|
Meroni M, Chiappori F, Paolini E, Longo M, De Caro E, Mosca E, Chiodi A, Merelli I, Badiali S, Maggioni M, Mezzelani A, Valenti L, Ludovica Fracanzani A, Dongiovanni P. A novel gene signature to diagnose MASLD in metabolically unhealthy obese individuals. Biochem Pharmacol 2023; 218:115925. [PMID: 37981173 DOI: 10.1016/j.bcp.2023.115925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
Visceral adipose tissue (VAT) contributes to metabolic dysfunction-associated steatotic liver disease (MASLD), releasing lipogenic substrates and cytokines which promote inflammation. Metabolic healthy obese individuals (MHO) may shift towardsunhealthy ones (MUHO) who develop MASLD, although the mechanisms are still unexplained. Therefore, we aimed to identify dysfunctional pathways and transcriptomic signatures shared by liver and VAT and to outline novel obesity-related biomarkers which feature MASLD in MUHO subjects, at higher risk of progressive liver disease and extrahepatic comorbidities. We performed RNA-sequencing in 167 hepatic samples and in a subset of 79 matched VAT, stratified in MHO and MUHO. A validation analysis was performed in hepatic samples and primary adipocytes from 12 bariatric patients, by qRT-PCR and western blot. We identified a transcriptomic signature that discriminate MUHO vs MHO, including 498 deregulated genes in liver and 189 in VAT. According to pathway and network analyses, oxidative phosphorylation resulted the only significantly downregulated pathway in both tissues in MUHO subjects. Next, we highlighted 5 genes commonly deregulated in liver and VAT, encompassing C6, IGF1, OXA1L, NDUFB11 and KLHL5 and we built a tissue-related score by integrating their expressions. Accordingly to RNAseq data, serum levels of C6 and IGF1, which are the only secreted proteins among those included in the gene signature were downregulated in MUHO vs MHO. Finally, the expression pattern of this 5-genes was confirmed in hepatic and VAT samples. We firstly identified the liver and VAT transcriptional phenotype of MUHO and a gene signature associated with the presence of MASLD in these at risk individuals.
Collapse
Affiliation(s)
- Marica Meroni
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Federica Chiappori
- National Research Council - Institute for Biomedical Technologies, (ITB-CNR), 20054 Segrate, Italy
| | - Erika Paolini
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Miriam Longo
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Emilia De Caro
- Life and Medical Sciences Institute (LIMES), University of Bonn, Germany; System Medicine, Deutsches Zentrum Für Neurodegenerativen Erkrankugen (DZNE), Bonn, Germany
| | - Ettore Mosca
- National Research Council - Institute for Biomedical Technologies, (ITB-CNR), 20054 Segrate, Italy
| | - Alice Chiodi
- National Research Council - Institute for Biomedical Technologies, (ITB-CNR), 20054 Segrate, Italy
| | - Ivan Merelli
- National Research Council - Institute for Biomedical Technologies, (ITB-CNR), 20054 Segrate, Italy
| | - Sara Badiali
- Department of Surgery, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Maggioni
- Department of Pathology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandra Mezzelani
- National Research Council - Institute for Biomedical Technologies, (ITB-CNR), 20054 Segrate, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Precision Medicine Lab, Biological Resource Center, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Ludovica Fracanzani
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
5
|
Sharma N, Millstein J. CausNet: generational orderings based search for optimal Bayesian networks via dynamic programming with parent set constraints. BMC Bioinformatics 2023; 24:46. [PMID: 36788490 PMCID: PMC9926787 DOI: 10.1186/s12859-023-05159-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 01/24/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Finding a globally optimal Bayesian Network using exhaustive search is a problem with super-exponential complexity, which severely restricts the number of variables that can feasibly be included. We implement a dynamic programming based algorithm with built-in dimensionality reduction and parent set identification. This reduces the search space substantially and can be applied to large-dimensional data. We use what we call 'generational orderings' based search for optimal networks, which is a novel way to efficiently search the space of possible networks given the possible parent sets. The algorithm supports both continuous and categorical data, as well as continuous, binary and survival outcomes. RESULTS We demonstrate the efficacy of our algorithm on both synthetic and real data. In simulations, our algorithm performs better than three state-of-art algorithms that are currently used extensively. We then apply it to an Ovarian Cancer gene expression dataset with 513 genes and a survival outcome. Our algorithm is able to find an optimal network describing the disease pathway consisting of 6 genes leading to the outcome node in just 3.4 min on a personal computer with a 2.3 GHz Intel Core i9 processor with 16 GB RAM. CONCLUSIONS Our generational orderings based search for optimal networks is both an efficient and highly scalable approach for finding optimal Bayesian Networks and can be applied to 1000 s of variables. Using specifiable parameters-correlation, FDR cutoffs, and in-degree-one can increase or decrease the number of nodes and density of the networks. Availability of two scoring option-BIC and Bge-and implementation for survival outcomes and mixed data types makes our algorithm very suitable for many types of high dimensional data in a variety of fields.
Collapse
Affiliation(s)
- Nand Sharma
- Division of Biostatistics, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, USA.
| | - Joshua Millstein
- grid.42505.360000 0001 2156 6853Division of Biostatistics, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, USA
| |
Collapse
|
6
|
Röhl A, Baek SH, Kachroo P, Morrow JD, Tantisira K, Silverman EK, Weiss ST, Sharma A, Glass K, DeMeo DL. Protein interaction networks provide insight into fetal origins of chronic obstructive pulmonary disease. Respir Res 2022; 23:69. [PMID: 35331221 PMCID: PMC8944072 DOI: 10.1186/s12931-022-01963-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 02/08/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a leading cause of death in adults that may have origins in early lung development. It is a complex disease, influenced by multiple factors including genetic variants and environmental factors. Maternal smoking during pregnancy may influence the risk for diseases during adulthood, potentially through epigenetic modifications including methylation. METHODS In this work, we explore the fetal origins of COPD by utilizing lung DNA methylation marks associated with in utero smoke (IUS) exposure, and evaluate the network relationships between methylomic and transcriptomic signatures associated with adult lung tissue from former smokers with and without COPD. To identify potential pathobiological mechanisms that may link fetal lung, smoke exposure and adult lung disease, we study the interactions (physical and functional) of identified genes using protein-protein interaction networks. RESULTS We build IUS-exposure and COPD modules, which identify connected subnetworks linking fetal lung smoke exposure to adult COPD. Studying the relationships and connectivity among the different modules for fetal smoke exposure and adult COPD, we identify enriched pathways, including the AGE-RAGE and focal adhesion pathways. CONCLUSIONS The modules identified in our analysis add new and potentially important insights to understanding the early life molecular perturbations related to the pathogenesis of COPD. We identify AGE-RAGE and focal adhesion as two biologically plausible pathways that may reveal lung developmental contributions to COPD. We were not only able to identify meaningful modules but were also able to study interconnections between smoke exposure and lung disease, augmenting our knowledge about the fetal origins of COPD.
Collapse
Affiliation(s)
- Annika Röhl
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Seung Han Baek
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Priyadarshini Kachroo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jarrett D Morrow
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kelan Tantisira
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Pediatric Respiratory Medicine, University of California San Diego, San Diego, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Amitabh Sharma
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Center for Complex Network Research, Northeastern University, Boston, MA, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
7
|
Tarozzi M, Bartoletti-Stella A, Dall'Olio D, Matteuzzi T, Baiardi S, Parchi P, Castellani G, Capellari S. Identification of recurrent genetic patterns from targeted sequencing panels with advanced data science: a case-study on sporadic and genetic neurodegenerative diseases. BMC Med Genomics 2022; 15:26. [PMID: 35144616 PMCID: PMC8830183 DOI: 10.1186/s12920-022-01173-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/02/2022] [Indexed: 11/10/2022] Open
Abstract
Background Targeted Next Generation Sequencing is a common and powerful approach used in both clinical and research settings. However, at present, a large fraction of the acquired genetic information is not used since pathogenicity cannot be assessed for most variants. Further complicating this scenario is the increasingly frequent description of a poli/oligogenic pattern of inheritance showing the contribution of multiple variants in increasing disease risk. We present an approach in which the entire genetic information provided by target sequencing is transformed into binary data on which we performed statistical, machine learning, and network analyses to extract all valuable information from the entire genetic profile. To test this approach and unbiasedly explore the presence of recurrent genetic patterns, we studied a cohort of 112 patients affected either by genetic Creutzfeldt–Jakob (CJD) disease caused by two mutations in the PRNP gene (p.E200K and p.V210I) with different penetrance or by sporadic Alzheimer disease (sAD). Results Unsupervised methods can identify functionally relevant sources of variation in the data, like haplogroups and polymorphisms that do not follow Hardy–Weinberg equilibrium, such as the NOTCH3 rs11670823 (c.3837 + 21 T > A). Supervised classifiers can recognize clinical phenotypes with high accuracy based on the mutational profile of patients. In addition, we found a similar alteration of allele frequencies compared the European population in sporadic patients and in V210I-CJD, a poorly penetrant PRNP mutation, and sAD, suggesting shared oligogenic patterns in different types of dementia. Pathway enrichment and protein–protein interaction network revealed different altered pathways between the two PRNP mutations. Conclusions We propose this workflow as a possible approach to gain deeper insights into the genetic information derived from target sequencing, to identify recurrent genetic patterns and improve the understanding of complex diseases. This work could also represent a possible starting point of a predictive tool for personalized medicine and advanced diagnostic applications. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01173-4.
Collapse
Affiliation(s)
- M Tarozzi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - A Bartoletti-Stella
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - D Dall'Olio
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - T Matteuzzi
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - S Baiardi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - P Parchi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - G Castellani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - S Capellari
- IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
8
|
Mosca E, Bersanelli M, Matteuzzi T, Di Nanni N, Castellani G, Milanesi L, Remondini D. Characterization and comparison of gene-centered human interactomes. Brief Bioinform 2021; 22:bbab153. [PMID: 34010955 PMCID: PMC8574298 DOI: 10.1093/bib/bbab153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 01/04/2023] Open
Abstract
The complex web of macromolecular interactions occurring within cells-the interactome-is the backbone of an increasing number of studies, but a clear consensus on the exact structure of this network is still lacking. Different genome-scale maps of human interactome have been obtained through several experimental techniques and functional analyses. Moreover, these maps can be enriched through literature-mining approaches, and different combinations of various 'source' databases have been used in the literature. It is therefore unclear to which extent the various interactomes yield similar results when used in the context of interactome-based approaches in network biology. We compared a comprehensive list of human interactomes on the basis of topology, protein complexes, molecular pathways, pathway cross-talk and disease gene prediction. In a general context of relevant heterogeneity, our study provides a series of qualitative and quantitative parameters that describe the state of the art of human interactomes and guidelines for selecting interactomes in future applications.
Collapse
Affiliation(s)
- Ettore Mosca
- Institute of Biomedical Technologies, National Research Council, Segrate (Milan), 20090, Italy
| | - Matteo Bersanelli
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele (Milan), 20090, Italy
| | - Tommaso Matteuzzi
- Department of Physics and Astronomy, University of Bologna, Bologna, 40127, Italy
| | - Noemi Di Nanni
- Institute of Biomedical Technologies, National Research Council, Segrate (Milan), 20090, Italy
| | - Gastone Castellani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, 40127, Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, National Research Council, Segrate (Milan), 20090, Italy
| | - Daniel Remondini
- Department of Physics and Astronomy, University of Bologna, Bologna, 40127, Italy
| |
Collapse
|
9
|
Picart-Armada S, Thompson WK, Buil A, Perera-Lluna A. The effect of statistical normalization on network propagation scores. Bioinformatics 2021; 37:845-852. [PMID: 33070187 PMCID: PMC8097756 DOI: 10.1093/bioinformatics/btaa896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 09/18/2020] [Accepted: 10/07/2020] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Network diffusion and label propagation are fundamental tools in computational biology, with applications like gene-disease association, protein function prediction and module discovery. More recently, several publications have introduced a permutation analysis after the propagation process, due to concerns that network topology can bias diffusion scores. This opens the question of the statistical properties and the presence of bias of such diffusion processes in each of its applications. In this work, we characterized some common null models behind the permutation analysis and the statistical properties of the diffusion scores. We benchmarked seven diffusion scores on three case studies: synthetic signals on a yeast interactome, simulated differential gene expression on a protein-protein interaction network and prospective gene set prediction on another interaction network. For clarity, all the datasets were based on binary labels, but we also present theoretical results for quantitative labels. RESULTS Diffusion scores starting from binary labels were affected by the label codification and exhibited a problem-dependent topological bias that could be removed by the statistical normalization. Parametric and non-parametric normalization addressed both points by being codification-independent and by equalizing the bias. We identified and quantified two sources of bias-mean value and variance-that yielded performance differences when normalizing the scores. We provided closed formulae for both and showed how the null covariance is related to the spectral properties of the graph. Despite none of the proposed scores systematically outperformed the others, normalization was preferred when the sought positive labels were not aligned with the bias. We conclude that the decision on bias removal should be problem and data-driven, i.e. based on a quantitative analysis of the bias and its relation to the positive entities. AVAILABILITY The code is publicly available at https://github.com/b2slab/diffuBench and the data underlying this article are available at https://github.com/b2slab/retroData. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Sergio Picart-Armada
- B2SLab, Departament d'Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, CIBER-BBN, Barcelona, 08028, Spain.,Esplugues de Llobregat, Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, Barcelona, 08950, Spain
| | - Wesley K Thompson
- Mental Health Center Sct. Hans, 4000 Roskilde, Denmark.,Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Alfonso Buil
- Mental Health Center Sct. Hans, 4000 Roskilde, Denmark
| | - Alexandre Perera-Lluna
- B2SLab, Departament d'Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, CIBER-BBN, Barcelona, 08028, Spain.,Esplugues de Llobregat, Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, Barcelona, 08950, Spain
| |
Collapse
|
10
|
Janyasupab P, Suratanee A, Plaimas K. Network diffusion with centrality measures to identify disease-related genes. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:2909-2929. [PMID: 33892577 DOI: 10.3934/mbe.2021147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Disease-related gene prioritization is one of the most well-established pharmaceutical techniques used to identify genes that are important to a biological process relevant to a disease. In identifying these essential genes, the network diffusion (ND) approach is a widely used technique applied in gene prioritization. However, there is still a large number of candidate genes that need to be evaluated experimentally. Therefore, it would be of great value to develop a new strategy to improve the precision of the prioritization. Given the efficiency and simplicity of centrality measures in capturing a gene that might be important to the network structure, herein, we propose a technique that extends the scope of ND through a centrality measure to identify new disease-related genes. Five common centrality measures with different aspects were examined for integration in the traditional ND model. A total of 40 diseases were used to test our developed approach and to find new genes that might be related to a disease. Results indicated that the best measure to combine with the diffusion is closeness centrality. The novel candidate genes identified by the model for all 40 diseases were provided along with supporting evidence. In conclusion, the integration of network centrality in ND is a simple but effective technique to discover more precise disease-related genes, which is extremely useful for biomedical science.
Collapse
Affiliation(s)
- Panisa Janyasupab
- Advanced Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Apichat Suratanee
- Intelligent and Nonlinear Dynamic Innovations Research Center, Department of Mathematics, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok 10800, Thailand
| | - Kitiporn Plaimas
- Advanced Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Omics Science and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
11
|
Erten C, Houdjedj A, Kazan H. Ranking cancer drivers via betweenness-based outlier detection and random walks. BMC Bioinformatics 2021; 22:62. [PMID: 33568049 PMCID: PMC7877041 DOI: 10.1186/s12859-021-03989-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 01/31/2021] [Indexed: 12/04/2022] Open
Abstract
Background Recent cancer genomic studies have generated detailed molecular data on a large number of cancer patients. A key remaining problem in cancer genomics is the identification of driver genes. Results We propose BetweenNet, a computational approach that integrates genomic data with a protein-protein interaction network to identify cancer driver genes. BetweenNet utilizes a measure based on betweenness centrality on patient specific networks to identify the so-called outlier genes that correspond to dysregulated genes for each patient. Setting up the relationship between the mutated genes and the outliers through a bipartite graph, it employs a random-walk process on the graph, which provides the final prioritization of the mutated genes. We compare BetweenNet against state-of-the art cancer gene prioritization methods on lung, breast, and pan-cancer datasets. Conclusions Our evaluations show that BetweenNet is better at recovering known cancer genes based on multiple reference databases. Additionally, we show that the GO terms and the reference pathways enriched in BetweenNet ranked genes and those that are enriched in known cancer genes overlap significantly when compared to the overlaps achieved by the rankings of the alternative methods.
Collapse
Affiliation(s)
- Cesim Erten
- Department of Computer Engineering, Antalya Bilim University, Antalya, Turkey
| | - Aissa Houdjedj
- Electrical and Computer Engineering Graduate Program, Antalya Bilim University, Antalya, Turkey
| | - Hilal Kazan
- Department of Computer Engineering, Antalya Bilim University, Antalya, Turkey.
| |
Collapse
|
12
|
Cho JW, Son J, Ha SJ, Lee I. Systems biology analysis identifies TNFRSF9 as a functional marker of tumor-infiltrating regulatory T-cell enabling clinical outcome prediction in lung cancer. Comput Struct Biotechnol J 2021; 19:860-868. [PMID: 33598101 PMCID: PMC7851794 DOI: 10.1016/j.csbj.2021.01.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 12/21/2022] Open
Abstract
Regulatory T cells (Tregs) are enriched in the tumor microenvironment and play key roles in immune evasion of cancer cells. Cell surface markers specific for tumor-infiltrating Tregs (TI-Tregs) can be effectively targeted to enhance antitumor immunity and used for stratification of immunotherapy outcomes. Here, we present a systems biology approach to identify functional cell surface markers for TI-Tregs. We selected differentially expressed genes for surface proteins of TI-Tregs and compared these with other CD4+ T cells using bulk RNA-sequencing data from murine lung cancer models. Thereafter, we filtered for human orthologues with conserved expression in TI-Tregs using single-cell transcriptome data from patients with non-small cell lung cancer (NSCLC). To evaluate the functional importance of expression-based markers of TI-Tregs, we utilized network-based measure of context-associated centrality in a Treg-specific coregulatory network. We identified TNFRSF9 (also known as 4-1BB or CD137), a previously reported target for enhancing antitumor immunity, among the final candidates for TI-Treg markers with high functional importance score. We found that the low TNFRSF9 expression level in Tregs was associated with enhanced overall survival rate and response to anti-PD-1 immunotherapy in patients with NSCLC, proposing that TNFRSF9 promotes immune suppressive activity of Tregs in tumor. Collectively, these results demonstrated that integrative transcriptome and network analysis can facilitate the discovery of functional markers of tumor-specific immune cells to develop novel therapeutic targets and biomarkers for boosting cancer immunotherapy.
Collapse
Affiliation(s)
- Jae-Won Cho
- Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jimin Son
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
13
|
Xie J, Yin Y, Yang F, Sun J, Wang J. Differential Network Analysis Reveals Regulatory Patterns in Neural Stem Cell Fate Decision. Interdiscip Sci 2021; 13:91-102. [PMID: 33439459 DOI: 10.1007/s12539-020-00415-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 12/11/2020] [Accepted: 12/22/2020] [Indexed: 11/30/2022]
Abstract
Deciphering regulatory patterns of neural stem cell (NSC) differentiation with multiple stages is essential to understand NSC differentiation mechanisms. Recent single-cell transcriptome datasets became available at individual differentiation. However, a systematic and integrative analysis of multiple datasets at multiple temporal stages of NSC differentiation is lacking. In this study, we propose a new method integrating prior information to construct three gene regulatory networks at pair-wise stages of transcriptome and apply this method to investigate five NSC differentiation paths on four different single-cell transcriptome datasets. By constructing gene regulatory networks for each path, we delineate their regulatory patterns via differential topology and network diffusion analyses. We find 12 common differentially expressed genes among the five NSC differentiation paths, with one common regulatory pattern (Gsk3b_App_Cdk5) shared by all paths. The identified regulatory pattern, partly supported by previous experimental evidence, is essential to all differentiation paths, but it plays a different role in each path when regulating other genes. Together, our integrative analysis provides both common and specific regulatory mechanisms for each of the five NSC differentiation paths.
Collapse
Affiliation(s)
- Jiang Xie
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Yiting Yin
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Fuzhang Yang
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Jiamin Sun
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai, China.
| |
Collapse
|
14
|
Singh RK, Yadav BS, Mohapatra TM. Molecular targets and system biology approaches for drug repurposing against SARS-CoV-2. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2020; 44:193. [PMID: 33230386 PMCID: PMC7675379 DOI: 10.1186/s42269-020-00444-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/03/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND COVID-19, a pandemic declared by WHO, has infected about 39.5 million and killed about 1.1 million people throughout the world. There is the urgent need of more studies to identify the novel drug targets and the drug candidates against it to handle the situation. MAIN BODY To virtually screen various drugs against SARS-CoV-2, the scientists need the detail information about the various drug targets identified till date. The present review provides the information about almost all the drug targets, including structural and non-structural proteins of virus as well as host cell surface receptors, that can be used for virtual screening of drugs. Moreover, this review also focuses on the different network analysis tools that have been used for the identification of new drug targets and candidate repurposable drugs against SARS-CoV-2. CONCLUSION This review provides important insights of various drug targets and the network analysis tools to young bioinformaticians and will help in creating pace to the drug repurposing strategy for COVID-19 disease.
Collapse
Affiliation(s)
- Rahul Kunwar Singh
- Department of Microbiology School of Life Sciences, H.N.B. Garhwal University, Srinagar (Garhwal), Uttarakhand 246174 India
| | | | - Tribhuvan Mohan Mohapatra
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005 India
| |
Collapse
|
15
|
Barel G, Herwig R. NetCore: a network propagation approach using node coreness. Nucleic Acids Res 2020; 48:e98. [PMID: 32735660 PMCID: PMC7515737 DOI: 10.1093/nar/gkaa639] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/22/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
We present NetCore, a novel network propagation approach based on node coreness, for phenotype–genotype associations and module identification. NetCore addresses the node degree bias in PPI networks by using node coreness in the random walk with restart procedure, and achieves improved re-ranking of genes after propagation. Furthermore, NetCore implements a semi-supervised approach to identify phenotype-associated network modules, which anchors the identification of novel candidate genes at known genes associated with the phenotype. We evaluated NetCore on gene sets from 11 different GWAS traits and showed improved performance compared to the standard degree-based network propagation using cross-validation. Furthermore, we applied NetCore to identify disease genes and modules for Schizophrenia GWAS data and pan-cancer mutation data. We compared the novel approach to existing network propagation approaches and showed the benefits of using NetCore in comparison to those. We provide an easy-to-use implementation, together with a high confidence PPI network extracted from ConsensusPathDB, which can be applied to various types of genomics data in order to obtain a re-ranking of genes and functionally relevant network modules.
Collapse
Affiliation(s)
- Gal Barel
- Department of Computational Molecular Biology, Max-Planck-Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany
| | - Ralf Herwig
- Department of Computational Molecular Biology, Max-Planck-Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany
| |
Collapse
|
16
|
Ruan P, Wang S. DiSNEP: a Disease-Specific gene Network Enhancement to improve Prioritizing candidate disease genes. Brief Bioinform 2020; 22:5925270. [PMID: 33064143 DOI: 10.1093/bib/bbaa241] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 07/25/2020] [Accepted: 08/29/2020] [Indexed: 12/27/2022] Open
Abstract
Biological network-based strategies are useful in prioritizing genes associated with diseases. Several comprehensive human gene networks such as STRING, GIANT and HumanNet were developed and used in network-assisted algorithms to identify disease-associated genes. However, none of these networks are disease-specific and may not accurately reflect gene interactions for a specific disease. Aiming to improve disease gene prioritization using networks, we propose a Disease-Specific Network Enhancement Prioritization (DiSNEP) framework. DiSNEP first enhances a comprehensive gene network specifically for a disease through a diffusion process on a gene-gene similarity matrix derived from disease omics data. The enhanced disease-specific gene network thus better reflects true gene interactions for the disease and may improve prioritizing disease-associated genes subsequently. In simulations, DiSNEP that uses an enhanced disease-specific network prioritizes more true signal genes than comparison methods using a general gene network or without prioritization. Applications to prioritize cancer-associated gene expression and DNA methylation signal genes for five cancer types from The Cancer Genome Atlas (TCGA) project suggest that more prioritized candidate genes by DiSNEP are cancer-related according to the DisGeNET database than those prioritized by the comparison methods, consistently across all five cancer types considered, and for both gene expression and DNA methylation signal genes.
Collapse
|
17
|
Ahmed R, Baali I, Erten C, Hoxha E, Kazan H. MEXCOwalk: mutual exclusion and coverage based random walk to identify cancer modules. Bioinformatics 2020; 36:872-879. [PMID: 31432076 DOI: 10.1093/bioinformatics/btz655] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/03/2019] [Accepted: 08/18/2019] [Indexed: 12/25/2022] Open
Abstract
MOTIVATION Genomic analyses from large cancer cohorts have revealed the mutational heterogeneity problem which hinders the identification of driver genes based only on mutation profiles. One way to tackle this problem is to incorporate the fact that genes act together in functional modules. The connectivity knowledge present in existing protein-protein interaction (PPI) networks together with mutation frequencies of genes and the mutual exclusivity of cancer mutations can be utilized to increase the accuracy of identifying cancer driver modules. RESULTS We present a novel edge-weighted random walk-based approach that incorporates connectivity information in the form of protein-protein interactions (PPIs), mutual exclusivity and coverage to identify cancer driver modules. MEXCOwalk outperforms several state-of-the-art computational methods on TCGA pan-cancer data in terms of recovering known cancer genes, providing modules that are capable of classifying normal and tumor samples and that are enriched for mutations in specific cancer types. Furthermore, the risk scores determined with output modules can stratify patients into low-risk and high-risk groups in multiple cancer types. MEXCOwalk identifies modules containing both well-known cancer genes and putative cancer genes that are rarely mutated in the pan-cancer data. The data, the source code and useful scripts are available at: https://github.com/abu-compbio/MEXCOwalk. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Rafsan Ahmed
- Electrical and Computer Engineering Graduate Program, Department of Computer Engineering, Antalya Bilim University, Antalya 07190, Turkey
| | - Ilyes Baali
- Electrical and Computer Engineering Graduate Program, Department of Computer Engineering, Antalya Bilim University, Antalya 07190, Turkey
| | - Cesim Erten
- Department of Computer Engineering, Antalya Bilim University, Antalya 07190, Turkey
| | - Evis Hoxha
- Department of Computer Engineering, Antalya Bilim University, Antalya 07190, Turkey
| | - Hilal Kazan
- Department of Computer Engineering, Antalya Bilim University, Antalya 07190, Turkey
| |
Collapse
|
18
|
Di Nanni N, Bersanelli M, Milanesi L, Mosca E. Network Diffusion Promotes the Integrative Analysis of Multiple Omics. Front Genet 2020; 11:106. [PMID: 32180795 PMCID: PMC7057719 DOI: 10.3389/fgene.2020.00106] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/29/2020] [Indexed: 02/01/2023] Open
Abstract
The development of integrative methods is one of the main challenges in bioinformatics. Network-based methods for the analysis of multiple gene-centered datasets take into account known and/or inferred relations between genes. In the last decades, the mathematical machinery of network diffusion—also referred to as network propagation—has been exploited in several network-based pipelines, thanks to its ability of amplifying association between genes that lie in network proximity. Indeed, network diffusion provides a quantitative estimation of network proximity between genes associated with one or more different data types, from simple binary vectors to real vectors. Therefore, this powerful data transformation method has also been increasingly used in integrative analyses of multiple collections of biological scores and/or one or more interaction networks. We present an overview of the state of the art of bioinformatics pipelines that use network diffusion processes for the integrative analysis of omics data. We discuss the fundamental ways in which network diffusion is exploited, open issues and potential developments in the field. Current trends suggest that network diffusion is a tool of broad utility in omics data analysis. It is reasonable to think that it will continue to be used and further refined as new data types arise (e.g. single cell datasets) and the identification of system-level patterns will be considered more and more important in omics data analysis.
Collapse
Affiliation(s)
- Noemi Di Nanni
- Institute of Biomedical Technologies, National Research Council, Milan, Italy.,Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy
| | - Matteo Bersanelli
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy.,National Institute of Nuclear Physics (INFN), Bologna, Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Ettore Mosca
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| |
Collapse
|
19
|
Bersanelli M, Mosca E, Milanesi L, Bazzani A, Castellani G. Frailness and resilience of gene networks predicted by detection of co-occurring mutations via a stochastic perturbative approach. Sci Rep 2020; 10:2643. [PMID: 32060296 PMCID: PMC7021762 DOI: 10.1038/s41598-020-59036-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 11/22/2019] [Indexed: 11/13/2022] Open
Abstract
In recent years complex networks have been identified as powerful mathematical frameworks for the adequate modeling of many applied problems in disparate research fields. Assuming a Master Equation (ME) modeling the exchange of information within the network, we set up a perturbative approach in order to investigate how node alterations impact on the network information flow. The main assumption of the perturbed ME (pME) model is that the simultaneous presence of multiple node alterations causes more or less intense network frailties depending on the specific features of the perturbation. In this perspective the collective behavior of a set of molecular alterations on a gene network is a particularly adapt scenario for a first application of the proposed method, since most diseases are neither related to a single mutation nor to an established set of molecular alterations. Therefore, after characterizing the method numerically, we applied as a proof of principle the pME approach to breast cancer (BC) somatic mutation data downloaded from Cancer Genome Atlas (TCGA) database. For each patient we measured the network frailness of over 90 significant subnetworks of the protein-protein interaction network, where each perturbation was defined by patient-specific somatic mutations. Interestingly the frailness measures depend on the position of the alterations on the gene network more than on their amount, unlike most traditional enrichment scores. In particular low-degree mutations play an important role in causing high frailness measures. The potential applicability of the proposed method is wide and suggests future development in the control theory context.
Collapse
Affiliation(s)
- Matteo Bersanelli
- Department of Physics and Astronomy, University of Bologna, Bologna, 40127, Italy. .,National Institute for Nuclear Physics (INFN), Bologna, 40127, Italy.
| | - Ettore Mosca
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, 20090, Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, 20090, Italy
| | - Armando Bazzani
- Department of Physics and Astronomy, University of Bologna, Bologna, 40127, Italy
| | - Gastone Castellani
- Department of Physics and Astronomy, University of Bologna, Bologna, 40127, Italy
| |
Collapse
|
20
|
Sinha N, Chowdhury S, Sarkar RR. Molecular basis of drug resistance in smoothened receptor: An
in silico
study of protein resistivity and specificity. Proteins 2019; 88:514-526. [DOI: 10.1002/prot.25830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/28/2019] [Accepted: 09/17/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Noopur Sinha
- Chemical Engineering and Process Development DivisionCSIR‐National Chemical Laboratory Pune Maharashtra India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad India
| | - Saikat Chowdhury
- Chemical Engineering and Process Development DivisionCSIR‐National Chemical Laboratory Pune Maharashtra India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad India
| | - Ram Rup Sarkar
- Chemical Engineering and Process Development DivisionCSIR‐National Chemical Laboratory Pune Maharashtra India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad India
| |
Collapse
|
21
|
Gabaldón T. Recent trends in molecular diagnostics of yeast infections: from PCR to NGS. FEMS Microbiol Rev 2019; 43:517-547. [PMID: 31158289 PMCID: PMC8038933 DOI: 10.1093/femsre/fuz015] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022] Open
Abstract
The incidence of opportunistic yeast infections in humans has been increasing over recent years. These infections are difficult to treat and diagnose, in part due to the large number and broad diversity of species that can underlie the infection. In addition, resistance to one or several antifungal drugs in infecting strains is increasingly being reported, severely limiting therapeutic options and showcasing the need for rapid detection of the infecting agent and its drug susceptibility profile. Current methods for species and resistance identification lack satisfactory sensitivity and specificity, and often require prior culturing of the infecting agent, which delays diagnosis. Recently developed high-throughput technologies such as next generation sequencing or proteomics are opening completely new avenues for more sensitive, accurate and fast diagnosis of yeast pathogens. These approaches are the focus of intensive research, but translation into the clinics requires overcoming important challenges. In this review, we provide an overview of existing and recently emerged approaches that can be used in the identification of yeast pathogens and their drug resistance profiles. Throughout the text we highlight the advantages and disadvantages of each methodology and discuss the most promising developments in their path from bench to bedside.
Collapse
Affiliation(s)
- Toni Gabaldón
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- ICREA, Pg Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
22
|
Di Nanni N, Gnocchi M, Moscatelli M, Milanesi L, Mosca E. Gene relevance based on multiple evidences in complex networks. Bioinformatics 2019; 36:865-871. [PMID: 31504182 PMCID: PMC9883679 DOI: 10.1093/bioinformatics/btz652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/17/2019] [Accepted: 08/19/2019] [Indexed: 02/02/2023] Open
Abstract
MOTIVATION Multi-omics approaches offer the opportunity to reconstruct a more complete picture of the molecular events associated with human diseases, but pose challenges in data analysis. Network-based methods for the analysis of multi-omics leverage the complex web of macromolecular interactions occurring within cells to extract significant patterns of molecular alterations. Existing network-based approaches typically address specific combinations of omics and are limited in terms of the number of layers that can be jointly analysed. In this study, we investigate the application of network diffusion to quantify gene relevance on the basis of multiple evidences (layers). RESULTS We introduce a gene score (mND) that quantifies the relevance of a gene in a biological process taking into account the network proximity of the gene and its first neighbours to other altered genes. We show that mND has a better performance over existing methods in finding altered genes in network proximity in one or more layers. We also report good performances in recovering known cancer genes. The pipeline described in this article is broadly applicable, because it can handle different types of inputs: in addition to multi-omics datasets, datasets that are stratified in many classes (e.g., cell clusters emerging from single cell analyses) or a combination of the two scenarios. AVAILABILITY AND IMPLEMENTATION The R package 'mND' is available at URL: https://www.itb.cnr.it/mnd. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Noemi Di Nanni
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy,Department of Industrial and Information Engineering, University of Pavia, Italy
| | - Matteo Gnocchi
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy
| | - Marco Moscatelli
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy
| | - Luciano Milanesi
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy
| | | |
Collapse
|
23
|
Di Nanni N, Bersanelli M, Cupaioli FA, Milanesi L, Mezzelani A, Mosca E. Network-Based Integrative Analysis of Genomics, Epigenomics and Transcriptomics in Autism Spectrum Disorders. Int J Mol Sci 2019; 20:E3363. [PMID: 31323926 PMCID: PMC6651137 DOI: 10.3390/ijms20133363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 01/16/2023] Open
Abstract
Current studies suggest that autism spectrum disorders (ASDs) may be caused by many genetic factors. In fact, collectively considering multiple studies aimed at characterizing the basic pathophysiology of ASDs, a large number of genes has been proposed. Addressing the problem of molecular data interpretation using gene networks helps to explain genetic heterogeneity in terms of shared pathways. Besides, the integrative analysis of multiple omics has emerged as an approach to provide a more comprehensive view of a disease. In this work, we carry out a network-based meta-analysis of the genes reported as associated with ASDs by studies that involved genomics, epigenomics, and transcriptomics. Collectively, our analysis provides a prioritization of the large number of genes proposed to be associated with ASDs, based on genes' relevance within the intracellular circuits, the strength of the supporting evidence of association with ASDs, and the number of different molecular alterations affecting genes. We discuss the presence of the prioritized genes in the SFARI (Simons Foundation Autism Research Initiative) database and in gene networks associated with ASDs by other investigations. Lastly, we provide the full results of our analyses to encourage further studies on common targets amenable to therapy.
Collapse
Affiliation(s)
- Noemi Di Nanni
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate (MI), Italy
- Department of Industrial and Information Engineering, University of Pavia, Via Ferrata 5, 27100 Pavia, Italy
| | - Matteo Bersanelli
- Department of Physics and Astronomy, University of Bologna, Via B. Pichat 6/2, 40127 Bologna, Italy
- National Institute of Nuclear Physics (INFN), 40127 Bologna, Italy
| | - Francesca Anna Cupaioli
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate (MI), Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate (MI), Italy
| | - Alessandra Mezzelani
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate (MI), Italy
| | - Ettore Mosca
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate (MI), Italy.
| |
Collapse
|
24
|
Alexander-Dann B, Pruteanu LL, Oerton E, Sharma N, Berindan-Neagoe I, Módos D, Bender A. Developments in toxicogenomics: understanding and predicting compound-induced toxicity from gene expression data. Mol Omics 2018; 14:218-236. [PMID: 29917034 PMCID: PMC6080592 DOI: 10.1039/c8mo00042e] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022]
Abstract
The toxicogenomics field aims to understand and predict toxicity by using 'omics' data in order to study systems-level responses to compound treatments. In recent years there has been a rapid increase in publicly available toxicological and 'omics' data, particularly gene expression data, and a corresponding development of methods for its analysis. In this review, we summarize recent progress relating to the analysis of RNA-Seq and microarray data, review relevant databases, and highlight recent applications of toxicogenomics data for understanding and predicting compound toxicity. These include the analysis of differentially expressed genes and their enrichment, signature matching, methods based on interaction networks, and the analysis of co-expression networks. In the future, these state-of-the-art methods will likely be combined with new technologies, such as whole human body models, to produce a comprehensive systems-level understanding of toxicity that reduces the necessity of in vivo toxicity assessment in animal models.
Collapse
Affiliation(s)
- Benjamin Alexander-Dann
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
| | - Lavinia Lorena Pruteanu
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
- Babeş-Bolyai University
, Institute for Doctoral Studies
,
1 Kogălniceanu Street
, Cluj-Napoca 400084
, Romania
- University of Medicine and Pharmacy “Iuliu Haţieganu”
, MedFuture Research Centre for Advanced Medicine
,
23 Marinescu Street/4-6 Pasteur Street
, Cluj-Napoca 400337
, Romania
| | - Erin Oerton
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
| | - Nitin Sharma
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
| | - Ioana Berindan-Neagoe
- University of Medicine and Pharmacy “Iuliu Haţieganu”
, MedFuture Research Centre for Advanced Medicine
,
23 Marinescu Street/4-6 Pasteur Street
, Cluj-Napoca 400337
, Romania
- University of Medicine and Pharmacy “Iuliu Haţieganu”
, Research Center for Functional Genomics
, Biomedicine and Translational Medicine
,
23 Marinescu Street
, Cluj-Napoca 400337
, Romania
- The Oncology Institute “Prof. Dr Ion Chiricuţă”
, Department of Functional Genomics and Experimental Pathology
,
34-36 Republicii Street
, Cluj-Napoca 400015
, Romania
| | - Dezső Módos
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
| | - Andreas Bender
- University of Cambridge
, Centre for Molecular Informatics
, Department of Chemistry
,
Lensfield Road
, Cambridge CB2 1EW
, UK
.
;
| |
Collapse
|
25
|
Picart-Armada S, Thompson WK, Buil A, Perera-Lluna A. diffuStats: an R package to compute diffusion-based scores on biological networks. Bioinformatics 2018; 34:533-534. [PMID: 29029016 PMCID: PMC5860365 DOI: 10.1093/bioinformatics/btx632] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/28/2017] [Accepted: 10/04/2017] [Indexed: 12/31/2022] Open
Abstract
Summary Label propagation and diffusion over biological networks are a common mathematical formalism in computational biology for giving context to molecular entities and prioritizing novel candidates in the area of study. There are several choices in conceiving the diffusion process-involving the graph kernel, the score definitions and the presence of a posterior statistical normalization-which have an impact on the results. This manuscript describes diffuStats, an R package that provides a collection of graph kernels and diffusion scores, as well as a parallel permutation analysis for the normalized scores, that eases the computation of the scores and their benchmarking for an optimal choice. Availability and implementation The R package diffuStats is publicly available in Bioconductor, https://bioconductor.org, under the GPL-3 license. Contact sergi.picart@upc.edu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Sergio Picart-Armada
- B2SLab, Departament d’Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, CIBER-BBN, Barcelona, Spain
- Department of Biomedical Engineering, Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Wesley K Thompson
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Roskilde, Denmark
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Alfonso Buil
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Roskilde, Denmark
| | - Alexandre Perera-Lluna
- B2SLab, Departament d’Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, CIBER-BBN, Barcelona, Spain
- Department of Biomedical Engineering, Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| |
Collapse
|
26
|
Mosca E, Bersanelli M, Gnocchi M, Moscatelli M, Castellani G, Milanesi L, Mezzelani A. Network Diffusion-Based Prioritization of Autism Risk Genes Identifies Significantly Connected Gene Modules. Front Genet 2017; 8:129. [PMID: 28993790 PMCID: PMC5622537 DOI: 10.3389/fgene.2017.00129] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 09/04/2017] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorder (ASD) is marked by a strong genetic heterogeneity, which is underlined by the low overlap between ASD risk gene lists proposed in different studies. In this context, molecular networks can be used to analyze the results of several genome-wide studies in order to underline those network regions harboring genetic variations associated with ASD, the so-called "disease modules." In this work, we used a recent network diffusion-based approach to jointly analyze multiple ASD risk gene lists. We defined genome-scale prioritizations of human genes in relation to ASD genes from multiple studies, found significantly connected gene modules associated with ASD and predicted genes functionally related to ASD risk genes. Most of them play a role in synapsis and neuronal development and function; many are related to syndromes that can be in comorbidity with ASD and the remaining are involved in epigenetics, cell cycle, cell adhesion and cancer.
Collapse
Affiliation(s)
- Ettore Mosca
- Bioinformatics Group, Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Italy
| | - Matteo Bersanelli
- Applied Physics Group, Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Matteo Gnocchi
- Bioinformatics Group, Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Italy
| | - Marco Moscatelli
- Bioinformatics Group, Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Italy
| | - Gastone Castellani
- Applied Physics Group, Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Luciano Milanesi
- Bioinformatics Group, Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Italy
| | - Alessandra Mezzelani
- Bioinformatics Group, Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Italy
| |
Collapse
|
27
|
Monti D, Ostan R, Borelli V, Castellani G, Franceschi C. Inflammaging and human longevity in the omics era. Mech Ageing Dev 2016; 165:129-138. [PMID: 28038993 DOI: 10.1016/j.mad.2016.12.008] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 12/21/2016] [Indexed: 11/24/2022]
Abstract
Inflammaging is a recent theory of aging originally proposed in 2000 where data and conceptualizations regarding the aging of the immune system (immunosenescence) and the evolution of immune responses from invertebrates to mammals converged. This theory has received an increasing number of citations and experimental confirmations. Here we present an updated version of inflammaging focused on omics data - particularly on glycomics - collected on centenarians, semi-supercentenarians and their offspring. Accordingly, we arrived to the following conclusions: i) inflammaging has a structure where specific combinations of pro- and anti-inflammatory mediators are involved; ii) inflammaging is systemic and more complex than we previously thought, as many organs, tissues and cell types participate in producing pro- and anti-inflammatory stimuli defined "molecular garbage"; iii) inflammaging is dynamic, can be propagated locally to neighboring cells and systemically from organ to organ by circulating products and microvesicles, and amplified by chronic age-related diseases constituting a "local fire", which in turn produces additional inflammatory stimuli and molecular garbage; iv) an integrated Systems Medicine approach is urgently needed to let emerge a robust and highly informative set/combination of omics markers able to better grasp the complex molecular core of inflammaging in elderly and centenarians.
Collapse
Affiliation(s)
- Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Rita Ostan
- Interdepartmental Centre "L. Galvani" (CIG) and Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy.
| | - Vincenzo Borelli
- Interdepartmental Centre "L. Galvani" (CIG) and Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy
| | - Gastone Castellani
- Department of Physics and Astronomy DIFA, University of Bologna, Viale Berti Pichat 6/2, 40127, Bologna, Italy
| | - Claudio Franceschi
- IRCCS, Institute of Neurological Sciences of Bologna, Via Altura 3, 40139 Bologna, Italy
| |
Collapse
|