1
|
Loscalzo J. Timing the Taming of Vascular Inflammation. N Engl J Med 2025; 392:712-714. [PMID: 39938099 DOI: 10.1056/nejme2416329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Affiliation(s)
- Joseph Loscalzo
- Brigham and Women's Hospital, Boston
- Harvard Medical School, Boston
| |
Collapse
|
2
|
Halu A, Chelvanambi S, Decano JL, Matamalas JT, Whelan M, Asano T, Kalicharran N, Singh SA, Loscalzo J, Aikawa M. Integrating pharmacogenomics and cheminformatics with diverse disease phenotypes for cell type-guided drug discovery. Genome Med 2025; 17:7. [PMID: 39833831 PMCID: PMC11744892 DOI: 10.1186/s13073-025-01431-x] [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: 01/19/2023] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Large-scale pharmacogenomic resources, such as the Connectivity Map (CMap), have greatly assisted computational drug discovery. However, despite their widespread use, CMap-based methods have thus far been agnostic to the biological activity of drugs as well as to the genomic effects of drugs in multiple disease contexts. Here, we present a network-based statistical approach, Pathopticon, that uses CMap to build cell type-specific gene-drug perturbation networks and integrates these networks with cheminformatic data and diverse disease phenotypes to prioritize drugs in a cell type-dependent manner. METHODS We build cell type-specific gene-drug perturbation networks from CMap data using a statistical procedure we call Quantile-based Instance Z-score Consensus (QUIZ-C). Using these networks and a large-scale disease-gene network consisting of 569 disease signatures from the Enrichr database, we calculate Pathophenotypic Congruity Scores (PACOS) between input gene signatures and drug perturbation signatures and combine these scores with cheminformatic data from ChEMBL to prioritize drugs. We benchmark our approach by calculating area under the receiver operating characteristic curves (AUROC) for 73 gene sets from the Molecular Signatures Database (MSigDB) using target gene expression profiles from the Comparative Toxicogenomics Database (CTD). We validate the drugs predicted in our proofs-of-concept using real-time polymerase chain reaction (qPCR) experiments. RESULTS Cell type-specific gene-drug perturbation networks built using QUIZ-C are topologically distinct, reflecting the biological uniqueness of the cell lines in CMap, and are enriched in known drug targets. Pathopticon demonstrates a better prediction performance than solely cheminformatic measures as well as state-of-the-art network and deep learning-based methods. Top predictions made by Pathopticon have high chemical structural diversity, suggesting their potential for building compound libraries. In proof-of-concept applications on vascular diseases, we demonstrate that Pathopticon helps guide in vitro experiments by identifying pathways that are potentially regulated by the predicted therapeutic candidates. CONCLUSIONS Our network-based analytical framework integrating pharmacogenomics and cheminformatics (available at https://github.com/r-duh/Pathopticon ) provides a feasible blueprint for a cell type-specific drug discovery and repositioning platform with broad implications for the efficiency and success of drug development.
Collapse
Affiliation(s)
- Arda Halu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA.
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA.
| | - Sarvesh Chelvanambi
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Julius L Decano
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Joan T Matamalas
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Mary Whelan
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Takaharu Asano
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Namitra Kalicharran
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Joseph Loscalzo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masanori Aikawa
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA.
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA.
| |
Collapse
|
3
|
Dai Z, Hu T, Wei J, Wang X, Cai C, Gu Y, Hu Y, Wang W, Wu Q, Fang J. Network-based identification and mechanism exploration of active ingredients against Alzheimer's disease via targeting endoplasmic reticulum stress from traditional chinese medicine. Comput Struct Biotechnol J 2024; 23:506-519. [PMID: 38261917 PMCID: PMC10796977 DOI: 10.1016/j.csbj.2023.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/25/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative disease that leads to dementia and poses a serious threat to the health of the elderly. Traditional Chinese medicine (TCM) presents as a promising novel therapeutic therapy for preventing and treating dementia. Studies have shown that natural products derived from kidney-tonifying herbs can effectively inhibit AD. Furthermore, endoplasmic reticulum (ER) stress is a critical factor in the pathology of AD. Regulation of ER stress is a crucial approach to prevent and treat AD. Thus, in this study, we first collected kidney-tonifying herbs, integrated chemical ingredients from multiple TCM databases, and constructed a comprehensive drug-target network. Subsequently, we employed the endophenotype network (network proximity) method to identify potential active ingredients in kidney-tonifying herbs that prevented AD via regulating ER stress. By combining the predicted outcomes, we discovered that 32 natural products could ameliorate AD pathology via regulating ER stress. After a comprehensive evaluation of the multi-network model and systematic pharmacological analyses, we further selected several promising compounds for in vitro testing in the APP-SH-SY5Y cell model. Experimental results showed that echinacoside and danthron were able to effectively reduce ER stress-mediated neuronal apoptosis by inhibiting the expression levels of BIP, p-PERK, ATF6, and CHOP in APP-SH-SY5Y cells. Overall, this study utilized the endophenotype network to preliminarily decipher the effective material basis and potential molecular mechanism of kidney-tonifying Chinese medicine for prevention and treatment of AD.
Collapse
Affiliation(s)
- Zhao Dai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Tian Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Junwen Wei
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xue Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Chuipu Cai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yong Gu
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Hainan Medical University, Haikou 570100, China
| | - Yunhui Hu
- Tasly Pharmaceutical Group Co., Ltd., Tianjin 300402, China
| | - Wenjia Wang
- Tasly Pharmaceutical Group Co., Ltd., Tianjin 300402, China
| | - Qihui Wu
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Hainan Medical University, Haikou 570100, China
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
4
|
Zhang S, Niu Q, Zong W, Song Q, Tian S, Wang J, Liu J, Zhang H, Wang Z, Li B. Endotype-driven Co-module mechanisms of danhong injection in the Co-treatment of cardiovascular and cerebrovascular diseases: A modular-based drug and disease integrated analysis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118287. [PMID: 38705429 DOI: 10.1016/j.jep.2024.118287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiovascular and cerebrovascular diseases are the leading causes of death worldwide and interact closely with each other. Danhong Injection (DHI) is a widely used preparation for the co-treatment of brain and heart diseases (CTBH). However, the underlying molecular endotype mechanisms of DHI in the CTBH remain unclear. AIM OF THIS STUDY To elucidate the underlying endotype mechanisms of DHI in the CTBH. MATERIALS AND METHODS In this study, we proposed a modular-based disease and drug-integrated analysis (MDDIA) strategy for elucidating the systematic CTBH mechanisms of DHI using high-throughput transcriptome-wide sequencing datasets of DHI in the treatment of patients with stable angina pectoris (SAP) and cerebral infarction (CI). First, we identified drug-targeted modules of DHI and disease modules of SAP and CI based on the gene co-expression networks of DHI therapy and the protein-protein interaction networks of diseases. Moreover, module proximity-based topological analyses were applied to screen CTBH co-module pairs and driver genes of DHI. At the same time, the representative driver genes were validated via in vitro experiments on hypoxia/reoxygenation-related cardiomyocytes and neuronal cell lines of H9C2 and HT22. RESULTS Seven drug-targeted modules of DHI and three disease modules of SAP and CI were identified by co-expression networks. Five modes of modular relationships between the drug and disease modules were distinguished by module proximity-based topological analyses. Moreover, 13 targeted module pairs and 17 driver genes associated with DHI in the CTBH were also screened. Finally, the representative driver genes AKT1, EDN1, and RHO were validated by in vitro experiments. CONCLUSIONS This study, based on clinical sequencing data and modular topological analyses, integrated diseases and drug targets. The CTBH mechanism of DHI may involve the altered expression of certain driver genes (SRC, STAT3, EDN1, CYP1A1, RHO, RELA) through various enriched pathways, including the Wnt signaling pathway.
Collapse
Affiliation(s)
- Siqi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qikai Niu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wenjing Zong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qi Song
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Siwei Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jingai Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huamin Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
5
|
Menichetti G, Barabási AL, Loscalzo J. Decoding the Foodome: Molecular Networks Connecting Diet and Health. Annu Rev Nutr 2024; 44:257-288. [PMID: 39207880 PMCID: PMC11610447 DOI: 10.1146/annurev-nutr-062322-030557] [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] [Indexed: 09/04/2024]
Abstract
Diet, a modifiable risk factor, plays a pivotal role in most diseases, from cardiovascular disease to type 2 diabetes mellitus, cancer, and obesity. However, our understanding of the mechanistic role of the chemical compounds found in food remains incomplete. In this review, we explore the "dark matter" of nutrition, going beyond the macro- and micronutrients documented by national databases to unveil the exceptional chemical diversity of food composition. We also discuss the need to explore the impact of each compound in the presence of associated chemicals and relevant food sources and describe the tools that will allow us to do so. Finally, we discuss the role of network medicine in understanding the mechanism of action of each food molecule. Overall, we illustrate the important role of network science and artificial intelligence in our ability to reveal nutrition's multifaceted role in health and disease.
Collapse
Affiliation(s)
- Giulia Menichetti
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA;
- Network Science Institute and Department of Physics, Northeastern University, Boston, Massachusetts, USA
- Harvard Data Science Initiative, Harvard University, Boston, Massachusetts, USA
| | - Albert-László Barabási
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA;
- Network Science Institute and Department of Physics, Northeastern University, Boston, Massachusetts, USA
- Department of Network and Data Science, Central European University, Budapest, Hungary
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
6
|
Pavlicev M, Wagner GP. Reading the palimpsest of cell interactions: What questions may we ask of the data? iScience 2024; 27:109670. [PMID: 38665209 PMCID: PMC11043885 DOI: 10.1016/j.isci.2024.109670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024] Open
Abstract
Biological function depends on the composition and structure of the organism, the latter describing the organization of interactions between parts. While cells in multicellular organisms are capable of a remarkable degree of autonomy, most functions do require cell communication: the coordination of functions (growth, differentiation, and apoptosis), the compartmentalization of cellular processes, and the integration of cells into higher levels of structural organization. A wealth of data on putative cell interactions has become available, yet its biological interpretation depends on our expectations about the structure of interaction networks. Here, we attempt to formulate basic questions to ask when interpreting cell interaction data. We build on the understanding that cells fulfill two general functions: the integrity-maintaining and the organismal service function. We derive the expected patterns of cell interactions considering two intertwined aspects: the functional and the evolutionary. Based on these, we propose guidelines for analysis and interpretation of transcriptional cell-interactome data.
Collapse
Affiliation(s)
- Mihaela Pavlicev
- Unit for Theoretical Biology, Department for Evolutionary Biology, University of Vienna, Vienna 1030, Austria
- Complexity Science Hub, Vienna 1090, Austria
| | - Günter P. Wagner
- Unit for Theoretical Biology, Department for Evolutionary Biology, University of Vienna, Vienna 1030, Austria
- Yale University, New Haven, CT 06520, USA
- Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
7
|
Fiocchi C. Omics and Multi-Omics in IBD: No Integration, No Breakthroughs. Int J Mol Sci 2023; 24:14912. [PMID: 37834360 PMCID: PMC10573814 DOI: 10.3390/ijms241914912] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/27/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
The recent advent of sophisticated technologies like sequencing and mass spectroscopy platforms combined with artificial intelligence-powered analytic tools has initiated a new era of "big data" research in various complex diseases of still-undetermined cause and mechanisms. The investigation of these diseases was, until recently, limited to traditional in vitro and in vivo biological experimentation, but a clear switch to in silico methodologies is now under way. This review tries to provide a comprehensive assessment of state-of-the-art knowledge on omes, omics and multi-omics in inflammatory bowel disease (IBD). The notion and importance of omes, omics and multi-omics in both health and complex diseases like IBD is introduced, followed by a discussion of the various omics believed to be relevant to IBD pathogenesis, and how multi-omics "big data" can generate new insights translatable into useful clinical tools in IBD such as biomarker identification, prediction of remission and relapse, response to therapy, and precision medicine. The pitfalls and limitations of current IBD multi-omics studies are critically analyzed, revealing that, regardless of the types of omes being analyzed, the majority of current reports are still based on simple associations of descriptive retrospective data from cross-sectional patient cohorts rather than more powerful longitudinally collected prospective datasets. Given this limitation, some suggestions are provided on how IBD multi-omics data may be optimized for greater clinical and therapeutic benefit. The review concludes by forecasting the upcoming incorporation of multi-omics analyses in the routine management of IBD.
Collapse
Affiliation(s)
- Claudio Fiocchi
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
8
|
Guthrie J, Ko¨stel Bal S, Lombardo SD, Mu¨ller F, Sin C, Hu¨tter CV, Menche J, Boztug K. AutoCore: A network-based definition of the core module of human autoimmunity and autoinflammation. SCIENCE ADVANCES 2023; 9:eadg6375. [PMID: 37656781 PMCID: PMC10848965 DOI: 10.1126/sciadv.adg6375] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 08/01/2023] [Indexed: 09/03/2023]
Abstract
Although research on rare autoimmune and autoinflammatory diseases has enabled definition of nonredundant regulators of homeostasis in human immunity, because of the single gene-single disease nature of many of these diseases, contributing factors were mostly unveiled in sequential and noncoordinated individual studies. We used a network-based approach for integrating a set of 186 inborn errors of immunity with predominant autoimmunity/autoinflammation into a comprehensive map of human immune dysregulation, which we termed "AutoCore." The AutoCore is located centrally within the interactome of all protein-protein interactions, connecting and pinpointing multidisease markers for a range of common, polygenic autoimmune/autoinflammatory diseases. The AutoCore can be subdivided into 19 endotypes that correspond to molecularly and phenotypically cohesive disease subgroups, providing a molecular mechanism-based disease classification and rationale toward systematic targeting for therapeutic purposes. Our study provides a proof of concept for using network-based methods to systematically investigate the molecular relationships between individual rare diseases and address a range of conceptual, diagnostic, and therapeutic challenges.
Collapse
Affiliation(s)
- Julia Guthrie
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Zimmermannplatz 10, A-1090 Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, A-1090 Vienna, Austria
- Max Perutz Labs, Vienna BioCenter Campus, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- Department of Structural and Computational Biology, University of Vienna, Dr.-Bohr-Gasse 9, 1030, Vienna Austria
| | - Sevgi Ko¨stel Bal
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Zimmermannplatz 10, A-1090 Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, A-1090 Vienna, Austria
- St. Anna Children’s Cancer Research Institute (CCRI), Zimmermannplatz 10, A-1090 Vienna, Austria
| | - Salvo Danilo Lombardo
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, A-1090 Vienna, Austria
- Max Perutz Labs, Vienna BioCenter Campus, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- Department of Structural and Computational Biology, University of Vienna, Dr.-Bohr-Gasse 9, 1030, Vienna Austria
| | - Felix Mu¨ller
- Max Perutz Labs, Vienna BioCenter Campus, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- Department of Structural and Computational Biology, University of Vienna, Dr.-Bohr-Gasse 9, 1030, Vienna Austria
| | - Celine Sin
- Max Perutz Labs, Vienna BioCenter Campus, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- Department of Structural and Computational Biology, University of Vienna, Dr.-Bohr-Gasse 9, 1030, Vienna Austria
| | - Christiane V. R. Hu¨tter
- Max Perutz Labs, Vienna BioCenter Campus, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter, A-1030 Vienna, Austria
| | - Jo¨rg Menche
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, A-1090 Vienna, Austria
- Max Perutz Labs, Vienna BioCenter Campus, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- Department of Structural and Computational Biology, University of Vienna, Dr.-Bohr-Gasse 9, 1030, Vienna Austria
- Faculty of Mathematics, University of Vienna, Oskar-Morgenstern-Platz 1, A-1090 Vienna, Austria
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Zimmermannplatz 10, A-1090 Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, A-1090 Vienna, Austria
- St. Anna Children’s Cancer Research Institute (CCRI), Zimmermannplatz 10, A-1090 Vienna, Austria
- St. Anna Children’s Hospital, Kinderspitalgasse 6, A-1090, Vienna, Austria
- Medical University of Vienna, Department of Pediatrics and Adolescent Medicine, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| |
Collapse
|
9
|
Pandey AK, Loscalzo J. Network medicine: an approach to complex kidney disease phenotypes. Nat Rev Nephrol 2023:10.1038/s41581-023-00705-0. [PMID: 37041415 DOI: 10.1038/s41581-023-00705-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 04/13/2023]
Abstract
Scientific reductionism has been the basis of disease classification and understanding for more than a century. However, the reductionist approach of characterizing diseases from a limited set of clinical observations and laboratory evaluations has proven insufficient in the face of an exponential growth in data generated from transcriptomics, proteomics, metabolomics and deep phenotyping. A new systematic method is necessary to organize these datasets and build new definitions of what constitutes a disease that incorporates both biological and environmental factors to more precisely describe the ever-growing complexity of phenotypes and their underlying molecular determinants. Network medicine provides such a conceptual framework to bridge these vast quantities of data while providing an individualized understanding of disease. The modern application of network medicine principles is yielding new insights into the pathobiology of chronic kidney diseases and renovascular disorders by expanding the understanding of pathogenic mediators, novel biomarkers and new options for renal therapeutics. These efforts affirm network medicine as a robust paradigm for elucidating new advances in the diagnosis and treatment of kidney disorders.
Collapse
Affiliation(s)
- Arvind K Pandey
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Saba L, Tagliagambe S. Quantitative medicine: Tracing the transition from holistic to reductionist approaches. A new "quantitative holism" is possible? J Public Health Res 2023; 12:22799036231182271. [PMID: 37361238 PMCID: PMC10286173 DOI: 10.1177/22799036231182271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
The practice of medicine has evolved significantly over time, from a more holistic to a reductionist or mechanistic approach. This paper briefly traces the history of medicine and the transition to quantitative medicine, which has enabled more personalized and targeted treatments, and improved understanding of the underlying biological mechanisms of disease. However, this shift has also presented some challenges and criticisms, including the danger of losing sight of the patient as a unique, whole individual. This paper explores the underlying principles and key contributions of quantitative medicine, as well as the context for its rise, including the development of new technologies and the influence of reductionist philosophies. The challenges and criticisms of this approach, and the need to balance reductionist and holistic approaches in order to achieve a comprehensive understanding of human health will be discussed. Ultimately, by integrating insights from philosophy, physics, and other fields, we may be able to develop new and innovative approaches that bridge the gap between reductionism and holism and improve patient outcomes with the new "quantitative holism."
Collapse
Affiliation(s)
- Luca Saba
- Luca Saba, University of Cagliari, SS 554 Monserrato, Cagliari 09124, Italy.
| | | |
Collapse
|
11
|
Sadegh S, Skelton J, Anastasi E, Maier A, Adamowicz K, Möller A, Kriege NM, Kronberg J, Haller T, Kacprowski T, Wipat A, Baumbach J, Blumenthal DB. Lacking mechanistic disease definitions and corresponding association data hamper progress in network medicine and beyond. Nat Commun 2023; 14:1662. [PMID: 36966134 PMCID: PMC10039912 DOI: 10.1038/s41467-023-37349-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/13/2023] [Indexed: 03/27/2023] Open
Abstract
A long-term objective of network medicine is to replace our current, mainly phenotype-based disease definitions by subtypes of health conditions corresponding to distinct pathomechanisms. For this, molecular and health data are modeled as networks and are mined for pathomechanisms. However, many such studies rely on large-scale disease association data where diseases are annotated using the very phenotype-based disease definitions the network medicine field aims to overcome. This raises the question to which extent the biases mechanistically inadequate disease annotations introduce in disease association data distort the results of studies which use such data for pathomechanism mining. We address this question using global- and local-scale analyses of networks constructed from disease association data of various types. Our results indicate that large-scale disease association data should be used with care for pathomechanism mining and that analyses of such data should be accompanied by close-up analyses of molecular data for well-characterized patient cohorts.
Collapse
Affiliation(s)
- Sepideh Sadegh
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - James Skelton
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
| | - Elisa Anastasi
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
| | - Andreas Maier
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Klaudia Adamowicz
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Anna Möller
- Biomedical Network Science Lab, Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Nils M Kriege
- Faculty of Computer Science, University of Vienna, Vienna, Austria
- Research Network Data Science, University of Vienna, Vienna, Austria
| | - Jaanika Kronberg
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Toomas Haller
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tim Kacprowski
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of Technische Universität Braunschweig and Hannover Medical School, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), TU Braunschweig, Braunschweig, Germany
| | - Anil Wipat
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Computational Biomedicine Lab, Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - David B Blumenthal
- Biomedical Network Science Lab, Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
12
|
Loscalzo J. Molecular interaction networks and drug development: Novel approach to drug target identification and drug repositioning. FASEB J 2023; 37:e22660. [PMID: 36468661 PMCID: PMC10107166 DOI: 10.1096/fj.202201683r] [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: 10/14/2022] [Revised: 10/27/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022]
Abstract
Conventional drug discovery requires identifying a protein target believed to be important for disease mechanism and screening compounds for those that beneficially alter the target's function. While this approach has been an effective one for decades, recent data suggest that its continued success is limited largely owing to the highly prevalent irreducibility of biologically complex systems that govern disease phenotype to a single primary disease driver. Network medicine, a new discipline that applies network science and systems biology to the analysis of complex biological systems and disease, offers a novel approach to overcoming these limitations of conventional drug discovery. Using the comprehensive protein-protein interaction network (interactome) as the template through which subnetworks that govern specific diseases are identified, potential disease drivers are unveiled and the effect of novel or repurposed drugs, used alone or in combination, is studied. This approach to drug discovery offers new and exciting unbiased possibilities for advancing our knowledge of disease mechanisms and precision therapeutics.
Collapse
Affiliation(s)
- Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
da Luz Moreira A, de Campos Lobato LF, de Lima Moreira JP, Luiz RR, Elia C, Fiocchi C, de Souza HSP. Geosocial Features and Loss of Biodiversity Underlie Variable Rates of Inflammatory Bowel Disease in a Large Developing Country: A Population-Based Study. Inflamm Bowel Dis 2022; 28:1696-1708. [PMID: 35089325 DOI: 10.1093/ibd/izab346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND The epidemiology of inflammatory bowel disease (IBD) in developing countries may uncover etiopathogenic factors. We investigated IBD prevalence in Brazil by investigating its geographic, spatial, and temporal distribution, and attempted to identify factors associated with its recent increase. METHODS A drug prescription database was queried longitudinally to identify patients and verify population distribution and density, race, urbanicity, sanitation, and Human Development Index. Prevalence was calculated using the number of IBD patients and the population estimated during the same decade. Data were matched to indices using linear regression analyses. RESULTS We identified 162 894 IBD patients, 59% with ulcerative colitis (UC) and 41% with Crohn's disease (CD). The overall prevalence of IBD was 80 per 100 000, with 46 per 100 000 for UC and 36 per 100 000 for CD. Estimated rates adjusted to total population showed that IBD more than triplicated from 2008 to 2017. The distribution of IBD demonstrated a South-to-North gradient that generally followed population apportionment. However, marked regional differences and disease clusters were identified that did not fit with conventionally accepted IBD epidemiological associations, revealing that the rise of IBD was variable. In some areas, loss of biodiversity was associated with high IBD prevalence. CONCLUSIONS When distribution is considered in the context of IBD prevalence, marked regional differences become evident. Despite a background of Westernization, hotspots of IBD are recognized that are not explained by population density, urbanicity, sanitation, or other indices but apparently are explained by biodiversity loss. Thus, the rise of IBD in developing countries is not uniform, but rather is one that varies depending on yet unexplored factors like geoecological conditions.
Collapse
Affiliation(s)
- Andre da Luz Moreira
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Inflammatory Bowel Disease Center, NYU Langone Health, NYU Grossman School of Medicine, New York, NY, USA
| | | | | | - Ronir Raggio Luiz
- Institute of Collective Health Studies, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Celeste Elia
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Claudio Fiocchi
- Department of Immunity & Inflammation, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Heitor Siffert Pereira de Souza
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Hickey SL, McKim A, Mancuso CA, Krishnan A. A network-based approach for isolating the chronic inflammation gene signatures underlying complex diseases towards finding new treatment opportunities. Front Pharmacol 2022; 13:995459. [PMCID: PMC9597699 DOI: 10.3389/fphar.2022.995459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Complex diseases are associated with a wide range of cellular, physiological, and clinical phenotypes. To advance our understanding of disease mechanisms and our ability to treat these diseases, it is critical to delineate the molecular basis and therapeutic avenues of specific disease phenotypes, especially those that are associated with multiple diseases. Inflammatory processes constitute one such prominent phenotype, being involved in a wide range of health problems including ischemic heart disease, stroke, cancer, diabetes mellitus, chronic kidney disease, non-alcoholic fatty liver disease, and autoimmune and neurodegenerative conditions. While hundreds of genes might play a role in the etiology of each of these diseases, isolating the genes involved in the specific phenotype (e.g., inflammation “component”) could help us understand the genes and pathways underlying this phenotype across diseases and predict potential drugs to target the phenotype. Here, we present a computational approach that integrates gene interaction networks, disease-/trait-gene associations, and drug-target information to accomplish this goal. We apply this approach to isolate gene signatures of complex diseases that correspond to chronic inflammation and use SAveRUNNER to prioritize drugs to reveal new therapeutic opportunities.
Collapse
Affiliation(s)
- Stephanie L. Hickey
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Alexander McKim
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, United States
- Genetics and Genome Sciences Program, Michigan State University, East Lansing, MI, United States
| | - Christopher A. Mancuso
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, United States
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Arjun Krishnan
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: Arjun Krishnan,
| |
Collapse
|
15
|
Network-based response module comprised of gene expression biomarkers predicts response to infliximab at treatment initiation in ulcerative colitis. Transl Res 2022; 246:78-86. [PMID: 35306220 DOI: 10.1016/j.trsl.2022.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/08/2022] [Indexed: 11/22/2022]
Abstract
This cross-cohort study aimed to (1) determine a network-based molecular signature that predicts the likelihood of inadequate response to the tumor necrosis factor-ɑ inhibitor (TNFi) therapy, infliximab, in ulcerative colitis (UC) patients, and (2) address biomarker irreproducibility across different cohort studies. Whole-transcriptome microarray data were derived from biopsies of affected colon tissue from 2 cohorts of infliximab-treated UC patients (training N = 24 and validation N = 22). Response was defined as endoscopic and histologic healing at 4-6 weeks and 8 weeks, respectively. From the training cohort, genes with RNA expression that significantly correlated with clinical response outcomes were mapped onto the Human Interactome network map of protein-protein interactions to identify a largest connected component (LCC) of proteins indicative of infliximab response status in UC. Expression levels of transcripts within the LCC were fed into a probabilistic neural network model to generate a classifier that predicts inadequate response to infliximab. A classifier predictive of inadequate response to infliximab was generated and tested in a cross-cohort, blinded fashion; the AUC was 0.83 and inadequate response was predicted with a 100% positive predictive value and 64% sensitivity. Genes separately identified from the 2 cohorts that correlated with response to infliximab appeared distinct but mapped onto the same network region of the Human Interactome, reflecting a common underlying biology of response among UC patients. Cross-cohort validation of a classifier predictive of infliximab response status in UC patients indicates that a molecular signature of non-response to TNFi therapies is present in patients' baseline gene expression data. The goal is to develop a diagnostic test that predicts which patients will have an inadequate response to targeted therapies and define new targets and pathways for therapeutic development.
Collapse
|
16
|
Sonawane AR, Aikawa E, Aikawa M. Connections for Matters of the Heart: Network Medicine in Cardiovascular Diseases. Front Cardiovasc Med 2022; 9:873582. [PMID: 35665246 PMCID: PMC9160390 DOI: 10.3389/fcvm.2022.873582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/19/2022] [Indexed: 01/18/2023] Open
Abstract
Cardiovascular diseases (CVD) are diverse disorders affecting the heart and vasculature in millions of people worldwide. Like other fields, CVD research has benefitted from the deluge of multiomics biomedical data. Current CVD research focuses on disease etiologies and mechanisms, identifying disease biomarkers, developing appropriate therapies and drugs, and stratifying patients into correct disease endotypes. Systems biology offers an alternative to traditional reductionist approaches and provides impetus for a comprehensive outlook toward diseases. As a focus area, network medicine specifically aids the translational aspect of in silico research. This review discusses the approach of network medicine and its application to CVD research.
Collapse
Affiliation(s)
- Abhijeet Rajendra Sonawane
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
17
|
You Y, Lai X, Pan Y, Zheng H, Vera J, Liu S, Deng S, Zhang L. Artificial intelligence in cancer target identification and drug discovery. Signal Transduct Target Ther 2022; 7:156. [PMID: 35538061 PMCID: PMC9090746 DOI: 10.1038/s41392-022-00994-0] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 03/14/2022] [Accepted: 04/05/2022] [Indexed: 02/08/2023] Open
Abstract
Artificial intelligence is an advanced method to identify novel anticancer targets and discover novel drugs from biology networks because the networks can effectively preserve and quantify the interaction between components of cell systems underlying human diseases such as cancer. Here, we review and discuss how to employ artificial intelligence approaches to identify novel anticancer targets and discover drugs. First, we describe the scope of artificial intelligence biology analysis for novel anticancer target investigations. Second, we review and discuss the basic principles and theory of commonly used network-based and machine learning-based artificial intelligence algorithms. Finally, we showcase the applications of artificial intelligence approaches in cancer target identification and drug discovery. Taken together, the artificial intelligence models have provided us with a quantitative framework to study the relationship between network characteristics and cancer, thereby leading to the identification of potential anticancer targets and the discovery of novel drug candidates.
Collapse
Affiliation(s)
- Yujie You
- College of Computer Science, Sichuan University, Chengdu, 610065, China
| | - Xin Lai
- Laboratory of Systems Tumor Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91052, Germany
| | - Yi Pan
- Faculty of Computer Science and Control Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Room D513, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China
| | - Huiru Zheng
- School of Computing, Ulster University, Belfast, BT15 1ED, UK
| | - Julio Vera
- Laboratory of Systems Tumor Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91052, Germany
| | - Suran Liu
- College of Computer Science, Sichuan University, Chengdu, 610065, China
| | - Senyi Deng
- Institute of Thoracic Oncology, Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610065, China.
| | - Le Zhang
- College of Computer Science, Sichuan University, Chengdu, 610065, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
18
|
Friedman SL, Pinzani M. Hepatic fibrosis 2022: Unmet needs and a blueprint for the future. Hepatology 2022; 75:473-488. [PMID: 34923653 DOI: 10.1002/hep.32285] [Citation(s) in RCA: 245] [Impact Index Per Article: 81.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022]
Abstract
Steady progress over four decades toward understanding the pathogenesis and clinical consequences of hepatic fibrosis has led to the expectation of effective antifibrotic drugs, yet none has been approved. Thus, an assessment of the field is timely, to clarify priorities and accelerate progress. Here, we highlight the successes to date but, more importantly, identify gaps and unmet needs, both experimentally and clinically. These include the need to better define cell-cell interactions and etiology-specific elements of fibrogenesis and their link to disease-specific drivers of portal hypertension. Success in treating viral hepatitis has revealed the remarkable capacity of the liver to degrade scar in reversing fibrosis, yet we know little of the mechanisms underlying this response. Thus, there is an exigent need to clarify the cellular and molecular mechanisms of fibrosis regression in order for therapeutics to mimic the liver's endogenous capacity. Better refined and more predictive in vitro and animal models will hasten drug development. From a clinical perspective, current diagnostics are improving but not always biologically plausible or sufficiently accurate to supplant biopsy. More urgently, digital pathology methods that leverage machine learning and artificial intelligence must be validated in order to capture more prognostic information from liver biopsies and better quantify the response to therapies. For more refined treatment of NASH, orthogonal approaches that integrate genetic, clinical, and pathological data sets may yield treatments for specific subphenotypes of the disease. Collectively, these and other advances will strengthen and streamline clinical trials and better link histologic responses to clinical outcomes.
Collapse
Affiliation(s)
- Scott L Friedman
- Division of Liver DiseasesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Massimo Pinzani
- Institute for Liver and Digestive HealthUniversity College LondonLondonUK
| |
Collapse
|
19
|
Mauch J, Thachil V, Tang WHW. Diagnostics and Prevention: Landscape for Technology Innovation in Precision Cardiovascular Medicine. ADVANCES IN CARDIOVASCULAR TECHNOLOGY 2022:603-624. [DOI: 10.1016/b978-0-12-816861-5.00004-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
20
|
Ghiassian SD, Withers JB, Santolini M, Saleh A, Akmaev VR. RETRACTED: Network-based response module comprised of gene expression biomarkers predicts response to infliximab at treatment initiation in ulcerative colitis. Transl Res 2022; 239:35-43. [PMID: 33965585 DOI: 10.1016/j.trsl.2021.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/12/2021] [Accepted: 04/28/2021] [Indexed: 11/25/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the authors after consulting with the Editors. During a follow-up study, the authors regretfully discovered that the microarray probe-to-gene mapping was incorrect. Although the methodology and primary findings remain the same, the identity of the biomarker genes are incorrect as a result of this honest mistake. The extent of the changes to correct this information necessitated the publication of a corrected version of this article: https://doi.org/10.1016/j.trsl.2022.03.006.
Collapse
Affiliation(s)
| | | | - Marc Santolini
- Center for Research and Interdisciplinarity (CRI), University Paris Descartes, Paris, France
| | - Alif Saleh
- Scipher Medicine Corporation, Waltham, Massachusetts
| | | |
Collapse
|
21
|
Guney E, Athie A. A needle for Alzheimer's in a haystack of claims data. NATURE AGING 2021; 1:1083-1085. [PMID: 37117523 DOI: 10.1038/s43587-021-00139-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- Emre Guney
- Research Programme on Biomedical Informatics, Hospital del Mar Medical Research Institute (IMIM), Pompeu Fabra University (UPF), Barcelona, Spain.
- Discovery and Data Science (DDS) Unit, STALICLA R&D SL, Barcelona, Spain.
| | | |
Collapse
|
22
|
Fang J, Zhang P, Zhou Y, Chiang CW, Tan J, Hou Y, Stauffer S, Li L, Pieper AA, Cummings J, Cheng F. Endophenotype-based in silico network medicine discovery combined with insurance record data mining identifies sildenafil as a candidate drug for Alzheimer's disease. NATURE AGING 2021; 1:1175-1188. [PMID: 35572351 PMCID: PMC9097949 DOI: 10.1038/s43587-021-00138-z] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We developed an endophenotype disease module-based methodology for Alzheimer's disease (AD) drug repurposing and identified sildenafil as a potential disease risk modifier. Based on retrospective case-control pharmacoepidemiologic analyses of insurance claims data for 7.23 million individuals, we found that sildenafil usage was significantly associated with a 69% reduced risk of AD (hazard ratio = 0.31, 95% confidence interval 0.25-0.39, P<1.0×10-8). Propensity score stratified analyses confirmed that sildenafil is significantly associated with a decreased risk of AD across all four drug cohorts we tested (diltiazem, glimepiride, losartan and metformin) after adjusting age, sex, race, and disease comorbidities. We also found that sildenafil increases neurite growth and decreases phospho-tau expression in AD patient-induced pluripotent stem cells-derived neuron models, supporting mechanistically its potential beneficial effect in Alzheimer's disease. The association between sildenafil use and decreased incidence of AD does not establish causality or its direction, which requires a randomized clinical trial approach.
Collapse
Affiliation(s)
- Jiansong Fang
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Pengyue Zhang
- Department of Biostatistics, School of Medicine, Indiana University
| | - Yadi Zhou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Chien-Wei Chiang
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Juan Tan
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Shaun Stauffer
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Andrew A. Pieper
- Harrington Discovery Institute, University Hospital Case Medical Center; Department of Psychiatry, Case Western Reserve University, Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Cleveland, OH 44106, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.,Correspondence to: Feixiong Cheng, Ph.D., Lerner Research Institute, Cleveland Clinic, , Tel: +1-216-4447654; Fax: +1-216-6361609
| |
Collapse
|
23
|
Alcalá-Corona SA, Sandoval-Motta S, Espinal-Enríquez J, Hernández-Lemus E. Modularity in Biological Networks. Front Genet 2021; 12:701331. [PMID: 34594357 PMCID: PMC8477004 DOI: 10.3389/fgene.2021.701331] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/23/2021] [Indexed: 01/13/2023] Open
Abstract
Network modeling, from the ecological to the molecular scale has become an essential tool for studying the structure, dynamics and complex behavior of living systems. Graph representations of the relationships between biological components open up a wide variety of methods for discovering the mechanistic and functional properties of biological systems. Many biological networks are organized into a modular structure, so methods to discover such modules are essential if we are to understand the biological system as a whole. However, most of the methods used in biology to this end, have a limited applicability, as they are very specific to the system they were developed for. Conversely, from the statistical physics and network science perspective, graph modularity has been theoretically studied and several methods of a very general nature have been developed. It is our perspective that in particular for the modularity detection problem, biology and theoretical physics/network science are less connected than they should. The central goal of this review is to provide the necessary background and present the most applicable and pertinent methods for community detection in a way that motivates their further usage in biological research.
Collapse
Affiliation(s)
- Sergio Antonio Alcalá-Corona
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Santiago Sandoval-Motta
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico.,National Council on Science and Technology, Mexico City, Mexico
| | - Jesús Espinal-Enríquez
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
24
|
Kafkas Ş, Althubaiti S, Gkoutos GV, Hoehndorf R, Schofield PN. Linking common human diseases to their phenotypes; development of a resource for human phenomics. J Biomed Semantics 2021; 12:17. [PMID: 34425897 PMCID: PMC8383460 DOI: 10.1186/s13326-021-00249-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/30/2021] [Indexed: 11/11/2022] Open
Abstract
Background In recent years a large volume of clinical genomics data has become available due to rapid advances in sequencing technologies. Efficient exploitation of this genomics data requires linkage to patient phenotype profiles. Current resources providing disease-phenotype associations are not comprehensive, and they often do not have broad coverage of the disease terminologies, particularly ICD-10, which is still the primary terminology used in clinical settings. Methods We developed two approaches to gather disease-phenotype associations. First, we used a text mining method that utilizes semantic relations in phenotype ontologies, and applies statistical methods to extract associations between diseases in ICD-10 and phenotype ontology classes from the literature. Second, we developed a semi-automatic way to collect ICD-10–phenotype associations from existing resources containing known relationships. Results We generated four datasets. Two of them are independent datasets linking diseases to their phenotypes based on text mining and semi-automatic strategies. The remaining two datasets are generated from these datasets and cover a subset of ICD-10 classes of common diseases contained in UK Biobank. We extensively validated our text mined and semi-automatically curated datasets by: comparing them against an expert-curated validation dataset containing disease–phenotype associations, measuring their similarity to disease–phenotype associations found in public databases, and assessing how well they could be used to recover gene–disease associations using phenotype similarity. Conclusion We find that our text mining method can produce phenotype annotations of diseases that are correct but often too general to have significant information content, or too specific to accurately reflect the typical manifestations of the sporadic disease. On the other hand, the datasets generated from integrating multiple knowledgebases are more complete (i.e., cover more of the required phenotype annotations for a given disease). We make all data freely available at 10.5281/zenodo.4726713. Supplementary Information The online version contains supplementary material available at (10.1186/s13326-021-00249-x).
Collapse
Affiliation(s)
- Şenay Kafkas
- Computational Bioscience Research Center (CBRC), Computer, Electrical, and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955, Saudi Arabia
| | - Sara Althubaiti
- Computational Bioscience Research Center (CBRC), Computer, Electrical, and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955, Saudi Arabia
| | - Georgios V Gkoutos
- Health Data Research UK, Midlands site, Edgbaston, Birmingham, B15 2TT, United Kingdom.,Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Robert Hoehndorf
- Computational Bioscience Research Center (CBRC), Computer, Electrical, and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955, Saudi Arabia.
| | - Paul N Schofield
- Department of Physiology, Development & Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| |
Collapse
|
25
|
Abstract
BACKGROUND Systems biology is a rapidly advancing field of science that allows us to look into disease mechanisms, patient diagnosis and stratification, and drug development in a completely new light. It is based on the utilization of unbiased computational systems free of the traditional experimental approaches based on personal choices of what is important and what select experiments should be performed to obtain the expected results. METHODS Systems biology can be applied to inflammatory bowel disease (IBD) by learning basic concepts of omes and omics and how omics-derived "big data" can be integrated to discover the biological networks underlying highly complex diseases like IBD. Once these biological networks (interactomes) are identified, then the molecules controlling the disease network can be singled out and specific blockers developed. RESULTS The field of systems biology in IBD is just emerging, and there is still limited information on how to best utilize its power to advance our understanding of Crohn disease and ulcerative colitis to develop novel therapeutic strategies. Few centers have embraced systems biology in IBD, but the creation of international consortia and large biobanks will make biosamples available to basic and clinical IBD investigators for further research studies. CONCLUSIONS The implementation of systems biology is indispensable and unavoidable, and the patient and medical communities will both benefit immensely from what it will offer in the near future.
Collapse
Affiliation(s)
- Claudio Fiocchi
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | |
Collapse
|
26
|
Shen J, Hou Y, Zhou Y, Mehra R, Jehi L, Cheng F. The Epidemiological and Mechanistic Understanding of the Neurological Manifestations of COVID-19: A Comprehensive Meta-Analysis and a Network Medicine Observation. Front Neurosci 2021; 15:606926. [PMID: 33732102 PMCID: PMC7959722 DOI: 10.3389/fnins.2021.606926] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/01/2021] [Indexed: 12/21/2022] Open
Abstract
The clinical characteristics and biological effects on the nervous system of infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain poorly understood. The aim of this study is to advance epidemiological and mechanistic understanding of the neurological manifestations of coronavirus disease 2019 (COVID-19) using stroke as a case study. In this study, we performed a meta-analysis of clinical studies reporting stroke history, intensive inflammatory response, and procoagulant state C-reactive protein (CRP), Procalcitonin (PCT), and coagulation indicator (D-dimer) in patients with COVID-19. Via network-based analysis of SARS-CoV-2 host genes and stroke-associated genes in the human protein-protein interactome, we inspected the underlying inflammatory mechanisms between COVID-19 and stroke. Finally, we further verified the network-based findings using three RNA-sequencing datasets generated from SARS-CoV-2 infected populations. We found that the overall pooled prevalence of stroke history was 2.98% (95% CI, 1.89-4.68; I 2=69.2%) in the COVID-19 population. Notably, the severe group had a higher prevalence of stroke (6.06%; 95% CI 3.80-9.52; I 2 = 42.6%) compare to the non-severe group (1.1%, 95% CI 0.72-1.71; I 2 = 0.0%). There were increased levels of CRP, PCT, and D-dimer in severe illness, and the pooled mean difference was 40.7 mg/L (95% CI, 24.3-57.1), 0.07 μg/L (95% CI, 0.04-0.10) and 0.63 mg/L (95% CI, 0.28-0.97), respectively. Vascular cell adhesion molecule 1 (VCAM-1), one of the leukocyte adhesion molecules, is suspected to play a vital role of SARS-CoV-2 mediated inflammatory responses. RNA-sequencing data analyses of the SARS-CoV-2 infected patients further revealed the relative importance of inflammatory responses in COVID-19-associated neurological manifestations. In summary, we identified an elevated vulnerability of those with a history of stroke to severe COVID-19 underlying inflammatory responses (i.e., VCAM-1) and procoagulant pathways, suggesting monotonic relationships, thus implicating causality.
Collapse
Affiliation(s)
- Jiayu Shen
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Yadi Zhou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Reena Mehra
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Lara Jehi
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
27
|
Maron BA, Wang RS, Shevtsov S, Drakos SG, Arons E, Wever-Pinzon O, Huggins GS, Samokhin AO, Oldham WM, Aguib Y, Yacoub MH, Rowin EJ, Maron BJ, Maron MS, Loscalzo J. Individualized interactomes for network-based precision medicine in hypertrophic cardiomyopathy with implications for other clinical pathophenotypes. Nat Commun 2021; 12:873. [PMID: 33558530 PMCID: PMC7870822 DOI: 10.1038/s41467-021-21146-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/13/2021] [Indexed: 12/19/2022] Open
Abstract
Progress in precision medicine is limited by insufficient knowledge of transcriptomic or proteomic features in involved tissues that define pathobiological differences between patients. Here, myectomy tissue from patients with obstructive hypertrophic cardiomyopathy and heart failure is analyzed using RNA-Seq, and the results are used to develop individualized protein-protein interaction networks. From this approach, hypertrophic cardiomyopathy is distinguished from dilated cardiomyopathy based on the protein-protein interaction network pattern. Within the hypertrophic cardiomyopathy cohort, the patient-specific networks are variable in complexity, and enriched for 30 endophenotypes. The cardiac Janus kinase 2-Signal Transducer and Activator of Transcription 3-collagen 4A2 (JAK2-STAT3-COL4A2) expression profile informed by the networks was able to discriminate two hypertrophic cardiomyopathy patients with extreme fibrosis phenotypes. Patient-specific network features also associate with other important hypertrophic cardiomyopathy clinical phenotypes. These proof-of-concept findings introduce personalized protein-protein interaction networks (reticulotypes) for characterizing patient-specific pathobiology, thereby offering a direct strategy for advancing precision medicine.
Collapse
Affiliation(s)
- Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Rui-Sheng Wang
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sergei Shevtsov
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Stavros G Drakos
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Elena Arons
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Omar Wever-Pinzon
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Gordon S Huggins
- Hypertrophic Cardiomyopathy Center, Cardiology Division, Tufts Medical Center, Boston, MA, USA
| | - Andriy O Samokhin
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - William M Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yasmine Aguib
- Department of Cardiac Surgery, Imperial College of London, London, UK
- The Magdi Yacoub Heart Center, Aswan, Egypt
| | - Magdi H Yacoub
- Department of Cardiac Surgery, Imperial College of London, London, UK
- The Magdi Yacoub Heart Center, Aswan, Egypt
| | - Ethan J Rowin
- Hypertrophic Cardiomyopathy Center, Cardiology Division, Tufts Medical Center, Boston, MA, USA
| | - Barry J Maron
- Hypertrophic Cardiomyopathy Center, Cardiology Division, Tufts Medical Center, Boston, MA, USA
| | - Martin S Maron
- Hypertrophic Cardiomyopathy Center, Cardiology Division, Tufts Medical Center, Boston, MA, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Paci P, Fiscon G, Conte F, Wang RS, Farina L, Loscalzo J. Gene co-expression in the interactome: moving from correlation toward causation via an integrated approach to disease module discovery. NPJ Syst Biol Appl 2021; 7:3. [PMID: 33479222 PMCID: PMC7819998 DOI: 10.1038/s41540-020-00168-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/19/2020] [Indexed: 01/29/2023] Open
Abstract
In this study, we integrate the outcomes of co-expression network analysis with the human interactome network to predict novel putative disease genes and modules. We first apply the SWItch Miner (SWIM) methodology, which predicts important (switch) genes within the co-expression network that regulate disease state transitions, then map them to the human protein-protein interaction network (PPI, or interactome) to predict novel disease-disease relationships (i.e., a SWIM-informed diseasome). Although the relevance of switch genes to an observed phenotype has been recently assessed, their performance at the system or network level constitutes a new, potentially fascinating territory yet to be explored. Quantifying the interplay between switch genes and human diseases in the interactome network, we found that switch genes associated with specific disorders are closer to each other than to other nodes in the network, and tend to form localized connected subnetworks. These subnetworks overlap between similar diseases and are situated in different neighborhoods for pathologically distinct phenotypes, consistent with the well-known topological proximity property of disease genes. These findings allow us to demonstrate how SWIM-based correlation network analysis can serve as a useful tool for efficient screening of potentially new disease gene associations. When integrated with an interactome-based network analysis, it not only identifies novel candidate disease genes, but also may offer testable hypotheses by which to elucidate the molecular underpinnings of human disease and reveal commonalities between seemingly unrelated diseases.
Collapse
Affiliation(s)
- Paola Paci
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Rome, Italy.
| | - Giulia Fiscon
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
- Fondazione per la Medicina Personalizzata, Via Goffredo Mameli, 3/1 Genova, Italy
| | - Federica Conte
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
| | - Rui-Sheng Wang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lorenzo Farina
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Rome, Italy
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
29
|
Creighton R, Schuch V, Urbanski AH, Giddaluru J, Costa-Martins AG, Nakaya HI. Network vaccinology. Semin Immunol 2020; 50:101420. [PMID: 33162295 DOI: 10.1016/j.smim.2020.101420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/31/2020] [Indexed: 01/21/2023]
Abstract
The structure and function of the immune system is governed by complex networks of interactions between cells and molecular components. Vaccination perturbs these networks, triggering specific pathways to induce cellular and humoral immunity. Systems vaccinology studies have generated vast data sets describing the genes related to vaccination, motivating the use of new approaches to identify patterns within the data. Here, we describe a framework called Network Vaccinology to explore the structure and function of biological networks responsible for vaccine-induced immunity. We demonstrate how the principles of graph theory can be used to identify modules of genes, proteins, and metabolites that are associated with innate and adaptive immune responses. Network vaccinology can be used to assess specific and shared molecular mechanisms of different types of vaccines, adjuvants, and routes of administration to direct rational design of the next generation of vaccines.
Collapse
Affiliation(s)
- Rachel Creighton
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Viviane Schuch
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alysson H Urbanski
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jeevan Giddaluru
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Scientific Platform Pasteur USP, São Paulo, Brazil
| | - Andre G Costa-Martins
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Scientific Platform Pasteur USP, São Paulo, Brazil
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Scientific Platform Pasteur USP, São Paulo, Brazil.
| |
Collapse
|
30
|
Lee LY, Pandey AK, Maron BA, Loscalzo J. Network medicine in Cardiovascular Research. Cardiovasc Res 2020; 117:2186-2202. [PMID: 33165538 DOI: 10.1093/cvr/cvaa321] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/08/2020] [Accepted: 10/30/2020] [Indexed: 12/21/2022] Open
Abstract
The ability to generate multi-omics data coupled with deeply characterizing the clinical phenotype of individual patients promises to improve understanding of complex cardiovascular pathobiology. There remains an important disconnection between the magnitude and granularity of these data and our ability to improve phenotype-genotype correlations for complex cardiovascular diseases. This shortcoming may be due to limitations associated with traditional reductionist analytical methods, which tend to emphasize a single molecular event in the pathogenesis of diseases more aptly characterized by crosstalk between overlapping molecular pathways. Network medicine is a rapidly growing discipline that considers diseases as the consequences of perturbed interactions between multiple interconnected biological components. This powerful integrative approach has enabled a number of important discoveries in complex disease mechanisms. In this review, we introduce the basic concepts of network medicine and highlight specific examples by which this approach has accelerated cardiovascular research. We also review how network medicine is well-positioned to promote rational drug design for patients with cardiovascular diseases, with particular emphasis on advancing precision medicine.
Collapse
Affiliation(s)
- Laurel Y Lee
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Arvind K Pandey
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.,Department of Cardiology, Boston VA Healthcare System, Boston, MA, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
31
|
Castrillon JA, Eng C, Cheng F. Pharmacogenomics for immunotherapy and immune-related cardiotoxicity. Hum Mol Genet 2020; 29:R186-R196. [PMID: 32620943 PMCID: PMC7574958 DOI: 10.1093/hmg/ddaa137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 06/25/2020] [Accepted: 07/01/2020] [Indexed: 12/20/2022] Open
Abstract
Immune checkpoint blockade (ICB) has become a standard of care in a subset of solid tumors. Although cancer survivorship has extended, rates of durable response of ICB remain poor; furthermore, cardiac adverse effects are emerging, which impact several mechanical aspects of the heart. Cardio-oncology programs implement a clinical assessment to curtail cardiovascular disease progression but are limited to the current clinical parameters used in cardiology. Pharmacogenomics provides the potential to unveil heritable and somatic genetic variations for guiding precision immunotherapy treatment to reduce the risk of immune-related cardiotoxicity. A better understanding of pharmacogenomics will optimize the current treatment selection and dosing of immunotherapy. Here, we summarize the recent pharmacogenomics studies in immunotherapy responsiveness and its related cardiotoxicity and highlight how patient genetics and epigenetics can facilitate researchers and clinicians in designing new approaches for precision immunotherapy. We highlight and discuss how single-cell technologies, human-induced pluripotent stem cells and systems pharmacogenomics accelerate future studies of precision cardio-oncology.
Collapse
Affiliation(s)
- Jessica A Castrillon
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
32
|
Wang B, Hu J, Wang Y, Zhang C, Zhou Y, Yu L, Guo X, Gao L, Chen Y. C3: connect separate connected components to form a succinct disease module. BMC Bioinformatics 2020; 21:433. [PMID: 33008305 PMCID: PMC7531168 DOI: 10.1186/s12859-020-03769-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/20/2020] [Indexed: 01/08/2023] Open
Abstract
Background Precise disease module is conducive to understanding the molecular mechanism of disease causation and identifying drug targets. However, due to the fragmentization of disease module in incomplete human interactome, how to determine connectivity pattern and detect a complete neighbourhood of disease based on this is still an open question. Results In this paper, we perform exploratory analysis leading to an important observation that through a few intermediate nodes, most separate connected components formed by disease-associated proteins can be effectively connected and eventually form a complete disease module. And based on the topological properties of these intermediate nodes, we propose a connect separate connected components (C3) method to detect a succinct disease module by introducing a relatively small number of intermediate nodes, which allows us to obtain more pure disease module than other methods. Then we apply C3 across a large corpus of diseases to validate this connectivity pattern of disease module. Furthermore, the connectivity of the perturbed genes in multi-omics data such as The Cancer Genome Atlas also fits this pattern. Conclusions C3 tool is not only useful in detecting a clearly-defined connected disease neighbourhood of 299 diseases and cancer with multi-omics data, but also helpful in better understanding the interconnection of phenotypically related genes in different omics data and studying complex pathological processes.
Collapse
Affiliation(s)
- Bingbo Wang
- School of Computer Science and Technology, Xidian University, Xi'an, People's Republic of China.
| | - Jie Hu
- School of Computer Science and Technology, Xidian University, Xi'an, People's Republic of China
| | - Yajun Wang
- School of Humanities and Foreign Languages, Xi'an University of Technology, Xi'an, People's Republic of China
| | - Chenxing Zhang
- School of Computer Science and Technology, Xidian University, Xi'an, People's Republic of China
| | - Yuanjun Zhou
- School of Computer Science and Technology, Xidian University, Xi'an, People's Republic of China
| | - Liang Yu
- School of Computer Science and Technology, Xidian University, Xi'an, People's Republic of China
| | - Xingli Guo
- School of Computer Science and Technology, Xidian University, Xi'an, People's Republic of China
| | - Lin Gao
- School of Computer Science and Technology, Xidian University, Xi'an, People's Republic of China
| | - Yunru Chen
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China.
| |
Collapse
|
33
|
Casas AI, Nogales C, Mucke HAM, Petraina A, Cuadrado A, Rojo AI, Ghezzi P, Jaquet V, Augsburger F, Dufrasne F, Soubhye J, Deshwal S, Di Sante M, Kaludercic N, Di Lisa F, Schmidt HHHW. On the Clinical Pharmacology of Reactive Oxygen Species. Pharmacol Rev 2020; 72:801-828. [PMID: 32859763 DOI: 10.1124/pr.120.019422] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Reactive oxygen species (ROS) have been correlated with almost every human disease. Yet clinical exploitation of these hypotheses by pharmacological modulation of ROS has been scarce to nonexistent. Are ROS, thus, irrelevant for disease? No. One key misconception in the ROS field has been its consideration as a rather detrimental metabolic by-product of cell metabolism, and thus, any approach eliminating ROS to a certain tolerable level would be beneficial. We now know, instead, that ROS at every concentration, low or high, can serve many essential signaling and metabolic functions. This likely explains why systemic, nonspecific antioxidants have failed in the clinic, often with neutral and sometimes even detrimental outcomes. Recently, drug development has focused, instead, on identifying and selectively modulating ROS enzymatic sources that in a given constellation cause disease while leaving ROS physiologic signaling and metabolic functions intact. As sources, the family of NADPH oxidases stands out as the only enzyme family solely dedicated to ROS formation. Selectively targeting disease-relevant ROS-related proteins is already quite advanced, as evidenced by several phase II/III clinical trials and the first drugs having passed registration. The ROS field is expanding by including target enzymes and maturing to resemble more and more modern, big data-enhanced drug discovery and development, including network pharmacology. By defining a disease based on a distinct mechanism, in this case ROS dysregulation, and not by a symptom or phenotype anymore, ROS pharmacology is leaping forward from a clinical underperformer to a proof of concept within the new era of mechanism-based precision medicine. SIGNIFICANCE STATEMENT: Despite being correlated to almost every human disease, nearly no ROS modulator has been translated to the clinics yet. Here, we move far beyond the old-fashioned misconception of ROS as detrimental metabolic by-products and suggest 1) novel pharmacological targeting focused on selective modulation of ROS enzymatic sources, 2) mechanism-based redefinition of diseases, and 3) network pharmacology within the ROS field, altogether toward the new era of ROS pharmacology in precision medicine.
Collapse
Affiliation(s)
- Ana I Casas
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Cristian Nogales
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Hermann A M Mucke
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Alexandra Petraina
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Antonio Cuadrado
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Ana I Rojo
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Pietro Ghezzi
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Vincent Jaquet
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Fiona Augsburger
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Francois Dufrasne
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Jalal Soubhye
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Soni Deshwal
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Moises Di Sante
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Nina Kaludercic
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Fabio Di Lisa
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalized Medicine, Maastricht University, School of Mental Health and Neuroscience (MHeNS), Maastricht, The Netherlands (A.I.C., C.N., A.P., H.H.H.W.S.); H. M. Pharma Consultancy, Wien, Austria (H.A.M.M.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain (A.C., A.I.R.); Brighton and Sussex Medical School, Falmer, United Kingdom (P.G.); Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland (V.J., F.A.); Microbiology, Bioorganic and Macromolecular Chemistry, RD3, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Bruxelles, Belgium (F.D., J.S.); and Department of Biomedical Sciences (S.D., M.D.S., F.D.L.) and CNR Neuroscience Institute (N.K., F.D.L.), University of Padova, Padova, Italy
| |
Collapse
|
34
|
Song E, Wang R, Leopold JA, Loscalzo J. Network determinants of cardiovascular calcification and repositioned drug treatments. FASEB J 2020; 34:11087-11100. [PMID: 32638415 PMCID: PMC7497212 DOI: 10.1096/fj.202001062r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/03/2020] [Accepted: 06/15/2020] [Indexed: 01/31/2023]
Abstract
Ectopic cardiovascular calcification is a highly prevalent pathology for which there are no effective novel or repurposed pharmacotherapeutics to prevent disease progression. We created a human calcification endophenotype module (ie, the "calcificasome") by mapping vascular calcification genes (proteins) to the human vascular smooth muscle-specific protein-protein interactome (218 nodes and 632 edges, P < 10-5 ). Network proximity analysis was used to demonstrate that the calcificasome overlapped significantly with endophenotype modules governing inflammation, thrombosis, and fibrosis in the human interactome (P < 0.001). A network-based drug repurposing analysis further revealed that everolimus, temsirolimus, and pomalidomide are predicted to target the calcificasome. The efficacy of these agents in limiting calcification was confirmed experimentally by treating human coronary artery smooth muscle cells in an in vitro calcification assay. Each of the drugs affected expression or activity of their predicted target in the network, and decreased calcification significantly (P < 0.009). An integrated network analytical approach identified novel mediators of ectopic cardiovascular calcification and biologically plausible candidate drugs that could be repurposed to target calcification. This methodological framework for drug repurposing has broad applicability to other diseases.
Collapse
Affiliation(s)
- Euijun Song
- Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Rui‐Sheng Wang
- Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Jane A. Leopold
- Division of Cardiovascular MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Joseph Loscalzo
- Department of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
35
|
Leopold JA, Maron BA, Loscalzo J. The application of big data to cardiovascular disease: paths to precision medicine. J Clin Invest 2020; 130:29-38. [PMID: 31895052 DOI: 10.1172/jci129203] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Advanced phenotyping of cardiovascular diseases has evolved with the application of high-resolution omics screening to populations enrolled in large-scale observational and clinical trials. This strategy has revealed that considerable heterogeneity exists at the genotype, endophenotype, and clinical phenotype levels in cardiovascular diseases, a feature of the most common diseases that has not been elucidated by conventional reductionism. In this discussion, we address genomic context and (endo)phenotypic heterogeneity, and examine commonly encountered cardiovascular diseases to illustrate the genotypic underpinnings of (endo)phenotypic diversity. We highlight the existing challenges in cardiovascular disease genotyping and phenotyping that can be addressed by the integration of big data and interpreted using novel analytical methodologies (network analysis). Precision cardiovascular medicine will only be broadly applied to cardiovascular patients once this comprehensive data set is subjected to unique, integrative analytical strategies that accommodate molecular and clinical heterogeneity rather than ignore or reduce it.
Collapse
|
36
|
Maturo MG, Soligo M, Gibson G, Manni L, Nardini C. The greater inflammatory pathway-high clinical potential by innovative predictive, preventive, and personalized medical approach. EPMA J 2020; 11:1-16. [PMID: 32140182 PMCID: PMC7028895 DOI: 10.1007/s13167-019-00195-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND LIMITATIONS Impaired wound healing (WH) and chronic inflammation are hallmarks of non-communicable diseases (NCDs). However, despite WH being a recognized player in NCDs, mainstream therapies focus on (un)targeted damping of the inflammatory response, leaving WH largely unaddressed, owing to three main factors. The first is the complexity of the pathway that links inflammation and wound healing; the second is the dual nature, local and systemic, of WH; and the third is the limited acknowledgement of genetic and contingent causes that disrupt physiologic progression of WH. PROPOSED APPROACH Here, in the frame of Predictive, Preventive, and Personalized Medicine (PPPM), we integrate and revisit current literature to offer a novel systemic view on the cues that can impact on the fate (acute or chronic inflammation) of WH, beyond the compartmentalization of medical disciplines and with the support of advanced computational biology. CONCLUSIONS This shall open to a broader understanding of the causes for WH going awry, offering new operational criteria for patients' stratification (prediction and personalization). While this may also offer improved options for targeted prevention, we will envisage new therapeutic strategies to reboot and/or boost WH, to enable its progression across its physiological phases, the first of which is a transient acute inflammatory response versus the chronic low-grade inflammation characteristic of NCDs.
Collapse
Affiliation(s)
- Maria Giovanna Maturo
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Greg Gibson
- Center for Integrative Genomics, School of Biological Sciences, Georgia Tech, Atlanta, GA USA
| | - Luigi Manni
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Christine Nardini
- IAC Institute for Applied Computing, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
- Bio Unit, Scientific and Medical Direction, SOL Group, Monza, Italy
| |
Collapse
|
37
|
Paci P, Fiscon G, Conte F, Licursi V, Morrow J, Hersh C, Cho M, Castaldi P, Glass K, Silverman EK, Farina L. Integrated transcriptomic correlation network analysis identifies COPD molecular determinants. Sci Rep 2020; 10:3361. [PMID: 32099002 PMCID: PMC7042269 DOI: 10.1038/s41598-020-60228-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/23/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex and heterogeneous syndrome. Network-based analysis implemented by SWIM software can be exploited to identify key molecular switches - called "switch genes" - for the disease. Genes contributing to common biological processes or defining given cell types are usually co-regulated and co-expressed, forming expression network modules. Consistently, we found that the COPD correlation network built by SWIM consists of three well-characterized modules: one populated by switch genes, all up-regulated in COPD cases and related to the regulation of immune response, inflammatory response, and hypoxia (like TIMP1, HIF1A, SYK, LY96, BLNK and PRDX4); one populated by well-recognized immune signature genes, all up-regulated in COPD cases; one where the GWAS genes AGER and CAVIN1 are the most representative module genes, both down-regulated in COPD cases. Interestingly, 70% of AGER negative interactors are switch genes including PRDX4, whose activation strongly correlates with the activation of known COPD GWAS interactors SERPINE2, CD79A, and POUF2AF1. These results suggest that SWIM analysis can identify key network modules related to complex diseases like COPD.
Collapse
Affiliation(s)
- Paola Paci
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy.
| | - Giulia Fiscon
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
| | - Federica Conte
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
| | - Valerio Licursi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Jarrett Morrow
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Craig Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Lorenzo Farina
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
38
|
Benincasa G, Marfella R, Della Mura N, Schiano C, Napoli C. Strengths and Opportunities of Network Medicine in Cardiovascular Diseases. Circ J 2020; 84:144-152. [DOI: 10.1253/circj.cj-19-0879] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Giuditta Benincasa
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania “Luigi Vanvitelli”
| | - Raffaele Marfella
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania “Luigi Vanvitelli”
| | | | - Concetta Schiano
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania “Luigi Vanvitelli”
| | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania “Luigi Vanvitelli”
- IRCCS-SDN
| |
Collapse
|
39
|
Li W, Deng G, Zhang J, Hu E, He Y, Lv J, Sun X, Wang K, Chen L. Identification of breast cancer risk modules via an integrated strategy. Aging (Albany NY) 2019; 11:12131-12146. [PMID: 31860871 PMCID: PMC6949069 DOI: 10.18632/aging.102546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/19/2019] [Indexed: 12/17/2022]
Abstract
Breast cancer is one of the most common malignant cancers among females worldwide. This complex disease is not caused by a single gene, but resulted from multi-gene interactions, which could be represented by biological networks. Network modules are composed of genes with significant similarities in terms of expression, function and disease association. Therefore, the identification of disease risk modules could contribute to understanding the molecular mechanisms underlying breast cancer. In this paper, an integrated disease risk module identification strategy was proposed according to a multi-objective programming model for two similarity criteria as well as significance of permutation tests in Markov random field module score, function consistency score and Pearson correlation coefficient difference score. Three breast cancer risk modules were identified from a breast cancer-related interaction network. Genes in these risk modules were confirmed to play critical roles in breast cancer by literature review. These risk modules were enriched in breast cancer-related pathways or functions and could distinguish between breast tumor and normal samples with high accuracy for not only the microarray dataset used for breast cancer risk module identification, but also another two independent datasets. Our integrated strategy could be extended to other complex diseases to identify their risk modules and reveal their pathogenesis.
Collapse
Affiliation(s)
- Wan Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Gui Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ji Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Erqiang Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yuehan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Junjie Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xilin Sun
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, the Fourth Hospital of Harbin Medical University, Harbin, China
| | - Kai Wang
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, the Fourth Hospital of Harbin Medical University, Harbin, China
| | - Lina Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
40
|
Biologically Defined or Biologically Informed Traits Are More Heritable Than Clinically Defined Ones: The Case of Oral and Dental Phenotypes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1197:179-189. [PMID: 31732942 DOI: 10.1007/978-3-030-28524-1_13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The genetic basis of oral health has long been theorized, but little information exists on the heritable variance in common oral and dental disease traits explained by the human genome. We sought to add to the evidence base of heritability of oral and dental traits using high-density genotype data in a well-characterized community-based cohort of middle-age adults. We used genome-wide association (GWAS) data combined with clinical and biomarker information in the Dental Atherosclerosis Risk In Communities (ARIC) cohort. Genotypes comprised SNPs directly typed on the Affymetrix Genome-Wide Human SNP Array 6.0 chip with minor allele frequency of >5% (n = 656,292) or were imputed using HapMap II-CEU (n = 2,104,905). We investigated 30 traits including "global" [e.g., number of natural teeth (NT) and incident tooth loss], clinically defined (e.g., dental caries via the DMFS index, periodontitis via the CDC/AAP and WW17 classifications), and biologically informed (e.g., subgingival pathogen colonization and "complex" traits). Heritability (i.e., variance explained; h2) was calculated using Visscher's Genome-wide Complex Trait Analysis (GCTA), using a random-effects mixed linear model and restricted maximum likelihood (REML) regression adjusting for ancestry (10 principal components), age, and sex. Heritability estimates were modest for clinical traits-NT = 0.11 (se = 0.07), severe chronic periodontitis (CDC/AAP) = 0.22 (se = 0.19), WW17 Stage 4 vs. 1/2 = 0.15 (se = 0.11). "High gingival index" and "high red complex colonization" had h2 > 0.50, while a periodontal complex trait defined by high IL-1β GCF expression and Aggregatibacter actinomycetemcomitans subgingival colonization had the highest h2 = 0.72 (se = 0.32). Our results indicate that all GWAS SNPs explain modest levels of the observed variance in clinical oral and dental measures. Subgingival bacterial colonization and complex phenotypes encompassing both bacterial colonization and local inflammatory response had the highest heritability, suggesting that these biologically informed traits capture aspects of the disease process and are promising targets for genomics investigations, according to the notion of precision oral health.
Collapse
|
41
|
Aguilar D, Lemonnier N, Koppelman GH, Melén E, Oliva B, Pinart M, Guerra S, Bousquet J, Anto JM. Understanding allergic multimorbidity within the non-eosinophilic interactome. PLoS One 2019; 14:e0224448. [PMID: 31693680 PMCID: PMC6834334 DOI: 10.1371/journal.pone.0224448] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The mechanisms explaining multimorbidity between asthma, dermatitis and rhinitis (allergic multimorbidity) are not well known. We investigated these mechanisms and their specificity in distinct cell types by means of an interactome-based analysis of expression data. METHODS Genes associated to the diseases were identified using data mining approaches, and their multimorbidity mechanisms in distinct cell types were characterized by means of an in silico analysis of the topology of the human interactome. RESULTS We characterized specific pathomechanisms for multimorbidities between asthma, dermatitis and rhinitis for distinct emergent non-eosinophilic cell types. We observed differential roles for cytokine signaling, TLR-mediated signaling and metabolic pathways for multimorbidities across distinct cell types. Furthermore, we also identified individual genes potentially associated to multimorbidity mechanisms. CONCLUSIONS Our results support the existence of differentiated multimorbidity mechanisms between asthma, dermatitis and rhinitis at cell type level, as well as mechanisms common to distinct cell types. These results will help understanding the biology underlying allergic multimorbidity, assisting in the design of new clinical studies.
Collapse
MESH Headings
- Asthma/epidemiology
- Asthma/genetics
- Asthma/immunology
- Blood Cells/immunology
- Blood Cells/metabolism
- Cytokines/immunology
- Cytokines/metabolism
- Datasets as Topic
- Dermatitis, Allergic Contact/epidemiology
- Dermatitis, Allergic Contact/genetics
- Dermatitis, Allergic Contact/immunology
- Dermatitis, Atopic/epidemiology
- Dermatitis, Atopic/genetics
- Dermatitis, Atopic/immunology
- Gene Expression Profiling
- Humans
- Immunity, Cellular/genetics
- Multimorbidity
- Protein Interaction Maps/genetics
- Protein Interaction Maps/immunology
- Rhinitis, Allergic/epidemiology
- Rhinitis, Allergic/genetics
- Rhinitis, Allergic/immunology
Collapse
Affiliation(s)
- Daniel Aguilar
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD), Instituto de Salud Carlos III, Barcelona, Spain
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
- 6AM Data Mining, Barcelona, Spain
| | - Nathanael Lemonnier
- Institute for Advanced Biosciences, Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes, Site Santé, Allée des Alpes, La Tronche, France
| | - Gerard H. Koppelman
- University of Groningen, University Medical Center Groningen, Beatrix Children’s Hospital, Department of Pediatric Pulmonology and Pediatric Allergology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Baldo Oliva
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Mariona Pinart
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| | - Stefano Guerra
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, United States of America
| | - Jean Bousquet
- Hopital Arnaud de Villeneuve University Hospital, Montpellier, France
- Charité, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Comprehensive Allergy Center, Department of Dermatology and Allergy, Berlin, Germany
| | - Josep M. Anto
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| |
Collapse
|
42
|
Schlotter F, Halu A, Goto S, Blaser MC, Body SC, Lee LH, Higashi H, DeLaughter DM, Hutcheson JD, Vyas P, Pham T, Rogers MA, Sharma A, Seidman CE, Loscalzo J, Seidman JG, Aikawa M, Singh SA, Aikawa E. Spatiotemporal Multi-Omics Mapping Generates a Molecular Atlas of the Aortic Valve and Reveals Networks Driving Disease. Circulation 2019; 138:377-393. [PMID: 29588317 DOI: 10.1161/circulationaha.117.032291] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND No pharmacological therapy exists for calcific aortic valve disease (CAVD), which confers a dismal prognosis without invasive valve replacement. The search for therapeutics and early diagnostics is challenging because CAVD presents in multiple pathological stages. Moreover, it occurs in the context of a complex, multi-layered tissue architecture; a rich and abundant extracellular matrix phenotype; and a unique, highly plastic, and multipotent resident cell population. METHODS A total of 25 human stenotic aortic valves obtained from valve replacement surgeries were analyzed by multiple modalities, including transcriptomics and global unlabeled and label-based tandem-mass-tagged proteomics. Segmentation of valves into disease stage-specific samples was guided by near-infrared molecular imaging, and anatomic layer-specificity was facilitated by laser capture microdissection. Side-specific cell cultures were subjected to multiple calcifying stimuli, and their calcification potential and basal/stimulated proteomes were evaluated. Molecular (protein-protein) interaction networks were built, and their central proteins and disease associations were identified. RESULTS Global transcriptional and protein expression signatures differed between the nondiseased, fibrotic, and calcific stages of CAVD. Anatomic aortic valve microlayers exhibited unique proteome profiles that were maintained throughout disease progression and identified glial fibrillary acidic protein as a specific marker of valvular interstitial cells from the spongiosa layer. CAVD disease progression was marked by an emergence of smooth muscle cell activation, inflammation, and calcification-related pathways. Proteins overrepresented in the disease-prone fibrosa are functionally annotated to fibrosis and calcification pathways, and we found that in vitro, fibrosa-derived valvular interstitial cells demonstrated greater calcification potential than those from the ventricularis. These studies confirmed that the microlayer-specific proteome was preserved in cultured valvular interstitial cells, and that valvular interstitial cells exposed to alkaline phosphatase-dependent and alkaline phosphatase-independent calcifying stimuli had distinct proteome profiles, both of which overlapped with that of the whole tissue. Analysis of protein-protein interaction networks found a significant closeness to multiple inflammatory and fibrotic diseases. CONCLUSIONS A spatially and temporally resolved multi-omics, and network and systems biology strategy identifies the first molecular regulatory networks in CAVD, a cardiac condition without a pharmacological cure, and describes a novel means of systematic disease ontology that is broadly applicable to comprehensive omics studies of cardiovascular diseases.
Collapse
Affiliation(s)
- Florian Schlotter
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Arda Halu
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.).,Channing Division of Network Medicine (A.H., A.S., M.A.)
| | - Shinji Goto
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Simon C Body
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA. Center for Perioperative Genomics and Department of Anesthesiology, Brigham and Women's Hospital, Boston, MA (S.C.B.)
| | - Lang H Lee
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Hideyuki Higashi
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Daniel M DeLaughter
- Department of Genetics, Harvard Medical School, Boston, MA (D.M.D., C.E.S., J.G.S.)
| | - Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.).,Department of Biomedical Engineering, Florida International University, Miami (J.D.H.)
| | - Payal Vyas
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Tan Pham
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Maximillian A Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Amitabh Sharma
- Channing Division of Network Medicine (A.H., A.S., M.A.)
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (D.M.D., C.E.S., J.G.S.).,Department of Medicine, Brigham and Women's Hospital, Boston, MA (C.E.S., J.L.).,Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Boston, MA (C.E.S., J.L.)
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (D.M.D., C.E.S., J.G.S.)
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.).,Channing Division of Network Medicine (A.H., A.S., M.A.).,Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.A., E.A.)
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.).,Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.A., E.A.)
| |
Collapse
|
43
|
Haider S, Ponnusamy K, Singh RKB, Chakraborti A, Bamezai RNK. Hamiltonian energy as an efficient approach to identify the significant key regulators in biological networks. PLoS One 2019; 14:e0221463. [PMID: 31442253 PMCID: PMC6707611 DOI: 10.1371/journal.pone.0221463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/07/2019] [Indexed: 12/27/2022] Open
Abstract
The topological characteristics of biological networks enable us to identify the key nodes in terms of modularity. However, due to a large size of the biological networks with many hubs and functional modules across intertwined layers within the network, it often becomes difficult to accomplish the task of identifying potential key regulators. We use for the first time a generalized formalism of Hamiltonian Energy (HE) with a recursive approach. The concept, when applied to the Apoptosis Regulatory Gene Network (ARGN), helped us identify 11 Motif hubs (MHs), which influenced the network up to motif levels. The approach adopted allowed to classify MHs into 5 significant motif hubs (S-MHs) and 6 non-significant motif hubs (NS-MHs). The significant motif hubs had a higher HE value and were considered as high-active key regulators; while the non-significant motif hubs had a relatively lower HE value and were considered as low-active key regulators, in network control mechanism. Further, we compared the results of the HE analyses with the topological characterization, after subjecting to the three conditions independently: (i) removing all MHs, (ii) removing only S-MHs, and (iii) removing only NS-MHs from the ARGN. This procedure allowed us to cross-validate the role of 5 S-MHs, NFk-B1, BRCA1, CEBPB, AR, and POU2F1 as the potential key regulators. The changes in HE calculations further showed that the removal of 5 S-MHs could cause perturbation at all levels of the network, a feature not discernible by topological analysis alone.
Collapse
Affiliation(s)
- Shazia Haider
- Department of Neurology, All India Institute of Medical Science (AIIMS), New Delhi, India
| | | | - R. K. Brojen Singh
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
- * E-mail: (RKBS); (AC); (RNKB)
| | - Anirban Chakraborti
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
- * E-mail: (RKBS); (AC); (RNKB)
| | - Rameshwar N. K. Bamezai
- Formerly at National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
- * E-mail: (RKBS); (AC); (RNKB)
| |
Collapse
|
44
|
Naviaux RK, Naviaux JC, Li K, Wang L, Monk JM, Bright AT, Koslik HJ, Ritchie JB, Golomb BA. Metabolic features of Gulf War illness. PLoS One 2019; 14:e0219531. [PMID: 31348786 PMCID: PMC6660083 DOI: 10.1371/journal.pone.0219531] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 06/27/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND More than 230,000 veterans-about 1/3 of US personnel deployed in the 1990-1991 Persian Gulf War-developed chronic, multi-symptom health problems now called "Gulf War illness" (GWI), for which mechanisms and objective diagnostic signatures continue to be sought. METHODS Targeted, broad-spectrum serum metabolomics was used to gain insights into the biology of GWI. 40 male participants, included 20 veterans who met both Kansas and CDC diagnostic criteria for GWI and 20 nonveteran controls without similar symptoms that were 1:1 matched to GWI cases by age, sex, and ethnicity. Serum samples were collected and archived at -80° C prior to testing. 358 metabolites from 46 biochemical pathways were measured by hydrophilic interaction liquid chromatography and tandem mass spectrometry. RESULTS Veterans with GWI, compared to healthy controls, had abnormalities in 8 of 46 biochemical pathways interrogated. Lipid abnormalities accounted for 78% of the metabolic impact. Fifteen ceramides and sphingomyelins, and four phosphatidylcholine lipids were increased. Five of the 8 pathways were shared with the previously reported metabolic phenotype of males with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). However, 4 of the 5 shared pathways were regulated in opposite directions; key pathways that were up-regulated in GWI were down-regulated in ME/CFS. The single pathway regulated in the same direction was purines, which were decreased. CONCLUSIONS Our data show that despite heterogeneous exposure histories, a metabolic phenotype of GWI was clearly distinguished from controls. Metabolomic differences between GWI and ME/CFS show that common clinical symptoms like fatigue can have different chemical mechanisms and different diagnostic implications. Larger studies will be needed to validate these findings.
Collapse
Affiliation(s)
- Robert K. Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Medicine, Division of Medical Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Pediatrics, Division of Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Pathology, Division of Comparative Pathology, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Jane C. Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Neurosciences, Division of Pediatric Neurology, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Medicine, Division of Medical Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Medicine, Division of Medical Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Jonathan M. Monk
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Medicine, Division of Medical Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - A. Taylor Bright
- The Mitochondrial and Metabolic Disease Center, University of California San Diego School of Medicine, San Diego, California, United States of America
- Department of Medicine, Division of Medical Genetics, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Hayley J. Koslik
- Department of Medicine, Division of General Internal Medicine, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Janis B. Ritchie
- Department of Medicine, Division of General Internal Medicine, University of California San Diego School of Medicine, San Diego, California, United States of America
| | - Beatrice A. Golomb
- Department of Medicine, Division of General Internal Medicine, University of California San Diego School of Medicine, San Diego, California, United States of America
| |
Collapse
|
45
|
Lee LYH, Loscalzo J. Network Medicine in Pathobiology. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1311-1326. [PMID: 31014954 DOI: 10.1016/j.ajpath.2019.03.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/05/2019] [Indexed: 12/11/2022]
Abstract
The past decade has witnessed exponential growth in the generation of high-throughput human data across almost all known dimensions of biological systems. The discipline of network medicine has rapidly evolved in parallel, providing an unbiased, comprehensive biological framework through which to interrogate and integrate systematically these large-scale, multi-omic data to enhance our understanding of disease mechanisms and to design drugs that reflect a deep knowledge of molecular pathobiology. In this review, we discuss the key principles of network medicine and the human disease network and explore the latest applications of network medicine in this multi-omic era. We also highlight the current conceptual and technological challenges, which serve as exciting opportunities by which to improve and expand the network-based applications beyond the artificial boundaries of the current state of human pathobiology.
Collapse
Affiliation(s)
| | - Joseph Loscalzo
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
46
|
Sonawane AR, Weiss ST, Glass K, Sharma A. Network Medicine in the Age of Biomedical Big Data. Front Genet 2019; 10:294. [PMID: 31031797 PMCID: PMC6470635 DOI: 10.3389/fgene.2019.00294] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
Network medicine is an emerging area of research dealing with molecular and genetic interactions, network biomarkers of disease, and therapeutic target discovery. Large-scale biomedical data generation offers a unique opportunity to assess the effect and impact of cellular heterogeneity and environmental perturbations on the observed phenotype. Marrying the two, network medicine with biomedical data provides a framework to build meaningful models and extract impactful results at a network level. In this review, we survey existing network types and biomedical data sources. More importantly, we delve into ways in which the network medicine approach, aided by phenotype-specific biomedical data, can be gainfully applied. We provide three paradigms, mainly dealing with three major biological network archetypes: protein-protein interaction, expression-based, and gene regulatory networks. For each of these paradigms, we discuss a broad overview of philosophies under which various network methods work. We also provide a few examples in each paradigm as a test case of its successful application. Finally, we delineate several opportunities and challenges in the field of network medicine. We hope this review provides a lexicon for researchers from biological sciences and network theory to come on the same page to work on research areas that require interdisciplinary expertise. Taken together, the understanding gained from combining biomedical data with networks can be useful for characterizing disease etiologies and identifying therapeutic targets, which, in turn, will lead to better preventive medicine with translational impact on personalized healthcare.
Collapse
Affiliation(s)
- Abhijeet R. Sonawane
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Amitabh Sharma
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA, United States
| |
Collapse
|
47
|
Baulina N, Osmak G, Kiselev I, Popova E, Boyko A, Kulakova O, Favorova O. MiRNAs from DLK1-DIO3 Imprinted Locus at 14q32 are Associated with Multiple Sclerosis: Gender-Specific Expression and Regulation of Receptor Tyrosine Kinases Signaling. Cells 2019; 8:cells8020133. [PMID: 30743997 PMCID: PMC6406543 DOI: 10.3390/cells8020133] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/01/2019] [Accepted: 02/07/2019] [Indexed: 02/07/2023] Open
Abstract
Relapsing-remitting multiple sclerosis (RRMS) is the most prevalent course of multiple sclerosis. It is an autoimmune inflammatory disease of the central nervous system. To investigate the gender-specific involvement of microRNAs (miRNAs) in RRMS pathogenesis, we compared miRNA profiles in peripheral blood mononuclear cells separately in men and women (eight RRMS patients versus four healthy controls of each gender) using high-throughput sequencing. In contrast to women, six downregulated and 26 upregulated miRNAs (padj < 0.05) were identified in men with RRMS. Genes encoding upregulated miRNAs are co-localized in DLK1-DIO3 imprinted locus on human chromosome 14q32. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) analysis was performed in independent groups of men (16 RRMS patients and 10 healthy controls) and women (20 RRMS patients and 10 healthy controls). Increased expression of miR-431, miR-127-3p, miR-379, miR-376c, miR-381, miR-410 and miR-656 was again demonstrated in male (padj < 0.05), but not in female RRMS patients. At the same time, the expression levels of these miRNAs were lower in healthy men than in healthy women, whereas in RRMS men they increased and reached or exceeded levels in RRMS women. In general, we demonstrated that expression levels of these miRNAs depend both on “health–disease” status and gender. Network-based enrichment analysis identified that receptor tyrosine kinases-activated pathways were enriched with products of genes targeted by miRNAs from DLK1-DIO3 locus. These results suggest the male-specific involvement of these miRNAs in RRMS pathogenesis via regulation of PI3K/Akt signaling.
Collapse
Affiliation(s)
- Natalia Baulina
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia.
| | - German Osmak
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia.
| | - Ivan Kiselev
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia.
| | - Ekaterina Popova
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia.
| | - Alexey Boyko
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia.
| | - Olga Kulakova
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia.
| | - Olga Favorova
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia.
| |
Collapse
|
48
|
A network biology approach to unraveling inherited axonopathies. Sci Rep 2019; 9:1692. [PMID: 30737464 PMCID: PMC6368620 DOI: 10.1038/s41598-018-37119-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/23/2018] [Indexed: 12/14/2022] Open
Abstract
Inherited axonopathies represent a spectrum of disorders unified by the common pathological mechanism of length-dependent axonal degeneration. Progressive axonal degeneration can lead to both Charcot-Marie-Tooth type 2 (CMT2) and Hereditary Spastic Paraplegia (HSP) depending on the affected neurons: peripheral motor and sensory nerves or central nervous system axons of the corticospinal tract and dorsal columns, respectively. Inherited axonopathies display an extreme degree of genetic heterogeneity of Mendelian high-penetrance genes. High locus heterogeneity is potentially advantageous to deciphering disease etiology by providing avenues to explore biological pathways in an unbiased fashion. Here, we investigate ‘gene modules’ in inherited axonopathies through a network-based analysis of the Human Integrated Protein-Protein Interaction rEference (HIPPIE) database. We demonstrate that CMT2 and HSP disease proteins are significantly more connected than randomly expected. We define these connected disease proteins as ‘proto-modules’ and show the topological relationship of these proto-modules by evaluating their overlap through a shortest-path based measurement. In particular, we observe that the CMT2 and HSP proto-modules significantly overlapped, demonstrating a shared genetic etiology. Comparison of both modules with other diseases revealed an overlapping relationship between HSP and hereditary ataxia and between CMT2 + HSP and hereditary ataxia. We then use the DIseAse Module Detection (DIAMOnD) algorithm to expand the proto-modules into comprehensive disease modules. Analysis of disease modules thus obtained reveals an enrichment of ribosomal proteins and pathways likely central to inherited axonopathy pathogenesis, including protein processing in the endoplasmic reticulum, spliceosome, and mRNA processing. Furthermore, we determine pathways specific to each axonopathy by analyzing the difference of the axonopathy modules. CMT2-specific pathways include glycolysis and gluconeogenesis-related processes, while HSP-specific pathways include processes involved in viral infection response. Unbiased characterization of inherited axonopathy disease modules will provide novel candidate disease genes, improve interpretation of candidate genes identified through patient data, and guide therapy development.
Collapse
|
49
|
Osmak GJ, Matveeva NA, Titov BV, Favorova OO. The Myocardial Infarction Associated Variant in the MIR196A2 Gene and Presumable Signaling Pathways to Involve miR-196a2 in the Pathological Phenotype. Mol Biol 2018. [DOI: 10.1134/s0026893318060146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
50
|
NGS-identified circulating miR-375 as a potential regulating component of myocardial infarction associated network. J Mol Cell Cardiol 2018; 121:173-179. [PMID: 30025897 DOI: 10.1016/j.yjmcc.2018.07.129] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/13/2018] [Accepted: 07/14/2018] [Indexed: 12/14/2022]
Abstract
Acute myocardial infarction (MI), the most severe type of coronary heart disease, is a leading cause of disability and mortality worldwide. In order to investigate the involvement of miRNAs in the pathologic processes related to MI, we performed the analysis of circulating miRNAs - stable short noncoding RNA molecules - in the peripheral blood plasma of MI patients compared to healthy controls (all persons were men and lived in European Russia) using next generation sequencing. We observed 20 miRNAs, which levels in plasma more than two-fold differed in MI patients (p < 0.05). Among them miR-208b and miR-375 passed threshold for multiple corrections (FC = 49.2, FDR-adjusted p-value = 0.0078 and FC = -6.4, FDR-adjusted p-value = 0.00076, respectively); these data were then validated using RT-qPCR (FC = 5.3, p-value = 0.028 and FC = -2.1, p-value = 0.0039, respectively). While for miR-208b we reidentified earlier observations, miR-375 was found to be associated with MI for the first time. To investigate the reasons for which miR-375 holds a special place among circulating miRNAs in MI, enrichment and network analyses of miR-375 target genes and their interactions were carried out. PIK3CA and TP53 genes, regulated by miR-375, were identified as the key players of MI disease module.
Collapse
|