1
|
Abdelrady YA, Thabet HS, Sayed AM. The future of metronomic chemotherapy: experimental and computational approaches of drug repurposing. Pharmacol Rep 2024:10.1007/s43440-024-00662-w. [PMID: 39432183 DOI: 10.1007/s43440-024-00662-w] [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: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
Metronomic chemotherapy (MC), long-term continuous administration of anticancer drugs, is gaining attention as an alternative to the traditional maximum tolerated dose (MTD) chemotherapy. By combining MC with other treatments, the therapeutic efficacy is enhanced while minimizing toxicity. MC employs multiple mechanisms, making it a versatile approach against various cancers. However, drug resistance limits the long-term effectiveness of MC, necessitating ongoing development of anticancer drugs. Traditional drug discovery is lengthy and costly due to processes like target protein identification, virtual screening, lead optimization, and safety and efficacy evaluations. Drug repurposing (DR), which screens FDA-approved drugs for new uses, is emerging as a cost-effective alternative. Both experimental and computational methods, such as protein binding assays, in vitro cytotoxicity tests, structure-based screening, and several types of association analyses (Similarity-Based, Network-Based, and Target Gene), along with retrospective clinical analyses, are employed for virtual screening. This review covers the mechanisms of MC, its application in various cancers, DR strategies, examples of repurposed drugs, and the associated challenges and future directions.
Collapse
Affiliation(s)
- Yousef A Abdelrady
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104, Freiburg, Germany
| | - Hayam S Thabet
- Microbiology Department, Faculty of Veterinary Medicine, Assiut University, Asyut, 71526, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Asyut, 71516, Egypt
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Kingdom of Saudi Arabia
| |
Collapse
|
2
|
Woodward DJ, Thorp JG, Middeldorp CM, Akóṣílè W, Derks EM, Gerring ZF. Leveraging pleiotropy for the improved treatment of psychiatric disorders. Mol Psychiatry 2024:10.1038/s41380-024-02771-7. [PMID: 39390223 DOI: 10.1038/s41380-024-02771-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024]
Abstract
Over 90% of drug candidates fail in clinical trials, while it takes 10-15 years and one billion US dollars to develop a single successful drug. Drug development is more challenging for psychiatric disorders, where disease comorbidity and complex symptom profiles obscure the identification of causal mechanisms for therapeutic intervention. One promising approach for determining more suitable drug candidates in clinical trials is integrating human genetic data into the selection process. Genome-wide association studies have identified thousands of replicable risk loci for psychiatric disorders, and sophisticated statistical tools are increasingly effective at using these data to pinpoint likely causal genes. These studies have also uncovered shared or pleiotropic genetic risk factors underlying comorbid psychiatric disorders. In this article, we argue that leveraging pleiotropic effects will provide opportunities to discover novel drug targets and identify more effective treatments for psychiatric disorders by targeting a common mechanism rather than treating each disease separately.
Collapse
Affiliation(s)
- Damian J Woodward
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- School of Biomedical Science, Queensland University of Technology, Brisbane, QLD, Australia.
| | - Jackson G Thorp
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Christel M Middeldorp
- Department of Child and Adolescent Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction and Development Research Institute, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Arkin Mental Health Care, Amsterdam, The Netherlands
- Levvel, Academic Center for Child and Adolescent Psychiatry, Amsterdam, The Netherlands
- Child Health Research Centre, University of Queensland, Brisbane, QLD, Australia
- Child and Youth Mental Health Service, Children's Health Queensland Hospital and Health Service, Brisbane, QLD, Australia
| | - Wọlé Akóṣílè
- Greater Brisbane Clinical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Eske M Derks
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Zachary F Gerring
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- Healthy Development and Ageing, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
| |
Collapse
|
3
|
Fan M, Jin C, Li D, Deng Y, Yao L, Chen Y, Ma YL, Wang T. Multi-level advances in databases related to systems pharmacology in traditional Chinese medicine: a 60-year review. Front Pharmacol 2023; 14:1289901. [PMID: 38035021 PMCID: PMC10682728 DOI: 10.3389/fphar.2023.1289901] [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] [Received: 09/06/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023] Open
Abstract
The therapeutic effects of traditional Chinese medicine (TCM) involve intricate interactions among multiple components and targets. Currently, computational approaches play a pivotal role in simulating various pharmacological processes of TCM. The application of network analysis in TCM research has provided an effective means to explain the pharmacological mechanisms underlying the actions of herbs or formulas through the lens of biological network analysis. Along with the advances of network analysis, computational science has coalesced around the core chain of TCM research: formula-herb-component-target-phenotype-ZHENG, facilitating the accumulation and organization of the extensive TCM-related data and the establishment of relevant databases. Nonetheless, recent years have witnessed a tendency toward homogeneity in the development and application of these databases. Advancements in computational technologies, including deep learning and foundation model, have propelled the exploration and modeling of intricate systems into a new phase, potentially heralding a new era. This review aims to delves into the progress made in databases related to six key entities: formula, herb, component, target, phenotype, and ZHENG. Systematically discussions on the commonalities and disparities among various database types were presented. In addition, the review raised the issue of research bottleneck in TCM computational pharmacology and envisions the forthcoming directions of computational research within the realm of TCM.
Collapse
Affiliation(s)
- Mengyue Fan
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ching Jin
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL, United States
| | - Daping Li
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yingshan Deng
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Yao
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongjun Chen
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu-Ling Ma
- Oxford Chinese Medicine Research Centre, University of Oxford, Oxford, United Kingdom
| | - Taiyi Wang
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Oxford Chinese Medicine Research Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
4
|
Truong TTT, Panizzutti B, Kim JH, Walder K. Repurposing Drugs via Network Analysis: Opportunities for Psychiatric Disorders. Pharmaceutics 2022; 14:1464. [PMID: 35890359 PMCID: PMC9319329 DOI: 10.3390/pharmaceutics14071464] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/30/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
Despite advances in pharmacology and neuroscience, the path to new medications for psychiatric disorders largely remains stagnated. Drug repurposing offers a more efficient pathway compared with de novo drug discovery with lower cost and less risk. Various computational approaches have been applied to mine the vast amount of biomedical data generated over recent decades. Among these methods, network-based drug repurposing stands out as a potent tool for the comprehension of multiple domains of knowledge considering the interactions or associations of various factors. Aligned well with the poly-pharmacology paradigm shift in drug discovery, network-based approaches offer great opportunities to discover repurposing candidates for complex psychiatric disorders. In this review, we present the potential of network-based drug repurposing in psychiatry focusing on the incentives for using network-centric repurposing, major network-based repurposing strategies and data resources, applications in psychiatry and challenges of network-based drug repurposing. This review aims to provide readers with an update on network-based drug repurposing in psychiatry. We expect the repurposing approach to become a pivotal tool in the coming years to battle debilitating psychiatric disorders.
Collapse
Affiliation(s)
- Trang T. T. Truong
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong 3220, Australia; (T.T.T.T.); (B.P.); (J.H.K.)
| | - Bruna Panizzutti
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong 3220, Australia; (T.T.T.T.); (B.P.); (J.H.K.)
| | - Jee Hyun Kim
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong 3220, Australia; (T.T.T.T.); (B.P.); (J.H.K.)
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville 3010, Australia
| | - Ken Walder
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong 3220, Australia; (T.T.T.T.); (B.P.); (J.H.K.)
| |
Collapse
|
5
|
Gilanchi S, Zali H, Faranoush M, Rezaei Tavirani M, Shahriary K, Daskareh M. Identification of Candidate Biomarkers for Idiopathic Thrombocytopenic Purpura by Bioinformatics Analysis of Microarray Data. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 19:275-289. [PMID: 33841542 PMCID: PMC8019887 DOI: 10.22037/ijpr.2020.113442.14305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Idiopathic Thrombocytopenic Purpura (ITP) is a multifactorial disease with decreased count of platelet that can lead to bruising and bleeding manifestations. This study was intended to identify critical genes associated with chronic ITP. The gene expression profile GSE46922 was downloaded from the Gene Expression Omnibus database to recognize Differentially Expressed Genes (DEGs) by R software. Gene ontology and pathway analyses were performed by DAVID. The biological network was constructed using the Cytoscape. Molecular Complex Detection (MCODE) was applied for detecting module analysis. Transcription factors were identified by the PANTHER classification system database and the gene regulatory network was constructed by Cytoscape. One hundred thirty-two DEGs were screened from comparison newly diagnosed ITP than chronic ITP. Biological process analysis revealed that the DEGs were enriched in terms of positive regulation of autophagy and prohibiting apoptosis in the chronic phase. KEGG pathway analysis showed that the DEGs were enriched in the ErbB signaling pathway, mRNA surveillance pathway, Estrogen signaling pathway, and Notch signaling pathway. Additionally, the biological network was established, and five modules were extracted from the network. ARRB1, VIM, SF1, BUB3, GRK5, and RHOG were detected as hub genes that also belonged to the modules. SF1 also was identified as a hub-TF gene. To sum up, microarray data analysis could perform a panel of genes that provides new clues for diagnosing chronic ITP.
Collapse
Affiliation(s)
- Samira Gilanchi
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Science, Tehran, Iran.,School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Faranoush
- Pediatric Growth and Development Research Center, Institute of Endocrinology, Iran University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Science, Tehran, Iran
| | | | - Mahyar Daskareh
- Department of Radiology, Ziyaian Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Kiriiri GK, Njogu PM, Mwangi AN. Exploring different approaches to improve the success of drug discovery and development projects: a review. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2020. [DOI: 10.1186/s43094-020-00047-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Abstract
Background
There has been a significant increase in the cost and timeline of delivering new drugs for clinical use over the last three decades. Despite the increased investments in research infrastructure by pharmaceutical companies and technological advances in the scientific tools available, efforts to increase the number of molecules coming through the drug development pipeline have largely been unfruitful.
Main body
A non-systematic review of the current literature was undertaken to enumerate the various strategies employed to improve the success rates in the pharmaceutical research and development. The review covers the exploitation of genomics and proteomics, complementarity of target-based and phenotypic efficacy screening platforms, drug repurposing and repositioning, collaborative research, focusing on underserved therapeutic fields, outsourcing strategy, and pharmaceutical modeling and artificial intelligence. Examples of successful drug discoveries achieved through application of these strategies are highlighted and discussed herein.
Conclusions
Genomics and proteomics have uncovered a wide array of potential drug targets and are facilitative of enhanced scrupulous target identification and validation thus reducing efficacy-related drug attrition. When used complementarily, phenotypic and target-based screening platforms would likely allow serendipitous drug discovery while increasing rationality in drug design. Drug repurposing and repositioning reduces financial risks in drug development accompanied by cost and time savings, while prolonging patent exclusivity hence increased returns on investment to the innovator company. Equally important, collaborative research is facilitative of cross-fertilization and refinement of ideas, while sharing resources and expertise, hence reducing overhead costs in the early stages of drug discovery. Underserved therapeutic fields are niche drug discovery areas that may be used to experiment and launch novel drug targets, while exploiting incentivized benefits afforded by drug regulatory authorities. Outsourcing allows the pharma industries to focus on their core competencies while deriving greater efficiency of specialist contract research organizations. The existing and emerging pharmaceutical modeling and artificial intelligence softwares and tools allow for in silico computation enabling more efficient computer-aided drug design. Careful selection and application of these strategies, singly or in combination, may potentially harness pharmaceutical research and innovation.
Collapse
|
7
|
Saha A, Kim Y, Gewirtz ADH, Jo B, Gao C, McDowell IC, Engelhardt BE, Battle A. Co-expression networks reveal the tissue-specific regulation of transcription and splicing. Genome Res 2017; 27:1843-1858. [PMID: 29021288 PMCID: PMC5668942 DOI: 10.1101/gr.216721.116] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 08/22/2017] [Indexed: 11/24/2022]
Abstract
Gene co-expression networks capture biologically important patterns in gene expression data, enabling functional analyses of genes, discovery of biomarkers, and interpretation of genetic variants. Most network analyses to date have been limited to assessing correlation between total gene expression levels in a single tissue or small sets of tissues. Here, we built networks that additionally capture the regulation of relative isoform abundance and splicing, along with tissue-specific connections unique to each of a diverse set of tissues. We used the Genotype-Tissue Expression (GTEx) project v6 RNA sequencing data across 50 tissues and 449 individuals. First, we developed a framework called Transcriptome-Wide Networks (TWNs) for combining total expression and relative isoform levels into a single sparse network, capturing the interplay between the regulation of splicing and transcription. We built TWNs for 16 tissues and found that hubs in these networks were strongly enriched for splicing and RNA binding genes, demonstrating their utility in unraveling regulation of splicing in the human transcriptome. Next, we used a Bayesian biclustering model that identifies network edges unique to a single tissue to reconstruct Tissue-Specific Networks (TSNs) for 26 distinct tissues and 10 groups of related tissues. Finally, we found genetic variants associated with pairs of adjacent nodes in our networks, supporting the estimated network structures and identifying 20 genetic variants with distant regulatory impact on transcription and splicing. Our networks provide an improved understanding of the complex relationships of the human transcriptome across tissues.
Collapse
|
8
|
Lötsch J, Lippmann C, Kringel D, Ultsch A. Integrated Computational Analysis of Genes Associated with Human Hereditary Insensitivity to Pain. A Drug Repurposing Perspective. Front Mol Neurosci 2017; 10:252. [PMID: 28848388 PMCID: PMC5550731 DOI: 10.3389/fnmol.2017.00252] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 07/26/2017] [Indexed: 12/31/2022] Open
Abstract
Genes causally involved in human insensitivity to pain provide a unique molecular source of studying the pathophysiology of pain and the development of novel analgesic drugs. The increasing availability of “big data” enables novel research approaches to chronic pain while also requiring novel techniques for data mining and knowledge discovery. We used machine learning to combine the knowledge about n = 20 genes causally involved in human hereditary insensitivity to pain with the knowledge about the functions of thousands of genes. An integrated computational analysis proposed that among the functions of this set of genes, the processes related to nervous system development and to ceramide and sphingosine signaling pathways are particularly important. This is in line with earlier suggestions to use these pathways as therapeutic target in pain. Following identification of the biological processes characterizing hereditary insensitivity to pain, the biological processes were used for a similarity analysis with the functions of n = 4,834 database-queried drugs. Using emergent self-organizing maps, a cluster of n = 22 drugs was identified sharing important functional features with hereditary insensitivity to pain. Several members of this cluster had been implicated in pain in preclinical experiments. Thus, the present concept of machine-learned knowledge discovery for pain research provides biologically plausible results and seems to be suitable for drug discovery by identifying a narrow choice of repurposing candidates, demonstrating that contemporary machine-learned methods offer innovative approaches to knowledge discovery from available evidence.
Collapse
Affiliation(s)
- Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-UniversityFrankfurt am Main, Germany.,Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group, Translational Medicine and Pharmacology (IME-TMP)Frankfurt am Main, Germany
| | - Catharina Lippmann
- Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group, Translational Medicine and Pharmacology (IME-TMP)Frankfurt am Main, Germany
| | - Dario Kringel
- Institute of Clinical Pharmacology, Goethe-UniversityFrankfurt am Main, Germany
| | - Alfred Ultsch
- DataBionics Research Group, University of MarburgMarburg, Germany
| |
Collapse
|
9
|
Ung MH, Varn FS, Cheng C. In silico frameworks for systematic pre-clinical screening of potential anti-leukemia therapeutics. Expert Opin Drug Discov 2016; 11:1213-1222. [PMID: 27689915 DOI: 10.1080/17460441.2016.1243524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Leukemia is a collection of highly heterogeneous cancers that arise from neoplastic transformation and clonal expansion of immature hematopoietic cells. Post-treatment recurrence is high, especially among elderly patients, thus necessitating more effective treatment modalities. Development of novel anti-leukemic compounds relies heavily on traditional in vitro screens which require extensive resources and time. Therefore, integration of in silico screens prior to experimental validation can improve the efficiency of pre-clinical drug development. Areas covered: This article reviews different methods and frameworks used to computationally screen for anti-leukemic agents. In particular, three approaches are discussed including molecular docking, transcriptomic integration, and network analysis. Expert opinion: Today's data deluge presents novel opportunities to develop computational tools and pipelines to screen for likely therapeutic candidates in the treatment of leukemia. Formal integration of these methodologies can accelerate and improve the efficiency of modern day anti-leukemic drug discovery and ease the economic and healthcare burden associated with it.
Collapse
Affiliation(s)
- Matthew H Ung
- a Department of Molecular and Systems Biology , Geisel School of Medicine at Dartmouth , Hanover , NH , USA
| | - Frederick S Varn
- a Department of Molecular and Systems Biology , Geisel School of Medicine at Dartmouth , Hanover , NH , USA
| | - Chao Cheng
- a Department of Molecular and Systems Biology , Geisel School of Medicine at Dartmouth , Hanover , NH , USA.,b Department of Biomedical Data Science , Geisel School of Medicine at Dartmouth , Lebanon , NH , USA.,c Norris Cotton Cancer Center , Lebanon , NH , USA
| |
Collapse
|
10
|
Multi-target therapeutics for neuropsychiatric and neurodegenerative disorders. Drug Discov Today 2016; 21:1886-1914. [PMID: 27506871 DOI: 10.1016/j.drudis.2016.08.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/20/2016] [Accepted: 08/01/2016] [Indexed: 12/30/2022]
Abstract
Historically, neuropsychiatric and neurodegenerative disease treatments focused on the 'magic bullet' concept; however multi-targeted strategies are increasingly attractive gauging from the escalating research in this area. Because these diseases are typically co-morbid, multi-targeted drugs capable of interacting with multiple targets will expand treatment to the co-morbid disease condition. Despite their theoretical efficacy, there are significant impediments to clinical success (e.g., difficulty titrating individual aspects of the drug and inconclusive pathophysiological mechanisms). The new and revised diagnostic frameworks along with studies detailing the endophenotypic characteristics of the diseases promise to provide the foundation for the circumvention of these impediments. This review serves to evaluate the various marketed and nonmarketed multi-targeted drugs with particular emphasis on their design strategy.
Collapse
|
11
|
Ung MH, Liu CC, Cheng C. Integrative analysis of cancer genes in a functional interactome. Sci Rep 2016; 6:29228. [PMID: 27356765 PMCID: PMC4928112 DOI: 10.1038/srep29228] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/16/2016] [Indexed: 11/09/2022] Open
Abstract
The post-genomic era has resulted in the accumulation of high-throughput cancer data from a vast array of genomic technologies including next-generation sequencing and microarray. As such, the large amounts of germline variant and somatic mutation data that have been generated from GWAS and sequencing projects, respectively, show great promise in providing a systems-level view of these genetic aberrations. In this study, we analyze publicly available GWAS, somatic mutation, and drug target data derived from large databanks using a network-based approach that incorporates directed edge information under a randomized network hypothesis testing procedure. We show that these three classes of disease-associated nodes exhibit non-random topological characteristics in the context of a functional interactome. Specifically, we show that drug targets tend to lie upstream of somatic mutations and disease susceptibility germline variants. In addition, we introduce a new approach to measuring hierarchy between drug targets, somatic mutants, and disease susceptibility genes by utilizing directionality and path length information. Overall, our results provide new insight into the intrinsic relationships between these node classes that broaden our understanding of cancer. In addition, our results align with current knowledge on the therapeutic actionability of GWAS and somatic mutant nodes, while demonstrating relationships between node classes from a global network perspective.
Collapse
Affiliation(s)
- Matthew H Ung
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, 03755 USA.,Program in Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, 03755 USA
| | - Chun-Chi Liu
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taiwan
| | - Chao Cheng
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, 03755 USA.,Program in Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, 03755 USA.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, 03766 USA
| |
Collapse
|
12
|
Chen J, Zhang S. Integrative analysis for identifying joint modular patterns of gene-expression and drug-response data. Bioinformatics 2016; 32:1724-32. [DOI: 10.1093/bioinformatics/btw059] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/27/2016] [Indexed: 12/13/2022] Open
|
13
|
Saykin AJ, Shen L, Yao X, Kim S, Nho K, Risacher SL, Ramanan VK, Foroud TM, Faber KM, Sarwar N, Munsie LM, Hu X, Soares HD, Potkin SG, Thompson PM, Kauwe JSK, Kaddurah-Daouk R, Green RC, Toga AW, Weiner MW. Genetic studies of quantitative MCI and AD phenotypes in ADNI: Progress, opportunities, and plans. Alzheimers Dement 2015; 11:792-814. [PMID: 26194313 PMCID: PMC4510473 DOI: 10.1016/j.jalz.2015.05.009] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 05/08/2015] [Accepted: 05/08/2015] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Genetic data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) have been crucial in advancing the understanding of Alzheimer's disease (AD) pathophysiology. Here, we provide an update on sample collection, scientific progress and opportunities, conceptual issues, and future plans. METHODS Lymphoblastoid cell lines and DNA and RNA samples from blood have been collected and banked, and data and biosamples have been widely disseminated. To date, APOE genotyping, genome-wide association study (GWAS), and whole exome and whole genome sequencing data have been obtained and disseminated. RESULTS ADNI genetic data have been downloaded thousands of times, and >300 publications have resulted, including reports of large-scale GWAS by consortia to which ADNI contributed. Many of the first applications of quantitative endophenotype association studies used ADNI data, including some of the earliest GWAS and pathway-based studies of biospecimen and imaging biomarkers, as well as memory and other clinical/cognitive variables. Other contributions include some of the first whole exome and whole genome sequencing data sets and reports in healthy controls, mild cognitive impairment, and AD. DISCUSSION Numerous genetic susceptibility and protective markers for AD and disease biomarkers have been identified and replicated using ADNI data and have heavily implicated immune, mitochondrial, cell cycle/fate, and other biological processes. Early sequencing studies suggest that rare and structural variants are likely to account for significant additional phenotypic variation. Longitudinal analyses of transcriptomic, proteomic, metabolomic, and epigenomic changes will also further elucidate dynamic processes underlying preclinical and prodromal stages of disease. Integration of this unique collection of multiomics data within a systems biology framework will help to separate truly informative markers of early disease mechanisms and potential novel therapeutic targets from the vast background of less relevant biological processes. Fortunately, a broad swath of the scientific community has accepted this grand challenge.
Collapse
Affiliation(s)
- Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Li Shen
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiaohui Yao
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; School of Informatics and Computing, Indiana University, Purdue University - Indianapolis, Indianapolis, IN, USA
| | - Sungeun Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Vijay K Ramanan
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tatiana M Foroud
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kelley M Faber
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Xiaolan Hu
- Bristol-Myers Squibb, Wallingford, CT, USA
| | | | - Steven G Potkin
- Department of Psychiatry and Human Behavior, University of California - Irvine, Irvine, CA, USA
| | - Paul M Thompson
- Department of Neurology, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA; Imaging Genetics Center, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, UT, USA; Department of Neuroscience, Brigham Young University, Provo, UT, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA; Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Robert C Green
- Partners Center for Personalized Genetic Medicine, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, Institute for Neuroimaging and Neuroinformatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Michael W Weiner
- Department of Radiology, University of California-San Francisco, San Francisco, CA, USA; Department of Medicine, University of California-San Francisco, San Francisco, CA, USA; Department of Psychiatry, University of California-San Francisco, San Francisco, CA, USA; Center for Imaging of Neurodegenerative Diseases, San Francisco VA Medical Center, San Francisco, CA, USA
| |
Collapse
|
14
|
Millan MJ, Goodwin GM, Meyer-Lindenberg A, Ove Ögren S. Learning from the past and looking to the future: Emerging perspectives for improving the treatment of psychiatric disorders. Eur Neuropsychopharmacol 2015; 25:599-656. [PMID: 25836356 DOI: 10.1016/j.euroneuro.2015.01.016] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 01/28/2015] [Indexed: 02/06/2023]
Abstract
Modern neuropsychopharmacology commenced in the 1950s with the serendipitous discovery of first-generation antipsychotics and antidepressants which were therapeutically effective yet had marked adverse effects. Today, a broader palette of safer and better-tolerated agents is available for helping people that suffer from schizophrenia, depression and other psychiatric disorders, while complementary approaches like psychotherapy also have important roles to play in their treatment, both alone and in association with medication. Nonetheless, despite considerable efforts, current management is still only partially effective, and highly-prevalent psychiatric disorders of the brain continue to represent a huge personal and socio-economic burden. The lack of success in discovering more effective pharmacotherapy has contributed, together with many other factors, to a relative disengagement by pharmaceutical firms from neuropsychiatry. Nonetheless, interest remains high, and partnerships are proliferating with academic centres which are increasingly integrating drug discovery and translational research into their traditional activities. This is, then, a time of transition and an opportune moment to thoroughly survey the field. Accordingly, the present paper, first, chronicles the discovery and development of psychotropic agents, focusing in particular on their mechanisms of action and therapeutic utility, and how problems faced were eventually overcome. Second, it discusses the lessons learned from past successes and failures, and how they are being applied to promote future progress. Third, it comprehensively surveys emerging strategies that are (1), improving our understanding of the diagnosis and classification of psychiatric disorders; (2), deepening knowledge of their underlying risk factors and pathophysiological substrates; (3), refining cellular and animal models for discovery and validation of novel therapeutic agents; (4), improving the design and outcome of clinical trials; (5), moving towards reliable biomarkers of patient subpopulations and medication efficacy and (6), promoting collaborative approaches to innovation by uniting key partners from the regulators, industry and academia to patients. Notwithstanding the challenges ahead, the many changes and ideas articulated herein provide new hope and something of a framework for progress towards the improved prevention and relief of psychiatric and other CNS disorders, an urgent mission for our Century.
Collapse
Affiliation(s)
- Mark J Millan
- Pole for Innovation in Neurosciences, IDR Servier, 125 chemin de ronde, 78290 Croissy sur Seine, France.
| | - Guy M Goodwin
- University Department of Psychiatry, Oxford University, Warneford Hospital, Oxford OX3 7JX, England, UK
| | - Andreas Meyer-Lindenberg
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, J5, D-68159 Mannheim, Germany
| | - Sven Ove Ögren
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, S-17177 Stockholm, Sweden
| |
Collapse
|
15
|
Talwar P, Silla Y, Grover S, Gupta M, Grewal GK, Kukreti R. Systems Pharmacology and Pharmacogenomics for Drug Discovery and Development. SYSTEMS AND SYNTHETIC BIOLOGY 2015. [DOI: 10.1007/978-94-017-9514-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
16
|
On 'polypharmacy' and multi-target agents, complementary strategies for improving the treatment of depression: a comparative appraisal. Int J Neuropsychopharmacol 2014; 17:1009-37. [PMID: 23719026 DOI: 10.1017/s1461145712001496] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Major depression is a heterogeneous disorder, both in terms of symptoms, ranging from anhedonia to cognitive impairment, and in terms of pathogenesis, with many interacting genetic, epigenetic, developmental and environmental causes. Accordingly, it seems unlikely that depressive states could be fully controlled by a drug possessing one discrete mechanism of action and, in the wake of disappointing results with several classes of highly selective agent, multi-modal treatment concepts are attracting attention. As concerns pharmacotherapy, there are essentially two core strategies. First, multi-target antidepressants that act via two or more complementary mechanisms and, second, polypharmacy, which refers to co-administration of two distinct drugs, usually in separate pills. Both multi-target agents and polypharmacy ideally couple a therapeutically unexploited action to a clinically established mechanism in order to enhance efficacy, moderate side-effects, accelerate onset of action and treat a broader range of symptoms. The melatonin MT1/MT2 agonist and 5-HT(2C) antagonist, agomelatine, which is effective in the short- and long-term treatment of depression, exemplifies the former approach, while evidence-based polypharmacy is illustrated by the adjunctive use of second-generation antipsychotics with serotonin reuptake inhibitors for treatment of resistant depression. Histone acetylation and methylation, ghrelin signalling, inflammatory modulators, metabotropic glutamate-7 receptors and trace amine-associated-1 receptors comprise attractive substrates for new multi-target and polypharmaceutical strategies. The present article outlines the rationale underpinning multi-modal approaches for treating depression, and critically compares and contrasts the pros and cons of established and potentially novel multi-target vs. polypharmaceutical treatments. On balance, the former appear the most promising for the elaboration, development and clinical implementation of innovative concepts for the more effective management of depression.
Collapse
|
17
|
Shen L, Thompson PM, Potkin SG, Bertram L, Farrer LA, Foroud TM, Green RC, Hu X, Huentelman MJ, Kim S, Kauwe JSK, Li Q, Liu E, Macciardi F, Moore JH, Munsie L, Nho K, Ramanan VK, Risacher SL, Stone DJ, Swaminathan S, Toga AW, Weiner MW, Saykin AJ. Genetic analysis of quantitative phenotypes in AD and MCI: imaging, cognition and biomarkers. Brain Imaging Behav 2014; 8:183-207. [PMID: 24092460 PMCID: PMC3976843 DOI: 10.1007/s11682-013-9262-z] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The Genetics Core of the Alzheimer's Disease Neuroimaging Initiative (ADNI), formally established in 2009, aims to provide resources and facilitate research related to genetic predictors of multidimensional Alzheimer's disease (AD)-related phenotypes. Here, we provide a systematic review of genetic studies published between 2009 and 2012 where either ADNI APOE genotype or genome-wide association study (GWAS) data were used. We review and synthesize ADNI genetic associations with disease status or quantitative disease endophenotypes including structural and functional neuroimaging, fluid biomarker assays, and cognitive performance. We also discuss the diverse analytical strategies used in these studies, including univariate and multivariate analysis, meta-analysis, pathway analysis, and interaction and network analysis. Finally, we perform pathway and network enrichment analyses of these ADNI genetic associations to highlight key mechanisms that may drive disease onset and trajectory. Major ADNI findings included all the top 10 AD genes and several of these (e.g., APOE, BIN1, CLU, CR1, and PICALM) were corroborated by ADNI imaging, fluid and cognitive phenotypes. ADNI imaging genetics studies discovered novel findings (e.g., FRMD6) that were later replicated on different data sets. Several other genes (e.g., APOC1, FTO, GRIN2B, MAGI2, and TOMM40) were associated with multiple ADNI phenotypes, warranting further investigation on other data sets. The broad availability and wide scope of ADNI genetic and phenotypic data has advanced our understanding of the genetic basis of AD and has nominated novel targets for future studies employing next-generation sequencing and convergent multi-omics approaches, and for clinical drug and biomarker development.
Collapse
Affiliation(s)
- Li Shen
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - Paul M. Thompson
- Imaging Genetics Center, Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA 90095 USA
| | - Steven G. Potkin
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92617 USA
| | - Lars Bertram
- Neuropsychiatric Genetics Group, Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | - Lindsay A. Farrer
- Biomedical Genetics L320, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118 USA
| | - Tatiana M. Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Robert C. Green
- Division of Genetics and Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115 USA
| | - Xiaolan Hu
- Clinical Genetics, Exploratory Clinical & Translational Research, Bristol-Myers Squibbs, Pennington, NJ 08534 USA
| | - Matthew J. Huentelman
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ 85004 USA
| | - Sungeun Kim
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - John S. K. Kauwe
- Departments of Biology, Neuroscience, Brigham Young University, 675 WIDB, Provo, UT 84602 USA
| | - Qingqin Li
- Department of Neuroscience Biomarkers, Janssen Research and Development, LLC, Raritan, NJ 08869 USA
| | - Enchi Liu
- Biomarker Discovery, Janssen Alzheimer Immunotherapy Research and Development, LLC, South San Francisco, CA 94080 USA
| | - Fabio Macciardi
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92617 USA
- Department of Sciences and Biomedical Technologies, University of Milan, Segrate, MI Italy
| | - Jason H. Moore
- Department of Genetics, Computational Genetics Laboratory, Dartmouth Medical School, Lebanon, NH 03756 USA
| | - Leanne Munsie
- Tailored Therapeutics, Eli Lilly and Company, Indianapolis, IN 46285 USA
| | - Kwangsik Nho
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - Vijay K. Ramanan
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Shannon L. Risacher
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - David J. Stone
- Merck Research Laboratories, 770 Sumneytown Pike, WP53B-120, West Point, PA 19486 USA
| | - Shanker Swaminathan
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA 90095 USA
| | - Michael W. Weiner
- Departments of Radiology, Medicine and Psychiatry, UC San Francisco, San Francisco, CA 94143 USA
| | - Andrew J. Saykin
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - for the Alzheimer’s Disease Neuroimaging Initiative
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
- Imaging Genetics Center, Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA 90095 USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92617 USA
- Neuropsychiatric Genetics Group, Max-Planck Institute for Molecular Genetics, Berlin, Germany
- Biomedical Genetics L320, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118 USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
- Division of Genetics and Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115 USA
- Clinical Genetics, Exploratory Clinical & Translational Research, Bristol-Myers Squibbs, Pennington, NJ 08534 USA
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ 85004 USA
- Departments of Biology, Neuroscience, Brigham Young University, 675 WIDB, Provo, UT 84602 USA
- Department of Neuroscience Biomarkers, Janssen Research and Development, LLC, Raritan, NJ 08869 USA
- Biomarker Discovery, Janssen Alzheimer Immunotherapy Research and Development, LLC, South San Francisco, CA 94080 USA
- Department of Sciences and Biomedical Technologies, University of Milan, Segrate, MI Italy
- Department of Genetics, Computational Genetics Laboratory, Dartmouth Medical School, Lebanon, NH 03756 USA
- Tailored Therapeutics, Eli Lilly and Company, Indianapolis, IN 46285 USA
- Merck Research Laboratories, 770 Sumneytown Pike, WP53B-120, West Point, PA 19486 USA
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA 90095 USA
- Departments of Radiology, Medicine and Psychiatry, UC San Francisco, San Francisco, CA 94143 USA
| |
Collapse
|
18
|
Ramanan VK, Saykin AJ. Pathways to neurodegeneration: mechanistic insights from GWAS in Alzheimer's disease, Parkinson's disease, and related disorders. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2013; 2:145-175. [PMID: 24093081 PMCID: PMC3783830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 08/25/2013] [Indexed: 06/02/2023]
Abstract
The discovery of causative genetic mutations in affected family members has historically dominated our understanding of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS). Nevertheless, most cases of neurodegenerative disease are not explained by Mendelian inheritance of known genetic variants, but instead are thought to have a complex etiology with numerous genetic and environmental factors contributing to susceptibility. Although unbiased genome-wide association studies (GWAS) have identified novel associations to neurodegenerative diseases, most of these hits explain only modest fractions of disease heritability. In addition, despite the substantial overlap of clinical and pathologic features among major neurodegenerative diseases, surprisingly few GWAS-implicated variants appear to exhibit cross-disease association. These realities suggest limitations of the focus on individual genetic variants and create challenges for the development of diagnostic and therapeutic strategies, which traditionally target an isolated molecule or mechanistic step. Recently, GWAS of complex diseases and traits have focused less on individual susceptibility variants and instead have emphasized the biological pathways and networks revealed by genetic associations. This new paradigm draws on the hypothesis that fundamental disease processes may be influenced on a personalized basis by a combination of variants - some common and others rare, some protective and others deleterious - in key genes and pathways. Here, we review and synthesize the major pathways implicated in neurodegeneration, focusing on GWAS from the most prevalent neurodegenerative disorders, AD and PD. Using literature mining, we also discover a novel regulatory network that is enriched with AD- and PD-associated genes and centered on the SP1 and AP-1 (Jun/Fos) transcription factors. Overall, this pathway- and network-driven model highlights several potential shared mechanisms in AD and PD that will inform future studies of these and other neurodegenerative disorders. These insights also suggest that biomarker and treatment strategies may require simultaneous targeting of multiple components, including some specific to disease stage, in order to assess and modulate neurodegeneration. Pathways and networks will provide ideal vehicles for integrating relevant findings from GWAS and other modalities to enhance clinical translation.
Collapse
Affiliation(s)
- Vijay K Ramanan
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of MedicineIndianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of MedicineIndianapolis, IN, USA
- Medical Scientist Training Program, Indiana University School of MedicineIndianapolis, IN, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of MedicineIndianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of MedicineIndianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of MedicineIndianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of MedicineIndianapolis, IN, USA
| |
Collapse
|
19
|
Genome-wide pathway analysis of memory impairment in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort implicates gene candidates, canonical pathways, and networks. Brain Imaging Behav 2013; 6:634-48. [PMID: 22865056 DOI: 10.1007/s11682-012-9196-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Memory deficits are prominent features of mild cognitive impairment (MCI) and Alzheimer's disease (AD). The genetic architecture underlying these memory deficits likely involves the combined effects of multiple genetic variants operative within numerous biological pathways. In order to identify functional pathways associated with memory impairment, we performed a pathway enrichment analysis on genome-wide association data from 742 Alzheimer's Disease Neuroimaging Initiative (ADNI) participants. A composite measure of memory was generated as the phenotype for this analysis by applying modern psychometric theory to item-level data from the ADNI neuropsychological test battery. Using the GSA-SNP software tool, we identified 27 canonical, expertly-curated pathways with enrichment (FDR-corrected p-value < 0.05) against this composite memory score. Processes classically understood to be involved in memory consolidation, such as neurotransmitter receptor-mediated calcium signaling and long-term potentiation, were highly represented among the enriched pathways. In addition, pathways related to cell adhesion, neuronal differentiation and guided outgrowth, and glucose- and inflammation-related signaling were also enriched. Among genes that were highly-represented in these enriched pathways, we found indications of coordinated relationships, including one large gene set that is subject to regulation by the SP1 transcription factor, and another set that displays co-localized expression in normal brain tissue along with known AD risk genes. These results 1) demonstrate that psychometrically-derived composite memory scores are an effective phenotype for genetic investigations of memory impairment and 2) highlight the promise of pathway analysis in elucidating key mechanistic targets for future studies and for therapeutic interventions.
Collapse
|
20
|
Yang M, Chen JL, Xu LW, Ji G. Navigating traditional chinese medicine network pharmacology and computational tools. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:731969. [PMID: 23983798 PMCID: PMC3747450 DOI: 10.1155/2013/731969] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/04/2013] [Indexed: 12/17/2022]
Abstract
The concept of "network target" has ushered in a new era in the field of traditional Chinese medicine (TCM). As a new research approach, network pharmacology is based on the analysis of network models and systems biology. Taking advantage of advancements in systems biology, a high degree of integration data analysis strategy and interpretable visualization provides deeper insights into the underlying mechanisms of TCM theories, including the principles of herb combination, biological foundations of herb or herbal formulae action, and molecular basis of TCM syndromes. In this study, we review several recent developments in TCM network pharmacology research and discuss their potential for bridging the gap between traditional and modern medicine. We briefly summarize the two main functional applications of TCM network models: understanding/uncovering and predicting/discovering. In particular, we focus on how TCM network pharmacology research is conducted and highlight different computational tools, such as network-based and machine learning algorithms, and sources that have been proposed and applied to the different steps involved in the research process. To make network pharmacology research commonplace, some basic network definitions and analysis methods are presented.
Collapse
Affiliation(s)
- Ming Yang
- Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai 200032, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jia-Lei Chen
- Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai 200032, China
| | - Li-Wen Xu
- Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai 200032, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
21
|
Csermely P, Korcsmáros T, Kiss HJM, London G, Nussinov R. Structure and dynamics of molecular networks: a novel paradigm of drug discovery: a comprehensive review. Pharmacol Ther 2013; 138:333-408. [PMID: 23384594 PMCID: PMC3647006 DOI: 10.1016/j.pharmthera.2013.01.016] [Citation(s) in RCA: 512] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 01/22/2013] [Indexed: 02/02/2023]
Abstract
Despite considerable progress in genome- and proteome-based high-throughput screening methods and in rational drug design, the increase in approved drugs in the past decade did not match the increase of drug development costs. Network description and analysis not only give a systems-level understanding of drug action and disease complexity, but can also help to improve the efficiency of drug design. We give a comprehensive assessment of the analytical tools of network topology and dynamics. The state-of-the-art use of chemical similarity, protein structure, protein-protein interaction, signaling, genetic interaction and metabolic networks in the discovery of drug targets is summarized. We propose that network targeting follows two basic strategies. The "central hit strategy" selectively targets central nodes/edges of the flexible networks of infectious agents or cancer cells to kill them. The "network influence strategy" works against other diseases, where an efficient reconfiguration of rigid networks needs to be achieved by targeting the neighbors of central nodes/edges. It is shown how network techniques can help in the identification of single-target, edgetic, multi-target and allo-network drug target candidates. We review the recent boom in network methods helping hit identification, lead selection optimizing drug efficacy, as well as minimizing side-effects and drug toxicity. Successful network-based drug development strategies are shown through the examples of infections, cancer, metabolic diseases, neurodegenerative diseases and aging. Summarizing >1200 references we suggest an optimized protocol of network-aided drug development, and provide a list of systems-level hallmarks of drug quality. Finally, we highlight network-related drug development trends helping to achieve these hallmarks by a cohesive, global approach.
Collapse
Affiliation(s)
- Peter Csermely
- Department of Medical Chemistry, Semmelweis University, P.O. Box 260, H-1444 Budapest 8, Hungary.
| | | | | | | | | |
Collapse
|
22
|
Duran-Frigola M, Mosca R, Aloy P. Structural Systems Pharmacology: The Role of 3D Structures in Next-Generation Drug Development. ACTA ACUST UNITED AC 2013; 20:674-84. [DOI: 10.1016/j.chembiol.2013.03.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 02/28/2013] [Accepted: 03/05/2013] [Indexed: 01/12/2023]
|
23
|
Current World Literature. Curr Opin Cardiol 2013; 28:369-79. [DOI: 10.1097/hco.0b013e328360f5be] [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: 11/26/2022]
|
24
|
Gerwick WH, Fenner AM. Drug discovery from marine microbes. MICROBIAL ECOLOGY 2013; 65:800-6. [PMID: 23274881 PMCID: PMC3640700 DOI: 10.1007/s00248-012-0169-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 12/13/2012] [Indexed: 05/04/2023]
Abstract
The marine environment has been a source of more than 20,000 inspirational natural products discovered over the past 50 years. From these efforts, 9 approved drugs and 12 current clinical trial agents have been discovered, either as natural products or as molecules inspired from the natural product structure. To a significant degree, these have come from collections of marine invertebrates largely obtained from shallow-water tropical ecosystems. However, there is a growing recognition that marine invertebrates are oftentimes populated with enormous quantities of "associated" or symbiotic microorganisms and that microorganisms are the true metabolic sources of these most valuable of marine natural products. Also, because of the inherently multidisciplinary nature of this field, a high degree of innovation is characteristic of marine natural product drug discovery efforts.
Collapse
Affiliation(s)
- William H Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92093, USA.
| | | |
Collapse
|
25
|
Affiliation(s)
- Nagasuma Chandra
- Indian Institute of Science, Department of Biochemistry,
Bangalore – 560012, India ,
| | - Jyothi Padiadpu
- Indian Institute of Science, Department of Biochemistry,
Bangalore – 560012, India
| |
Collapse
|
26
|
Pathway analysis of genomic data: concepts, methods, and prospects for future development. Trends Genet 2012; 28:323-32. [PMID: 22480918 DOI: 10.1016/j.tig.2012.03.004] [Citation(s) in RCA: 215] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/02/2012] [Accepted: 03/07/2012] [Indexed: 12/31/2022]
Abstract
Genome-wide data sets are increasingly being used to identify biological pathways and networks underlying complex diseases. In particular, analyzing genomic data through sets defined by functional pathways offers the potential of greater power for discovery and natural connections to biological mechanisms. With the burgeoning availability of next-generation sequencing, this is an opportune moment to revisit strategies for pathway-based analysis of genomic data. Here, we synthesize relevant concepts and extant methodologies to guide investigators in study design and execution. We also highlight ongoing challenges and proposed solutions. As relevant analytical strategies mature, pathways and networks will be ideally placed to integrate data from diverse -omics sources to harness the extensive, rich information related to disease and treatment mechanisms.
Collapse
|
27
|
Millan MJ, Agid Y, Brüne M, Bullmore ET, Carter CS, Clayton NS, Connor R, Davis S, Deakin B, DeRubeis RJ, Dubois B, Geyer MA, Goodwin GM, Gorwood P, Jay TM, Joëls M, Mansuy IM, Meyer-Lindenberg A, Murphy D, Rolls E, Saletu B, Spedding M, Sweeney J, Whittington M, Young LJ. Cognitive dysfunction in psychiatric disorders: characteristics, causes and the quest for improved therapy. Nat Rev Drug Discov 2012; 11:141-68. [PMID: 22293568 DOI: 10.1038/nrd3628] [Citation(s) in RCA: 815] [Impact Index Per Article: 67.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Studies of psychiatric disorders have traditionally focused on emotional symptoms such as depression, anxiety and hallucinations. However, poorly controlled cognitive deficits are equally prominent and severely compromise quality of life, including social and professional integration. Consequently, intensive efforts are being made to characterize the cellular and cerebral circuits underpinning cognitive function, define the nature and causes of cognitive impairment in psychiatric disorders and identify more effective treatments. Successful development will depend on rigorous validation in animal models as well as in patients, including measures of real-world cognitive functioning. This article critically discusses these issues, highlighting the challenges and opportunities for improving cognition in individuals suffering from psychiatric disorders.
Collapse
Affiliation(s)
- Mark J Millan
- Institut de Recherche Servier, 78290 Croissy/Seine, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
The era of targeted cancer therapies has arrived. However, due to the complexity of biological systems, the current progress is far from enough. From biological network modeling to structural/dynamic network analysis, network systems biology provides unique insight into the potential mechanisms underlying the growth and progression of cancer cells. It has also introduced great changes into the research paradigm of cancer-associated drug discovery and drug resistance.
Collapse
Affiliation(s)
- Ting-Ting Zhou
- Department of Immunology, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100850, P. R. China.
| |
Collapse
|