1
|
Sinha S, McLaren E, Mullick M, Singh S, Boland BS, Ghosh P. FORWARD: A Learning Framework for Logical Network Perturbations to Prioritize Targets for Drug Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.602603. [PMID: 39071297 PMCID: PMC11275938 DOI: 10.1101/2024.07.16.602603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Despite advances in artificial intelligence (AI), target-based drug development remains a costly, complex and imprecise process. We introduce F.O.R.W.A.R.D [ Framework for Outcome-based Research and Drug Development ], a network-based target prioritization approach and test its utility in the challenging therapeutic area of Inflammatory Bowel Diseases (IBD), which is a chronic condition of multifactorial origin. F.O.R.W.A.R.D leverages real-world outcomes, using a machine-learning classifier trained on transcriptomic data from seven prospective randomized clinical trials involving four drugs. It establishes a molecular signature of remission as the therapeutic goal and computes, by integrating principles of network connectivity, the likelihood that a drug's action on its target(s) will induce the remission-associated genes. Benchmarking F.O.R.W.A.R.D against 210 completed clinical trials on 52 targets showed a perfect predictive accuracy of 100%. The success of F.O.R.W.A.R.D was achieved despite differences in targets, mechanisms, and trial designs. F.O.R.W.A.R.D-driven in-silico phase '0' trials revealed its potential to inform trial design, justify re-trialing failed drugs, and guide early terminations. With its extendable applications to other therapeutic areas and its iterative refinement with emerging trials, F.O.R.W.A.R.D holds the promise to transform drug discovery by generating foresight from hindsight and impacting research and development as well as human-in-the-loop clinical decision-making.
Collapse
|
2
|
Sayed IM, Vo DT, Alcantara J, Inouye KM, Pranadinata RF, Luo L, Boland CR, Goyal NP, Kuo DJ, Huang SC, Sahoo D, Ghosh P, Das S. Molecular Signatures for Microbe-Associated Colorectal Cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.26.595902. [PMID: 38853996 PMCID: PMC11160670 DOI: 10.1101/2024.05.26.595902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Genetic factors and microbial imbalances play crucial roles in colorectal cancers (CRCs), yet the impact of infections on cancer initiation remains poorly understood. While bioinformatic approaches offer valuable insights, the rising incidence of CRCs creates a pressing need to precisely identify early CRC events. We constructed a network model to identify continuum states during CRC initiation spanning normal colonic tissue to pre-cancer lesions (adenomatous polyps) and examined the influence of microbes and host genetics. Methods A Boolean network was built using a publicly available transcriptomic dataset from healthy and adenoma affected patients to identify an invariant Microbe-Associated Colorectal Cancer Signature (MACS). We focused on Fusobacterium nucleatum ( Fn ), a CRC-associated microbe, as a model bacterium. MACS-associated genes and proteins were validated by RT-qPCR, RNA seq, ELISA, IF and IHCs in tissues and colon-derived organoids from genetically predisposed mice ( CPC-APC Min+/- ) and patients (FAP, Lynch Syndrome, PJS, and JPS). Results The MACS that is upregulated in adenomas consists of four core genes/proteins: CLDN2/Claudin-2 (leakiness), LGR5/leucine-rich repeat-containing receptor (stemness), CEMIP/cell migration-inducing and hyaluronan-binding protein (epithelial-mesenchymal transition) and IL8/Interleukin-8 (inflammation). MACS was induced upon Fn infection, but not in response to infection with other enteric bacteria or probiotics. MACS induction upon Fn infection was higher in CPC-APC Min+/- organoids compared to WT controls. The degree of MACS expression in the patient-derived organoids (PDOs) generally corresponded with the known lifetime risk of CRCs. Conclusions Computational prediction followed by validation in the organoid-based disease model identified the early events in CRC initiation. MACS reveals that the CRC-associated microbes induce a greater risk in the genetically predisposed hosts, suggesting its potential use for risk prediction and targeted cancer prevention.
Collapse
|
3
|
Horowitz A, Yu H, Pandey S, Mishra B, Sahoo D. C1QA is an invariant biomarker for tissue macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577475. [PMID: 38328228 PMCID: PMC10849641 DOI: 10.1101/2024.01.26.577475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Macrophages play a pivotal role in immune responses, particularly in the context of combating microbial threats within tissues. The identification of reliable biomarkers associated with macrophage function is essential for understanding their diverse roles in host defense. This study investigates the potential of C1QA as an invariant biomarker for tissue macrophages, focusing on its correlation with the anti-microbial pathway. C1QA, a component of the complement system, has been previously implicated in various immune functions. Our research delves into the specific association of C1QA with tissue-resident macrophages and its implications in the context of anti-microbial responses. Through comprehensive systems biology and Boolean analysis of gene expression, we aim to establish C1QA as a consistent and reliable marker for identifying tissue macrophages. Furthermore, we explore the functional significance of C1QA in the anti-microbial pathway. This research seeks to provide valuable insights into the molecular mechanisms underlying the anti-microbial functions of tissue macrophages, with C1QA emerging as a potential key player in this intricate regulatory network. Understanding the relationship between C1QA, tissue macrophages, and the anti-microbial pathway could pave the way for the development of targeted therapeutic strategies aimed at enhancing the host's ability to combat infections. Ultimately, our findings contribute to the expanding knowledge of macrophage biology and may have implications for the diagnosis and treatment of infectious diseases. One Sentence Summary C1QA is a fundamental biomarker of tissue macrophages.
Collapse
|
4
|
Sinha S, Alcantara J, Perry K, Castillo V, Espinoza CR, Taheri S, Vidales E, Tindle C, Adel A, Amirfakhri S, Sawires JR, Yang J, Bouvet M, Sahoo D, Ghosh P. Machine-Learning Identifies a Strategy for Differentiation Therapy in Solid Tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.13.557628. [PMID: 37745574 PMCID: PMC10515918 DOI: 10.1101/2023.09.13.557628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
BACKGROUND Although differentiation therapy can cure some hematologic malignancies, its curative potential remains unrealized in solid tumors. This is because conventional computational approaches succumb to the thunderous noise of inter-/intratumoral heterogeneity. Using colorectal cancers (CRCs) as an example, here we outline a machine learning(ML)-based approach to track, differentiate, and selectively target cancer stem cells (CSCs). METHODS A transcriptomic network was built and validated using healthy colon and CRC tissues in diverse gene expression datasets (~5,000 human and >300 mouse samples). Therapeutic targets and perturbation strategies were prioritized using ML, with the goal of reinstating the expression of a transcriptional identifier of the differentiated colonocyte, CDX2, whose loss in poorly differentiated (CSC-enriched) CRCs doubles the risk of relapse/death. The top candidate target was then engaged with a clinical-grade drug and tested on 3 models: CRC lines in vitro, xenografts in mice, and in a prospective cohort of healthy (n = 3) and CRC (n = 23) patient-derived organoids (PDOs). RESULTS The drug shifts the network predictably, induces CDX2 and crypt differentiation, and shows cytotoxicity in all 3 models, with a high degree of selectivity towards all CDX2-negative cell lines, xenotransplants, and PDOs. The potential for effective pairing of therapeutic efficacy (IC50) and biomarker (CDX2-low state) is confirmed in PDOs using multivariate analyses. A 50-gene signature of therapeutic response is derived and tested on 9 independent cohorts (~1700 CRCs), revealing the impact of CDX2-reinstatement therapy could translate into a ~50% reduction in the risk of mortality/recurrence. CONCLUSIONS Findings not only validate the precision of the ML approach in targeting CSCs, and objectively assess its impact on clinical outcome, but also exemplify the use of ML in yielding clinical directive information for enhancing personalized medicine.
Collapse
|
5
|
Anandachar MS, Roy S, Sinha S, Boadi A, Katkar GD, Ghosh P. Diverse gut pathogens exploit the host engulfment pathway via a conserved mechanism. J Biol Chem 2023; 299:105390. [PMID: 37890785 PMCID: PMC10696401 DOI: 10.1016/j.jbc.2023.105390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/22/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Macrophages clear infections by engulfing and digesting pathogens within phagolysosomes. Pathogens escape this fate by engaging in a molecular arms race; they use WxxxE motif-containing "effector" proteins to subvert the host cells they invade and seek refuge within protective vacuoles. Here, we define the host component of the molecular arms race as an evolutionarily conserved polar "hot spot" on the PH domain of ELMO1 (Engulfment and Cell Motility protein 1), which is targeted by diverse WxxxE effectors. Using homology modeling and site-directed mutagenesis, we show that a lysine triad within the "patch" directly binds all WxxxE effectors tested: SifA (Salmonella), IpgB1 and IpgB2 (Shigella), and Map (enteropathogenic Escherichia coli). Using an integrated SifA-host protein-protein interaction network, in silico network perturbation, and functional studies, we show that the major consequences of preventing SifA-ELMO1 interaction are reduced Rac1 activity and microbial invasion. That multiple effectors of diverse structure, function, and sequence bind the same hot spot on ELMO1 suggests that the WxxxE effector(s)-ELMO1 interface is a convergence point of intrusion detection and/or host vulnerability. We conclude that the interface may represent the fault line in coevolved molecular adaptations between pathogens and the host, and its disruption may serve as a therapeutic strategy.
Collapse
Affiliation(s)
- Mahitha Shree Anandachar
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA; Department of Pathology, University of California San Diego, San Diego, California, USA
| | - Suchismita Roy
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA
| | - Saptarshi Sinha
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA
| | - Agyekum Boadi
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA
| | - Gajanan D Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA.
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA; Department of Medicine, University of California San Diego, San Diego, California, USA.
| |
Collapse
|
6
|
Anandachar MS, Roy S, Sinha S, Agyekum B, Ibeawuchi SR, Gementera H, Amamoto A, Katkar GD, Ghosh P. Diverse Gut Pathogens Exploit the Host Engulfment Pathway via a Conserved Mechanism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.09.536168. [PMID: 37066267 PMCID: PMC10104235 DOI: 10.1101/2023.04.09.536168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Macrophages clear infections by engulfing and digesting pathogens within phagolysosomes. Pathogens escape this fate by engaging in a molecular arms race; they use WxxxE motif-containing effector proteins to subvert the host cells they invade and seek refuge within protective vacuoles. Here we define the host component of the molecular arms race as an evolutionarily conserved polar hotspot on the PH-domain of ELMO1 (Engulfment and Cell Motility1), which is targeted by diverse WxxxE-effectors. Using homology modeling and site-directed mutagenesis, we show that a lysine triad within the patch directly binds all WxxxE-effectors tested: SifA (Salmonella), IpgB1 and IpgB2 (Shigella), and Map (enteropathogenic E. coli). Using an integrated SifA-host protein-protein interaction (PPI) network, in-silico network perturbation, and functional studies we show that the major consequences of preventing SifA-ELMO1 interaction are reduced Rac1 activity and microbial invasion. That multiple effectors of diverse structure, function, and sequence bind the same hotpot on ELMO1 suggests that the WxxxE-effector(s)-ELMO1 interface is a convergence point of intrusion detection and/or host vulnerability. We conclude that the interface may represent the fault line in co-evolved molecular adaptations between pathogens and the host and its disruption may serve as a therapeutic strategy.
Collapse
|
7
|
Dadlani E, Dash T, Sahoo D. An AI-assisted Investigation of Tumor-Associated Macrophages and their Polarization in Colorectal Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551559. [PMID: 37577482 PMCID: PMC10418212 DOI: 10.1101/2023.08.01.551559] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Tumor-associated Macrophages (or TAMs) are amongst the most common cells that play a significant role in the initiation and progression of colorectal cancer (CRC). Recently, Ghosh et al. proposed distinguishing signatures for identifying macrophage polarization states, namely, immuno-reactive and immuno-tolerant, using the concept of Boolean implications and Boolean networks. Their signature, called the Signature of Macrophage Reactivity and Tolerance (SMaRT), comprises of 338 human genes (equivalently, 298 mouse genes). However, SMaRT was constructed using datasets that were not specialized towards any particular disease. In this paper, (a) we perform a comprehensive analysis of the SMaRT signature on single-cell human and mouse colorectal cancer RNA-seq datasets; (b) we then adopt a technique akin to transfer learning to construct a "refined" SMaRT signature for investigating TAMs and their polarization in the CRC tumor microenvironment. Towards validation of our refined gene signature, we use (a) 5 pseudo-bulk RNA-seq datasets derived from single-cell human datasets; and (b) 5 large-cohort microarray datasets from humans. Furthermore, we investigate the translational potential of our refined gene signature in problems related to MSS/MSI (4 datasets) and CIMP+/CIMP- status (4 datasets). Overall, our refined gene signature and its extensive validation provide a path for its adoption in clinical practice in diagnosing colorectal cancer and associated attributes.
Collapse
|
8
|
Ghosh P, Sinha S, Katkar GD, Vo D, Taheri S, Dang D, Das S, Sahoo D. Machine learning identifies signatures of macrophage reactivity and tolerance that predict disease outcomes. EBioMedicine 2023; 94:104719. [PMID: 37516087 PMCID: PMC10388732 DOI: 10.1016/j.ebiom.2023.104719] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND Single-cell transcriptomic studies have greatly improved organ-specific insights into macrophage polarization states are essential for the initiation and resolution of inflammation in all tissues; however, such insights are yet to translate into therapies that can predictably alter macrophage fate. METHOD Using machine learning algorithms on human macrophages, here we reveal the continuum of polarization states that is shared across diverse contexts. A path, comprised of 338 genes accurately identified both physiologic and pathologic spectra of "reactivity" and "tolerance", and remained relevant across tissues, organs, species, and immune cells (>12,500 diverse datasets). FINDINGS This 338-gene signature identified macrophage polarization states at single-cell resolution, in physiology and across diverse human diseases, and in murine pre-clinical disease models. The signature consistently outperformed conventional signatures in the degree of transcriptome-proteome overlap, and in detecting disease states; it also prognosticated outcomes across diverse acute and chronic diseases, e.g., sepsis, liver fibrosis, aging, and cancers. Crowd-sourced genetic and pharmacologic studies confirmed that model-rationalized interventions trigger predictable macrophage fates. INTERPRETATION These findings provide a formal and universally relevant definition of macrophage states and a predictive framework (http://hegemon.ucsd.edu/SMaRT) for the scientific community to develop macrophage-targeted precision diagnostics and therapeutics. FUNDING This work was supported by the National Institutes for Health (NIH) grant R01-AI155696 (to P.G, D.S and S.D). Other sources of support include: R01-GM138385 (to D.S), R01-AI141630 (to P.G), R01-DK107585 (to S.D), and UG3TR003355 (to D.S, S.D, and P.G). D.S was also supported by two Padres Pedal the Cause awards (Padres Pedal the Cause/RADY #PTC2017 and San Diego NCI Cancer Centers Council (C3) #PTC2017). S.S, G.D.K, and D.D were supported through The American Association of Immunologists (AAI) Intersect Fellowship Program for Computational Scientists and Immunologists. We also acknowledge support from the Padres Pedal the Cause #PTC2021 and the Torey Coast Foundation, La Jolla (P.G and D.S). D.S, P.G, and S.D were also supported by the Leona M. and Harry B. Helmsley Charitable Trust.
Collapse
Affiliation(s)
- Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, USA; Department of Medicine, University of California San Diego, USA; Moores Cancer Center, University of California San Diego, USA.
| | - Saptarshi Sinha
- Department of Cellular and Molecular Medicine, University of California San Diego, USA; Department of Pediatrics, University of California San Diego, USA
| | - Gajanan D Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, USA
| | - Daniella Vo
- Department of Pediatrics, University of California San Diego, USA
| | - Sahar Taheri
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, USA
| | - Dharanidhar Dang
- Department of Pediatrics, University of California San Diego, USA
| | - Soumita Das
- Moores Cancer Center, University of California San Diego, USA; Department of Pathology, University of California San Diego, USA
| | - Debashis Sahoo
- Moores Cancer Center, University of California San Diego, USA; Department of Pediatrics, University of California San Diego, USA; Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, USA.
| |
Collapse
|
9
|
Zage PE, Huo Y, Subramonian D, Le Clorennec C, Ghosh P, Sahoo D. Identification of a novel gene signature for neuroblastoma differentiation using a Boolean implication network. Genes Chromosomes Cancer 2023; 62:313-331. [PMID: 36680522 PMCID: PMC10257350 DOI: 10.1002/gcc.23124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
Although induction of differentiation represents an effective strategy for neuroblastoma treatment, the mechanisms underlying neuroblastoma differentiation are poorly understood. We generated a computational model of neuroblastoma differentiation consisting of interconnected gene clusters identified based on symmetric and asymmetric gene expression relationships. We identified a differentiation signature consisting of series of gene clusters comprised of 1251 independent genes that predicted neuroblastoma differentiation in independent datasets and in neuroblastoma cell lines treated with agents known to induce differentiation. This differentiation signature was associated with patient outcomes in multiple independent patient cohorts and validated the role of MYCN expression as a marker of neuroblastoma differentiation. Our results further identified novel genes associated with MYCN via asymmetric Boolean implication relationships that would not have been identified using symmetric computational approaches and that were associated with both neuroblastoma differentiation and patient outcomes. Our differentiation signature included a cluster of genes involved in intracellular signaling and growth factor receptor trafficking pathways that is strongly associated with neuroblastoma differentiation, and we validated the associations of UBE4B, a gene within this cluster, with neuroblastoma cell and tumor differentiation. Our findings demonstrate that Boolean network analyses of symmetric and asymmetric gene expression relationships can identify novel genes and pathways relevant for neuroblastoma tumor differentiation that could represent potential therapeutic targets.
Collapse
Affiliation(s)
- Peter E. Zage
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego (UCSD), La Jolla, CA
| | - Yuchen Huo
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego (UCSD), La Jolla, CA
| | - Divya Subramonian
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego (UCSD), La Jolla, CA
| | - Christophe Le Clorennec
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego (UCSD), La Jolla, CA
| | - Pradipta Ghosh
- Department of Medicine, UCSD, La Jolla, CA
- Department of Cellular and Molecular Medicine, UCSD, La Jolla, CA
- Veterans Affairs Medical Center, La Jolla, CA
| | - Debashis Sahoo
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego (UCSD), La Jolla, CA
- Department of Computer Science and Engineering, Jacobs School of Engineering, UCSD, La Jolla, CA
| |
Collapse
|
10
|
Ye Q, Guo NL. Hub Genes in Non-Small Cell Lung Cancer Regulatory Networks. Biomolecules 2022; 12:1782. [PMID: 36551208 PMCID: PMC9776006 DOI: 10.3390/biom12121782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022] Open
Abstract
There are currently no accurate biomarkers for optimal treatment selection in early-stage non-small cell lung cancer (NSCLC). Novel therapeutic targets are needed to improve NSCLC survival outcomes. This study systematically evaluated the association between genome-scale regulatory network centralities and NSCLC tumorigenesis, proliferation, and survival in early-stage NSCLC patients. Boolean implication networks were used to construct multimodal networks using patient DNA copy number variation, mRNA, and protein expression profiles. T statistics of differential gene/protein expression in tumors versus non-cancerous adjacent tissues, dependency scores in in vitro CRISPR-Cas9/RNA interference (RNAi) screening of human NSCLC cell lines, and hazard ratios in univariate Cox modeling of the Cancer Genome Atlas (TCGA) NSCLC patients were correlated with graph theory centrality metrics. Hub genes in multi-omics networks involving gene/protein expression were associated with oncogenic, proliferative potentials and poor patient survival outcomes (p < 0.05, Pearson's correlation). Immunotherapy targets PD1, PDL1, CTLA4, and CD27 were ranked as top hub genes within the 10th percentile in most constructed multi-omics networks. BUB3, DNM1L, EIF2S1, KPNB1, NMT1, PGAM1, and STRAP were discovered as important hub genes in NSCLC proliferation with oncogenic potential. These results support the importance of hub genes in NSCLC tumorigenesis, proliferation, and prognosis, with implications in prioritizing therapeutic targets to improve patient survival outcomes.
Collapse
Affiliation(s)
- Qing Ye
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
- Lane Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Nancy Lan Guo
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
11
|
Subramanian R, Sahoo D. Boolean implication analysis of single-cell data predicts retinal cell type markers. BMC Bioinformatics 2022; 23:378. [PMID: 36114457 PMCID: PMC9482279 DOI: 10.1186/s12859-022-04915-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/25/2022] [Indexed: 11/15/2022] Open
Abstract
Background The retina is a complex tissue containing multiple cell types that are essential for vision. Understanding the gene expression patterns of various retinal cell types has potential applications in regenerative medicine. Retinal organoids (optic vesicles) derived from pluripotent stem cells have begun to yield insights into the transcriptomics of developing retinal cell types in humans through single cell RNA-sequencing studies. Previous methods of gene reporting have relied upon techniques in vivo using microarray data, or correlational and dimension reduction methods for analyzing single cell RNA-sequencing data computationally. We aimed to develop a state-of-the-art Boolean method that filtered out noise, could be applied to a wide variety of datasets and lent insight into gene expression over differentiation. Results Here, we present a bioinformatic approach using Boolean implication to discover genes which are retinal cell type-specific or involved in retinal cell fate. We apply this approach to previously published retina and retinal organoid datasets and improve upon previously published correlational methods. Our method improves the prediction accuracy of marker genes of retinal cell types and discovers several new high confidence cone and rod-specific genes. Conclusions The results of this study demonstrate the benefits of a Boolean approach that considers asymmetric relationships. We have shown a statistically significant improvement from correlational, symmetric methods in the prediction accuracy of retinal cell-type specific genes. Furthermore, our method contains no cell or tissue-specific tuning and hence could impact other areas of gene expression analyses in cancer and other human diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04915-4.
Collapse
|
12
|
Stobdan T, Sahoo D, Haddad GG. A Boolean approach for novel hypoxia-related gene discovery. PLoS One 2022; 17:e0273524. [PMID: 36006949 PMCID: PMC9409593 DOI: 10.1371/journal.pone.0273524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/09/2022] [Indexed: 11/19/2022] Open
Abstract
Hypoxia plays a major role in the etiology and pathogenesis of most of the leading causes of morbidity and mortality, whether cardiovascular diseases, cancer, respiratory diseases or stroke. Despite active research on hypoxia-signaling pathways, the understanding of regulatory mechanisms, especially in specific tissues, still remain elusive. With the accessibility of thousands of potentially diverse genomic datasets, computational methods are utilized to generate new hypotheses. Here we utilized Boolean implication relationship, a powerful method to probe symmetrically and asymmetrically related genes, to identify novel hypoxia related genes. We used a well-known hypoxia-responsive gene, VEGFA, with very large human expression datasets (n = 25,955) to identify novel hypoxia-responsive candidate gene/s. Further, we utilized in-vitro analysis using human endothelial cells exposed to 1% O2 environment for 2, 8, 24 and 48 hours to validate top candidate genes. Out of the top candidate genes (n = 19), 84% genes were previously reported as hypoxia related, validating our results. However, we identified FAM114A1 as a novel candidate gene significantly upregulated in the endothelial cells at 8, 24 and 48 hours of 1% O2 environment. Additional evidence, particularly the localization of intronic miRNA and numerous HREs further support and strengthen our finding. Current results on FAM114A1 provide an example demonstrating the utility of powerful computational methods, like Boolean implications, in playing a major role in hypothesis building and discovery.
Collapse
Affiliation(s)
- Tsering Stobdan
- Department of Pediatrics, Division of Respiratory Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Debashis Sahoo
- Department of Pediatrics, Division of Respiratory Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, United States of America
| | - Gabriel G. Haddad
- Department of Pediatrics, Division of Respiratory Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Rady Children’s Hospital, San Diego, California, United States of America
| |
Collapse
|
13
|
Sinha S, Castillo V, Espinoza CR, Tindle C, Fonseca AG, Dan JM, Katkar GD, Das S, Sahoo D, Ghosh P. COVID-19 lung disease shares driver AT2 cytopathic features with Idiopathic pulmonary fibrosis. EBioMedicine 2022; 82:104185. [PMID: 35870428 PMCID: PMC9297827 DOI: 10.1016/j.ebiom.2022.104185] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/06/2022] [Accepted: 07/06/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND In the aftermath of Covid-19, some patients develop a fibrotic lung disease, i.e., post-COVID-19 lung disease (PCLD), for which we currently lack insights into pathogenesis, disease models, or treatment options. METHODS Using an AI-guided approach, we analyzed > 1000 human lung transcriptomic datasets associated with various lung conditions using two viral pandemic signatures (ViP and sViP) and one covid lung-derived signature. Upon identifying similarities between COVID-19 and idiopathic pulmonary fibrosis (IPF), we subsequently dissected the basis for such similarity from molecular, cytopathic, and immunologic perspectives using a panel of IPF-specific gene signatures, alongside signatures of alveolar type II (AT2) cytopathies and of prognostic monocyte-driven processes that are known drivers of IPF. Transcriptome-derived findings were used to construct protein-protein interaction (PPI) network to identify the major triggers of AT2 dysfunction. Key findings were validated in hamster and human adult lung organoid (ALO) pre-clinical models of COVID-19 using immunohistochemistry and qPCR. FINDINGS COVID-19 resembles IPF at a fundamental level; it recapitulates the gene expression patterns (ViP and IPF signatures), cytokine storm (IL15-centric), and the AT2 cytopathic changes, e.g., injury, DNA damage, arrest in a transient, damage-induced progenitor state, and senescence-associated secretory phenotype (SASP). These immunocytopathic features were induced in pre-clinical COVID models (ALO and hamster) and reversed with effective anti-CoV-2 therapeutics in hamsters. PPI-network analyses pinpointed ER stress as one of the shared early triggers of both diseases, and IHC studies validated the same in the lungs of deceased subjects with COVID-19 and SARS-CoV-2-challenged hamster lungs. Lungs from tg-mice, in which ER stress is induced specifically in the AT2 cells, faithfully recapitulate the host immune response and alveolar cytopathic changes that are induced by SARS-CoV-2. INTERPRETATION Like IPF, COVID-19 may be driven by injury-induced ER stress that culminates into progenitor state arrest and SASP in AT2 cells. The ViP signatures in monocytes may be key determinants of prognosis. The insights, signatures, disease models identified here are likely to spur the development of therapies for patients with IPF and other fibrotic interstitial lung diseases. FUNDING This work was supported by the National Institutes for Health grants R01- GM138385 and AI155696 and funding from the Tobacco-Related disease Research Program (R01RG3780).
Collapse
Affiliation(s)
- Saptarshi Sinha
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Vanessa Castillo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Celia R Espinoza
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Courtney Tindle
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Ayden G Fonseca
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jennifer M Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Gajanan D Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Soumita Das
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | - Debashis Sahoo
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA 92093, USA.
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
14
|
Sinha S, Castillo V, Espinoza CR, Tindle C, Fonseca AG, Dan JM, Katkar GD, Das S, Sahoo D, Ghosh P. COVID-19 lung disease shares driver AT2 cytopathic features with Idiopathic pulmonary fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.11.28.470269. [PMID: 34873597 PMCID: PMC8647648 DOI: 10.1101/2021.11.28.470269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Background In the aftermath of Covid-19, some patients develop a fibrotic lung disease, i.e., p ost- C OVID-19 l ung d isease (PCLD), for which we currently lack insights into pathogenesis, disease models, or treatment options. Method Using an AI-guided approach, we analyzed > 1000 human lung transcriptomic datasets associated with various lung conditions using two viral pandemic signatures (ViP and sViP) and one covid lung-derived signature. Upon identifying similarities between COVID-19 and idiopathic pulmonary fibrosis (IPF), we subsequently dissected the basis for such similarity from molecular, cytopathic, and immunologic perspectives using a panel of IPF-specific gene signatures, alongside signatures of alveolar type II (AT2) cytopathies and of prognostic monocyte-driven processes that are known drivers of IPF. Transcriptome-derived findings were used to construct protein-protein interaction (PPI) network to identify the major triggers of AT2 dysfunction. Key findings were validated in hamster and human adult lung organoid (ALO) pre-clinical models of COVID-19 using immunohistochemistry and qPCR. Findings COVID-19 resembles IPF at a fundamental level; it recapitulates the gene expression patterns (ViP and IPF signatures), cytokine storm (IL15-centric), and the AT2 cytopathic changes, e.g., injury, DNA damage, arrest in a transient, damage-induced progenitor state, and senescence-associated secretory phenotype (SASP). These immunocytopathic features were induced in pre-clinical COVID models (ALO and hamster) and reversed with effective anti-CoV-2 therapeutics in hamsters. PPI-network analyses pinpointed ER stress as one of the shared early triggers of both diseases, and IHC studies validated the same in the lungs of deceased subjects with COVID-19 and SARS-CoV-2-challenged hamster lungs. Lungs from tg - mice, in which ER stress is induced specifically in the AT2 cells, faithfully recapitulate the host immune response and alveolar cytopathic changes that are induced by SARS-CoV-2. Interpretation Like IPF, COVID-19 may be driven by injury-induced ER stress that culminates into progenitor state arrest and SASP in AT2 cells. The ViP signatures in monocytes may be key determinants of prognosis. The insights, signatures, disease models identified here are likely to spur the development of therapies for patients with IPF and other fibrotic interstitial lung diseases. Funding This work was supported by the National Institutes for Health grants R01-GM138385 and AI155696 and funding from the Tobacco-Related disease Research Program (R01RG3780). One Sentence Summary Severe COVID-19 triggers cellular processes seen in fibrosing Interstitial Lung Disease. RESEARCH IN CONTEXT Evidence before this study: In its aftermath, the COVID-19 pandemic has left many survivors, almost a third of those who recovered, with a mysterious long-haul form of the disease which culminates in a fibrotic form of interstitial lung disease (post-COVID-19 ILD). Post-COVID-19 ILD remains a largely unknown entity. Currently, we lack insights into the core cytopathic features that drive this condition.Added value of this study: Using an AI-guided approach, which involves the use of sets of gene signatures, protein-protein network analysis, and a hamster model of COVID-19, we have revealed here that COVID-19 -lung fibrosis resembles IPF, the most common form of ILD, at a fundamental levelâ€"showing similar gene expression patterns in the lungs and blood, and dysfunctional AT2 processes (ER stress, telomere instability, progenitor cell arrest, and senescence). These findings are insightful because AT2 cells are known to contain an elegant quality control network to respond to intrinsic or extrinsic stress; a failure of such quality control results in diverse cellular phenotypes, of which ER stress appears to be a point of convergence, which appears to be sufficient to drive downstream fibrotic remodeling in the lung.Implications of all the available evidence: Because unbiased computational methods identified the shared fundamental aspects of gene expression and cellular processes between COVID-19 and IPF, the impact of our findings is likely to go beyond COVID-19 or any viral pandemic. The insights, tools (disease models, gene signatures, and biomarkers), and mechanisms identified here are likely to spur the development of therapies for patients with IPF and, other fibrotic interstitial lung diseases, all of whom have limited or no treatment options. To dissect the validated prognostic biomarkers to assess and track the risk of pulmonary fibrosis and develop therapeutics to halt fibrogenic progression.
Collapse
|
15
|
Katkar GD, Sayed IM, Anandachar MS, Castillo V, Vidales E, Toobian D, Usmani F, Sawires JR, Leriche G, Yang J, Sandborn WJ, Das S, Sahoo D, Ghosh P. Artificial intelligence-rationalized balanced PPARα/γ dual agonism resets dysregulated macrophage processes in inflammatory bowel disease. Commun Biol 2022; 5:231. [PMID: 35288651 PMCID: PMC8921270 DOI: 10.1038/s42003-022-03168-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
A computational platform, Boolean network explorer (BoNE), has recently been developed to infuse AI-enhanced precision into drug discovery; it enables invariant Boolean Implication Networks of disease maps for prioritizing high-value targets. Here we used BoNE to query an Inflammatory Bowel Disease (IBD)-map and prioritize a therapeutic strategy that involves dual agonism of two nuclear receptors, PPARα/γ. Balanced agonism of PPARα/γ was predicted to modulate macrophage processes, ameliorate colitis, 'reset' the gene expression network from disease to health. Predictions were validated using a balanced and potent PPARα/γ-dual-agonist (PAR5359) in Citrobacter rodentium- and DSS-induced murine colitis models. Using inhibitors and agonists, we show that balanced-dual agonism promotes bacterial clearance efficiently than individual agonists, both in vivo and in vitro. PPARα is required and sufficient to induce the pro-inflammatory cytokines and cellular ROS, which are essential for bacterial clearance and immunity, whereas PPARγ-agonism blunts these responses, delays microbial clearance; balanced dual agonism achieved controlled inflammation while protecting the gut barrier and 'reversal' of the transcriptomic network. Furthermore, dual agonism reversed the defective bacterial clearance observed in PBMCs derived from IBD patients. These findings not only deliver a macrophage modulator for use as barrier-protective therapy in IBD, but also highlight the potential of BoNE to rationalize combination therapy.
Collapse
Affiliation(s)
- Gajanan D Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, USA
| | - Ibrahim M Sayed
- Department of Pathology, University of California San Diego, San Diego, USA.,Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | | | - Vanessa Castillo
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, USA
| | - Eleadah Vidales
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, USA
| | - Daniel Toobian
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, USA
| | - Fatima Usmani
- Department of Pathology, University of California San Diego, San Diego, USA
| | - Joseph R Sawires
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, USA
| | - Geoffray Leriche
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, USA
| | - Jerry Yang
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, USA
| | - William J Sandborn
- Department of Medicine, University of California San Diego, San Diego, USA.
| | - Soumita Das
- Department of Pathology, University of California San Diego, San Diego, USA.
| | - Debashis Sahoo
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, San Diego, USA. .,Department of Pediatrics, University of California San Diego, San Diego, USA. .,Rebecca and John Moore Comprehensive Cancer Center, University of California San Diego, San Diego, USA.
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, USA. .,Department of Medicine, University of California San Diego, San Diego, USA. .,Rebecca and John Moore Comprehensive Cancer Center, University of California San Diego, San Diego, USA. .,Veterans Affairs Medical Center, La Jolla, San Diego, USA.
| |
Collapse
|
16
|
Ye Q, Singh S, Qian PR, Guo NL. Immune-Omics Networks of CD27, PD1, and PDL1 in Non-Small Cell Lung Cancer. Cancers (Basel) 2021; 13:4296. [PMID: 34503105 PMCID: PMC8428355 DOI: 10.3390/cancers13174296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 01/03/2023] Open
Abstract
To date, there are no prognostic/predictive biomarkers to select chemotherapy, immunotherapy, and radiotherapy in individual non-small cell lung cancer (NSCLC) patients. Major immune-checkpoint inhibitors (ICIs) have more DNA copy number variations (CNV) than mutations in The Cancer Genome Atlas (TCGA) NSCLC tumors. Nevertheless, CNV-mediated dysregulated gene expression in NSCLC is not well understood. Integrated CNV and transcriptional profiles in NSCLC tumors (n = 371) were analyzed using Boolean implication networks for the identification of a multi-omics CD27, PD1, and PDL1 network, containing novel prognostic genes and proliferation genes. A 5-gene (EIF2AK3, F2RL3, FOSL1, SLC25A26, and SPP1) prognostic model was developed and validated for patient stratification (p < 0.02, Kaplan-Meier analyses) in NSCLC tumors (n = 1163). A total of 13 genes (COPA, CSE1L, EIF2B3, LSM3, MCM5, PMPCB, POLR1B, POLR2F, PSMC3, PSMD11, RPL32, RPS18, and SNRPE) had a significant impact on proliferation in 100% of the NSCLC cell lines in both CRISPR-Cas9 (n = 78) and RNA interference (RNAi) assays (n = 92). Multiple identified genes were associated with chemoresponse and radiotherapy response in NSCLC cell lines (n = 117) and patient tumors (n = 966). Repurposing drugs were discovered based on this immune-omics network to improve NSCLC treatment.
Collapse
Affiliation(s)
- Qing Ye
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (S.S.); (P.R.Q.)
- Lane Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Salvi Singh
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (S.S.); (P.R.Q.)
- Lane Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Peter R. Qian
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (S.S.); (P.R.Q.)
| | - Nancy Lan Guo
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (S.S.); (P.R.Q.)
- Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
17
|
Reynoso S, Castillo V, Katkar GD, Lopez-Sanchez I, Taheri S, Espinoza C, Rohena C, Sahoo D, Gagneux P, Ghosh P. GIV/Girdin, a non-receptor modulator for Gαi/s, regulates spatiotemporal signaling during sperm capacitation and is required for male fertility. eLife 2021; 10:69160. [PMID: 34409938 PMCID: PMC8376251 DOI: 10.7554/elife.69160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
For a sperm to successfully fertilize an egg, it must first undergo capacitation in the female reproductive tract and later undergo acrosomal reaction (AR) upon encountering an egg surrounded by its vestment. How premature AR is avoided despite rapid surges in signaling cascades during capacitation remains unknown. Using a combination of conditional knockout (cKO) mice and cell-penetrating peptides, we show that GIV (CCDC88A), a guanine nucleotide-exchange modulator (GEM) for trimeric GTPases, is highly expressed in spermatocytes and is required for male fertility. GIV is rapidly phosphoregulated on key tyrosine and serine residues in human and murine spermatozoa. These phosphomodifications enable GIV-GEM to orchestrate two distinct compartmentalized signaling programs in the sperm tail and head; in the tail, GIV enhances PI3K→Akt signals, sperm motility and survival, whereas in the head it inhibits cAMP surge and premature AR. Furthermore, GIV transcripts are downregulated in the testis and semen of infertile men. These findings exemplify the spatiotemporally segregated signaling programs that support sperm capacitation and shed light on a hitherto unforeseen cause of infertility in men.
Collapse
Affiliation(s)
- Sequoyah Reynoso
- Department of Pathology, School of Medicine, University of California San Diego, San Diego, United States
| | - Vanessa Castillo
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, San Diego, United States
| | - Gajanan Dattatray Katkar
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, San Diego, United States
| | - Inmaculada Lopez-Sanchez
- Department of Medicine, School of Medicine, University of California San Diego, San Diego, United States
| | - Sahar Taheri
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, San Diego, United States
| | - Celia Espinoza
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, San Diego, United States
| | - Cristina Rohena
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, San Diego, United States
| | - Debashis Sahoo
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, San Diego, United States.,Moore's Comprehensive Cancer Center, University of California San Diego, San Diego, United States.,Department of Pediatrics, School of Medicine, University of California San Diego, San Diego, United States
| | - Pascal Gagneux
- Department of Pathology, School of Medicine, University of California San Diego, San Diego, United States
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, San Diego, United States.,Department of Medicine, School of Medicine, University of California San Diego, San Diego, United States.,Moore's Comprehensive Cancer Center, University of California San Diego, San Diego, United States.,Veterans Affairs Medical Center, Washington DC, United States
| |
Collapse
|
18
|
Artificial intelligence guided discovery of a barrier-protective therapy in inflammatory bowel disease. Nat Commun 2021; 12:4246. [PMID: 34253728 PMCID: PMC8275683 DOI: 10.1038/s41467-021-24470-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/21/2021] [Indexed: 12/19/2022] Open
Abstract
Modeling human diseases as networks simplify complex multi-cellular processes, helps understand patterns in noisy data that humans cannot find, and thereby improves precision in prediction. Using Inflammatory Bowel Disease (IBD) as an example, here we outline an unbiased AI-assisted approach for target identification and validation. A network was built in which clusters of genes are connected by directed edges that highlight asymmetric Boolean relationships. Using machine-learning, a path of continuum states was pinpointed, which most effectively predicted disease outcome. This path was enriched in gene-clusters that maintain the integrity of the gut epithelial barrier. We exploit this insight to prioritize one target, choose appropriate pre-clinical murine models for target validation and design patient-derived organoid models. Potential for treatment efficacy is confirmed in patient-derived organoids using multivariate analyses. This AI-assisted approach identifies a first-in-class gut barrier-protective agent in IBD and predicted Phase-III success of candidate agents. Traditional drug discovery process use differential, Bayesian and other network based approaches. We developed a Boolean approach for building disease maps and prioritizing pre-clinical models to discover a first-in-class therapy to restore and protect the leaky gut barrier in inflammatory bowel disease.
Collapse
|
19
|
Sahoo D, Katkar GD, Khandelwal S, Behroozikhah M, Claire A, Castillo V, Tindle C, Fuller M, Taheri S, Rogers TF, Beutler N, Ramirez SI, Rawlings SA, Pretorius V, Smith DM, Burton DR, Alexander LEC, Duran J, Crotty S, Dan JM, Das S, Ghosh P. AI-guided discovery of the invariant host response to viral pandemics. EBioMedicine 2021; 68:103390. [PMID: 34127431 PMCID: PMC8193764 DOI: 10.1016/j.ebiom.2021.103390] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Coronavirus Disease 2019 (Covid-19) continues to challenge the limits of our knowledge and our healthcare system. Here we sought to define the host immune response, a.k.a, the "cytokine storm" that has been implicated in fatal COVID-19 using an AI-based approach. METHOD Over 45,000 transcriptomic datasets of viral pandemics were analyzed to extract a 166-gene signature using ACE2 as a 'seed' gene; ACE2 was rationalized because it encodes the receptor that facilitates the entry of SARS-CoV-2 (the virus that causes COVID-19) into host cells. An AI-based approach was used to explore the utility of the signature in navigating the uncharted territory of Covid-19, setting therapeutic goals, and finding therapeutic solutions. FINDINGS The 166-gene signature was surprisingly conserved across all viral pandemics, including COVID-19, and a subset of 20-genes classified disease severity, inspiring the nomenclatures ViP and severe-ViP signatures, respectively. The ViP signatures pinpointed a paradoxical phenomenon wherein lung epithelial and myeloid cells mount an IL15 cytokine storm, and epithelial and NK cell senescence and apoptosis determine severity/fatality. Precise therapeutic goals could be formulated; these goals were met in high-dose SARS-CoV-2-challenged hamsters using either neutralizing antibodies that abrogate SARS-CoV-2•ACE2 engagement or a directly acting antiviral agent, EIDD-2801. IL15/IL15RA were elevated in the lungs of patients with fatal disease, and plasma levels of the cytokine prognosticated disease severity. INTERPRETATION The ViP signatures provide a quantitative and qualitative framework for titrating the immune response in viral pandemics and may serve as a powerful unbiased tool to rapidly assess disease severity and vet candidate drugs. FUNDING This work was supported by the National Institutes for Health (NIH) [grants CA151673 and GM138385 (to DS) and AI141630 (to P.G), DK107585-05S1 (SD) and AI155696 (to P.G, D.S and S.D), U19-AI142742 (to S. C, CCHI Cooperative Centers for Human Immunology)]; Research Grants Program Office (RGPO) from the University of California Office of the President (UCOP) (R00RG2628 & R00RG2642 to P.G, D.S and S.D); the UC San Diego Sanford Stem Cell Clinical Center (to P.G, D.S and S.D); LJI Institutional Funds (to S.C); the VA San Diego Healthcare System Institutional funds (to L.C.A). GDK was supported through The American Association of Immunologists Intersect Fellowship Program for Computational Scientists and Immunologists. ONE SENTENCE SUMMARY The host immune response in COVID-19.
Collapse
Affiliation(s)
- Debashis Sahoo
- Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, MC 0730, Leichtag Building 132, La Jolla, CA 92093-0831, USA; Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San Diego, USA; Moores Cancer Center, University of California San Diego, USA.
| | - Gajanan D Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, USA
| | - Soni Khandelwal
- Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, MC 0730, Leichtag Building 132, La Jolla, CA 92093-0831, USA
| | - Mahdi Behroozikhah
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San Diego, USA
| | - Amanraj Claire
- Department of Cellular and Molecular Medicine, University of California San Diego, USA
| | - Vanessa Castillo
- Department of Cellular and Molecular Medicine, University of California San Diego, USA
| | - Courtney Tindle
- Department of Cellular and Molecular Medicine, University of California San Diego, USA
| | - MacKenzie Fuller
- Department of Cellular and Molecular Medicine, University of California San Diego, USA
| | - Sahar Taheri
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San Diego, USA
| | - Thomas F Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sydney I Ramirez
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Stephen A Rawlings
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | | | - Davey M Smith
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Dennis R Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Laura E Crotty Alexander
- Pulmonary Critical Care Section, Veterans Affairs (VA) San Diego Healthcare System, La Jolla, California; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Jason Duran
- Division of Cardiology, Department of Internal Medicine, UC San Diego Medical Center, La Jolla 92037
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Jennifer M Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Soumita Das
- Department of Pathology, University of California San Diego, USA.
| | - Pradipta Ghosh
- Moores Cancer Center, University of California San Diego, USA; Department of Cellular and Molecular Medicine, University of California San Diego, USA; Medicine, University of California San Diego, USA.
| |
Collapse
|
20
|
Vo D, Singh SC, Safa S, Sahoo D. Boolean implication analysis unveils candidate universal relationships in microbiome data. BMC Bioinformatics 2021; 22:49. [PMID: 33546590 PMCID: PMC7863539 DOI: 10.1186/s12859-020-03941-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/16/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Microbiomes consist of bacteria, viruses, and other microorganisms, and are responsible for many different functions in both organisms and the environment. Past analyses of microbiomes focused on using correlation to determine linear relationships between microbes and diseases. Weak correlations due to nonlinearity between microbe pairs may cause researchers to overlook critical components of the data. With the abundance of available microbiome, we need a method that comprehensively studies microbiomes and how they are related to each other. RESULTS We collected publicly available datasets from human, environment, and animal samples to determine both symmetric and asymmetric Boolean implication relationships between a pair of microbes. We then found relationships that are potentially invariants, meaning they will hold in any microbe community. In other words, if we determine there is a relationship between two microbes, we expect the relationship to hold in almost all contexts. We discovered that around 330,000 pairs of microbes universally exhibit the same relationship in almost all the datasets we studied, thus making them good candidates for invariants. Our results also confirm known biological properties and seem promising in terms of disease diagnosis. CONCLUSIONS Since the relationships are likely universal, we expect them to hold in clinical settings, as well as general populations. If these strong invariants are present in disease settings, it may provide insight into prognostic, predictive, or therapeutic properties of clinically relevant diseases. For example, our results indicate that there is a difference in the microbe distributions between patients who have or do not have IBD, eczema and psoriasis. These new analyses may improve disease diagnosis and drug development in terms of accuracy and efficiency.
Collapse
Affiliation(s)
- Daniella Vo
- Department of Bioinformatics and Systems Biology, University of California San Diego, La Jolla, CA, 92093-083, USA
| | - Shayal Charisma Singh
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, 92093-083, USA
| | - Sara Safa
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, La Jolla, CA, 92093-083, USA
| | - Debashis Sahoo
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, La Jolla, CA, 92093-083, USA.
- Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, MC 0730, Leichtag Building 132, La Jolla, CA, 92093-083, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093-083, USA.
| |
Collapse
|
21
|
Dang D, Taheri S, Das S, Ghosh P, Prince LS, Sahoo D. Computational Approach to Identifying Universal Macrophage Biomarkers. Front Physiol 2020; 11:275. [PMID: 32322218 PMCID: PMC7156600 DOI: 10.3389/fphys.2020.00275] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 03/10/2020] [Indexed: 12/11/2022] Open
Abstract
Macrophages engulf and digest microbes, cellular debris, and various disease-associated cells throughout the body. Understanding the dynamics of macrophage gene expression is crucial for studying human diseases. As both bulk RNAseq and single cell RNAseq datasets become more numerous and complex, identifying a universal and reliable marker of macrophage cell becomes paramount. Traditional approaches have relied upon tissue specific expression patterns. To identify universal biomarkers of macrophage, we used a previously published computational approach called BECC (Boolean Equivalent Correlated Clusters) that was originally used to identify conserved cell cycle genes. We performed BECC analysis using the known macrophage marker CD14 as a seed gene. The main idea behind BECC is that it uses massive database of public gene expression dataset to establish robust co-expression patterns identified using a combination of correlation, linear regression and Boolean equivalences. Our analysis identified and validated FCER1G and TYROBP as novel universal biomarkers for macrophages in human and mouse tissues.
Collapse
Affiliation(s)
- Dharanidhar Dang
- Department of Computer Science and Engineering, University of California, San Diego, San Diego, CA, United States.,Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
| | - Sahar Taheri
- Department of Computer Science and Engineering, University of California, San Diego, San Diego, CA, United States
| | - Soumita Das
- Department of Pathology, University of California, San Diego, San Diego, CA, United States
| | - Pradipta Ghosh
- Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, United States.,Moores Cancer Center, San Diego, CA, United States
| | - Lawrence S Prince
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States.,Rady Children's Hospital, San Diego, CA, United States
| | - Debashis Sahoo
- Department of Computer Science and Engineering, University of California, San Diego, San Diego, CA, United States.,Department of Pediatrics, University of California, San Diego, San Diego, CA, United States.,Moores Cancer Center, San Diego, CA, United States
| |
Collapse
|
22
|
Sayed IM, Suarez K, Lim E, Singh S, Pereira M, Ibeawuchi SR, Katkar G, Dunkel Y, Mittal Y, Chattopadhyay R, Guma M, Boland BS, Dulai PS, Sandborn WJ, Ghosh P, Das S. Host engulfment pathway controls inflammation in inflammatory bowel disease. FEBS J 2020; 287:3967-3988. [PMID: 32003126 DOI: 10.1111/febs.15236] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 12/20/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022]
Abstract
Chronic diseases, including inflammatory bowel disease (IBD) urgently need new biomarkers as a significant proportion of patients, do not respond to current medications. Inflammation is a common factor in these diseases, and microbial sensing in the intestinal tract is critical to initiate the inflammation. We have identified ELMO1 (engulfment and cell motility protein 1) as a microbial sensor in epithelial and phagocytic cells that turns on inflammatory signals. Using a stem cell-based 'gut-in-a-dish' coculture model, we studied the interactions between microbes, epithelium, and monocytes in the context of IBD. To mimic the in vivo cell physiology, enteroid-derived monolayers (EDMs) were generated from the organoids isolated from WT and ELMO1-/- mice and colonic biopsies of IBD patients. The EDMs were infected with the IBD-associated microbes to monitor the inflammatory responses. ELMO1-depleted EDMs displayed a significant reduction in bacterial internalization, a decrease in pro-inflammatory cytokine productions and monocyte recruitment. The expression of ELMO1 is elevated in the colonic epithelium and in the inflammatory infiltrates within the lamina propria of IBD patients where the higher expression is positively correlated with the elevated expression of pro-inflammatory cytokines, MCP-1 and TNF-α. MCP-1 is released from the epithelium and recruits monocytes to the site of inflammation. Once recruited, monocytes require ELMO1 to engulf the bacteria and propagate a robust TNF-α storm. These findings highlight that the dysregulated epithelial ELMO1 → MCP-1 axis can serve as an early biomarker in the diagnostics of IBD and other inflammatory disorders.
Collapse
Affiliation(s)
- Ibrahim M Sayed
- Department of Pathology, University of California, San Diego, CA, USA
| | - Katherine Suarez
- Department of Pathology, University of California, San Diego, CA, USA
| | - Eileen Lim
- Department of Pathology, University of California, San Diego, CA, USA
| | - Sujay Singh
- Department of Pathology, University of California, San Diego, CA, USA
| | - Matheus Pereira
- Department of Pathology, University of California, San Diego, CA, USA
| | | | - Gajanan Katkar
- Department of Cellular & Molecular Medicine, University of California, San Diego, CA, USA
| | - Ying Dunkel
- Department of Medicine, University of California, San Diego, CA, USA
| | - Yash Mittal
- Department of Medicine, University of California, San Diego, CA, USA
| | - Ranajoy Chattopadhyay
- Department of Cellular & Molecular Medicine, University of California, San Diego, CA, USA
| | - Monica Guma
- Department of Medicine, University of California, San Diego, CA, USA
| | - Brigid S Boland
- Department of Medicine, University of California, San Diego, CA, USA
| | - Parambir S Dulai
- Department of Medicine, University of California, San Diego, CA, USA
| | | | - Pradipta Ghosh
- Department of Medicine, University of California, San Diego, CA, USA.,Department of Cellular & Molecular Medicine, University of California, San Diego, CA, USA
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA, USA
| |
Collapse
|
23
|
Pandey S, Sahoo D. Identification of gene expression logical invariants in Arabidopsis. PLANT DIRECT 2019; 3:e00123. [PMID: 31245766 PMCID: PMC6508763 DOI: 10.1002/pld3.123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/28/2018] [Accepted: 02/01/2019] [Indexed: 06/09/2023]
Abstract
Numerous gene expression datasets from diverse tissue samples from the plant variety Arabidopsis thaliana have been already deposited in the public domain. There have been several attempts to do large scale meta-analyses of all of these datasets. Most of these analyses summarize pairwise gene expression relationships using correlation, or identify differentially expressed genes in two conditions. We propose here a new large scale meta-analysis of the publicly available Arabidopsis datasets to identify Boolean logical relationships between genes. Boolean logic is a branch of mathematics that deals with two possible values. In the context of gene expression datasets we use qualitative high and low expression values. A strong logical relationship between genes emerges if at least one of the quadrants is sparsely populated. We pointed out serious issues in the data normalization steps widely accepted and published recently in this context. We put together a web resource where gene expression relationships can be explored online which helps visualize the logical relationships between genes. We believe that this website will be useful in identifying important genes in different biological context. The web link is http://hegemon.ucsd.edu/plant/.
Collapse
|
24
|
Abstract
TRAF3-interacting protein 3 (TRAF3IP3) is expressed in the immune system and participates in cell maturation, tissue development, and immune response. In a previous study, we reported that TRAF3IP3 levels were substantially increased in the vasculature of breast cancer tissues, suggesting a proangiogenic role. In this study, we investigated TRAF3IP3 tumorigenic function. TRAF3IP3 protein was present in several cancer cell lines, with highest levels in melanoma. In addition, tumor microarray analysis on 23 primary melanoma and nine positive lymph nodes revealed that 70% of human primary melanoma and 66% of lymph node metastases were positive for TRAF3IP3. Importantly, TRAF3IP3 downregulation correlated with an 83% reduction of tumor growth in a subcutaneous xenograft mouse model (n=10, P=0.005). Immunohistochemistry analysis of the tumors revealed that TRAF3IP3-shRNA tumors had increased apoptosis (n=4, P<0.01) and reduced microvascular density (n=4, P<0.002). In addition, TRAF3IP3 downregulation in malignant endothelial cells reduced tube formation in a Matrigel tube formation assay. In melanoma cells, decreased levels of TRAF3IP3 were also associated with reduced viability (n=4, P=0.03) and proliferation (n=3, P=0.03), together with increased sensitivity to ultraviolet-induced apoptosis (n=4, P=0.0004). Furthermore, TRAF3IP3 downregulation correlated with increased amounts of interferon-γ. Interferon-γ inhibits tumor growth and angiogenesis, thus suggesting a new pathway for TRAF3IP3 in cancer. Collectively, the association of TRAF3IP3 with malignant properties of melanoma suggest a clinical potential for targeted therapy.
Collapse
|
25
|
Sahoo D, Wei W, Auman H, Hurtado-Coll A, Carroll PR, Fazli L, Gleave ME, Lin DW, Nelson PS, Simko J, Thompson IM, Leach RJ, Troyer DA, True LD, McKenney JK, Feng Z, Brooks JD. Boolean analysis identifies CD38 as a biomarker of aggressive localized prostate cancer. Oncotarget 2018; 9:6550-6561. [PMID: 29464091 PMCID: PMC5814231 DOI: 10.18632/oncotarget.23973] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/23/2017] [Indexed: 01/19/2023] Open
Abstract
The introduction of serum Prostate Specific Antigen (PSA) testing nearly 30 years ago has been associated with a significant shift towards localized disease and decreased deaths due to prostate cancer. Recognition that PSA testing has caused over diagnosis and over treatment of prostate cancer has generated considerable controversy over its value, and has spurred efforts to identify prognostic biomarkers to distinguish patients who need treatment from those that can be observed. Recent studies show that cancer is heterogeneous and forms a hierarchy of tumor cell populations. We developed a method of identifying prostate cancer differentiation states related to androgen signaling using Boolean logic. Using gene expression data, we identified two markers, CD38 and ARG2, that group prostate cancer into three differentiation states. Cancers with CD38-, ARG2- expression patterns, corresponding to an undifferentiated state, had significantly lower 10-year recurrence-free survival compared to the most differentiated group (CD38+ARG2+). We carried out immunohistochemical (IHC) staining for these two markers in a single institution (Stanford; n = 234) and multi-institution (Canary; n = 1326) cohorts. IHC staining for CD38 and ARG2 in the Stanford cohort demonstrated that combined expression of CD38 and ARG2 was prognostic. In the Canary cohort, low CD38 protein expression by IHC was significantly associated with recurrence-free survival (RFS), seminal vesicle invasion (SVI), extra-capsular extension (ECE) in univariable analysis. In multivariable analysis, ARG2 and CD38 IHC staining results were not independently associated with RFS, overall survival, or disease-specific survival after adjusting for other factors including SVI, ECE, Gleason score, pre-operative PSA, and surgical margins.
Collapse
Affiliation(s)
- Debashis Sahoo
- Department of Pediatrics and Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
| | - Wei Wei
- The Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heidi Auman
- Canary Foundation, Canary Center at Stanford, Palo Alto, CA, USA
| | - Antonio Hurtado-Coll
- The Prostate Center at Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter R Carroll
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
| | - Ladan Fazli
- The Prostate Center at Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin E Gleave
- The Prostate Center at Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel W Lin
- Department of Urology, University of Washington Medical Center, Seattle, WA, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jeff Simko
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Ian M Thompson
- CHRISTUS Medical Center Hospital, San Antonio, Texas, USA
| | - Robin J Leach
- Department of Urology, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Dean A Troyer
- Eastern Virginia Medical School, Pathology, Microbiology and Molecular Biology, Norfolk, VA, USA
| | - Lawrence D True
- Department of Pathology, University of Washington Medical Center, Seattle, WA, USA
| | | | - Ziding Feng
- The Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James D Brooks
- Department of Urology, Stanford University, Stanford, CA, USA
| |
Collapse
|
26
|
Peng S, Wang K, Gu Y, Chen Y, Nan X, Xing J, Cui Q, Chen Y, Ge Q, Zhao H. TRAF3IP3, a novel autophagy up-regulated gene, is involved in marginal zone B lymphocyte development and survival. Clin Exp Immunol 2015; 182:57-68. [PMID: 26011558 DOI: 10.1111/cei.12658] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2015] [Indexed: 12/26/2022] Open
Abstract
Tumour necrosis factor receptor-associated factor 3 (TRAF3) interacting protein 3 (TRAF3IP3; also known as T3JAM) is expressed specifically in immune organs and tissues. To investigate the impact of TRAF3IP3 on immunity, we generated Traf3ip3 knock-out (KO) mice. Interestingly, these mice exhibited a significant reduction in the number of common lymphoid progenitors (CLPs) and inhibition of B cell development in the bone marrow. Furthermore, Traf3ip3 KO mice lacked marginal zone (MZ) B cells in the spleen. Traf3ip3 KO mice also exhibited a reduced amount of serum natural antibodies and impaired T cell-independent type II (TI-II) responses to trinitrophenol (TNP)-Ficoll antigen. Additionally, our results showed that Traf3ip3 promotes autophagy via an ATG16L1-binding motif, and MZ B cells isolated from mutant mice showed a diminished level of autophagy and a high rate of apoptosis. These results suggest that TRAF3IP3 contributes to MZ B cell survival by up-regulating autophagy, thereby promoting the TI-II immune response.
Collapse
Affiliation(s)
- S Peng
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China.,Human Disease Genomics Center, Peking University, Beijing, China
| | - K Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Y Gu
- Human Disease Genomics Center, Peking University, Beijing, China.,Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Y Chen
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China.,Human Disease Genomics Center, Peking University, Beijing, China
| | - X Nan
- Human Disease Genomics Center, Peking University, Beijing, China.,Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - J Xing
- Human Disease Genomics Center, Peking University, Beijing, China.,Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Q Cui
- Human Disease Genomics Center, Peking University, Beijing, China.,Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Y Chen
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Q Ge
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - H Zhao
- Human Disease Genomics Center, Peking University, Beijing, China.,Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
27
|
Guo NL, Wan YW. Network-based identification of biomarkers coexpressed with multiple pathways. Cancer Inform 2014; 13:37-47. [PMID: 25392692 PMCID: PMC4218687 DOI: 10.4137/cin.s14054] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/25/2014] [Accepted: 06/29/2014] [Indexed: 02/07/2023] Open
Abstract
Unraveling complex molecular interactions and networks and incorporating clinical information in modeling will present a paradigm shift in molecular medicine. Embedding biological relevance via modeling molecular networks and pathways has become increasingly important for biomarker identification in cancer susceptibility and metastasis studies. Here, we give a comprehensive overview of computational methods used for biomarker identification, and provide a performance comparison of several network models used in studies of cancer susceptibility, disease progression, and prognostication. Specifically, we evaluated implication networks, Boolean networks, Bayesian networks, and Pearson’s correlation networks in constructing gene coexpression networks for identifying lung cancer diagnostic and prognostic biomarkers. The results show that implication networks, implemented in Genet package, identified sets of biomarkers that generated an accurate prediction of lung cancer risk and metastases; meanwhile, implication networks revealed more biologically relevant molecular interactions than Boolean networks, Bayesian networks, and Pearson’s correlation networks when evaluated with MSigDB database.
Collapse
Affiliation(s)
- Nancy Lan Guo
- Mary Babb Randolph Cancer Center/School of Public Health, West Virginia University, Morgantown, WV, USA
| | - Ying-Wooi Wan
- Mary Babb Randolph Cancer Center/School of Public Health, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
28
|
Kong XD, Liu N, Xu XJ. Bioinformatics analysis of biomarkers and transcriptional factor motifs in Down syndrome. ACTA ACUST UNITED AC 2014; 47:834-41. [PMID: 25118625 PMCID: PMC4181218 DOI: 10.1590/1414-431x20143792] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/07/2014] [Indexed: 01/15/2023]
Abstract
In this study, biomarkers and transcriptional factor motifs were identified in order
to investigate the etiology and phenotypic severity of Down syndrome. GSE 1281, GSE
1611, and GSE 5390 were downloaded from the gene expression ominibus (GEO). A robust
multiarray analysis (RMA) algorithm was applied to detect differentially expressed
genes (DEGs). In order to screen for biological pathways and to interrogate the Kyoto
Encyclopedia of Genes and Genomes (KEGG) pathway database, the database for
annotation, visualization, and integrated discovery (DAVID) was used to carry out a
gene ontology (GO) function enrichment for DEGs. Finally, a transcriptional
regulatory network was constructed, and a hypergeometric distribution test was
applied to select for significantly enriched transcriptional factor motifs.
CBR1, DYRK1A, HMGN1,
ITSN1, RCAN1, SON,
TMEM50B, and TTC3 were each up-regulated
two-fold in Down syndrome samples compared to normal samples; of these,
SON and TTC3 were newly reported.
CBR1, DYRK1A, HMGN1,
ITSN1, RCAN1, SON,
TMEM50B, and TTC3 were located on human
chromosome 21 (mouse chromosome 16). The DEGs were significantly enriched in
macromolecular complex subunit organization and focal adhesion pathways. Eleven
significantly enriched transcription factor motifs (PAX5,
EGR1, XBP1, SREBP1,
OLF1, MZF1, NFY,
NFKAPPAB, MYCMAX, NFE2, and
RP58) were identified. The DEGs and transcription factor motifs
identified in our study provide biomarkers for the understanding of Down syndrome
pathogenesis and progression.
Collapse
Affiliation(s)
- X D Kong
- Prenatal Diagnosis Center, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - N Liu
- Prenatal Diagnosis Center, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - X J Xu
- Prenatal Diagnosis Center, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
29
|
Sinha S, Tsang EK, Zeng H, Meister M, Dill DL. Mining TCGA data using Boolean implications. PLoS One 2014; 9:e102119. [PMID: 25054200 PMCID: PMC4108374 DOI: 10.1371/journal.pone.0102119] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 06/16/2014] [Indexed: 01/08/2023] Open
Abstract
Boolean implications (if-then rules) provide a conceptually simple, uniform and highly scalable way to find associations between pairs of random variables. In this paper, we propose to use Boolean implications to find relationships between variables of different data types (mutation, copy number alteration, DNA methylation and gene expression) from the glioblastoma (GBM) and ovarian serous cystadenoma (OV) data sets from The Cancer Genome Atlas (TCGA). We find hundreds of thousands of Boolean implications from these data sets. A direct comparison of the relationships found by Boolean implications and those found by commonly used methods for mining associations show that existing methods would miss relationships found by Boolean implications. Furthermore, many relationships exposed by Boolean implications reflect important aspects of cancer biology. Examples of our findings include cis relationships between copy number alteration, DNA methylation and expression of genes, a new hierarchy of mutations and recurrent copy number alterations, loss-of-heterozygosity of well-known tumor suppressors, and the hypermethylation phenotype associated with IDH1 mutations in GBM. The Boolean implication results used in the paper can be accessed at http://crookneck.stanford.edu/microarray/TCGANetworks/.
Collapse
Affiliation(s)
- Subarna Sinha
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| | - Emily K. Tsang
- Biomedical Informatics Program, Stanford University, Stanford, California, United States of America
| | - Haoyang Zeng
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Michela Meister
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| | - David L. Dill
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| |
Collapse
|
30
|
Clonal precursor of bone, cartilage, and hematopoietic niche stromal cells. Proc Natl Acad Sci U S A 2013; 110:12643-8. [PMID: 23858471 DOI: 10.1073/pnas.1310212110] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Organs are composites of tissue types with diverse developmental origins, and they rely on distinct stem and progenitor cells to meet physiological demands for cellular production and homeostasis. How diverse stem cell activity is coordinated within organs is not well understood. Here we describe a lineage-restricted, self-renewing common skeletal progenitor (bone, cartilage, stromal progenitor; BCSP) isolated from limb bones and bone marrow tissue of fetal, neonatal, and adult mice. The BCSP clonally produces chondrocytes (cartilage-forming) and osteogenic (bone-forming) cells and at least three subsets of stromal cells that exhibit differential expression of cell surface markers, including CD105 (or endoglin), Thy1 [or CD90 (cluster of differentiation 90)], and 6C3 [ENPEP glutamyl aminopeptidase (aminopeptidase A)]. These three stromal subsets exhibit differential capacities to support hematopoietic (blood-forming) stem and progenitor cells. Although the 6C3-expressing subset demonstrates functional stem cell niche activity by maintaining primitive hematopoietic stem cell (HSC) renewal in vitro, the other stromal populations promote HSC differentiation to more committed lines of hematopoiesis, such as the B-cell lineage. Gene expression analysis and microscopic studies further reveal a microenvironment in which CD105-, Thy1-, and 6C3-expressing marrow stroma collaborate to provide cytokine signaling to HSCs and more committed hematopoietic progenitors. As a result, within the context of bone as a blood-forming organ, the BCSP plays a critical role in supporting hematopoiesis through its generation of diverse osteogenic and hematopoietic-promoting stroma, including HSC supportive 6C3(+) niche cells.
Collapse
|
31
|
Rosin DP, Rontani D, Gauthier DJ, Schöll E. Experiments on autonomous Boolean networks. CHAOS (WOODBURY, N.Y.) 2013; 23:025102. [PMID: 23822500 DOI: 10.1063/1.4807481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
We realize autonomous Boolean networks by using logic gates in their autonomous mode of operation on a field-programmable gate array. This allows us to implement time-continuous systems with complex dynamical behaviors that can be conveniently interconnected into large-scale networks with flexible topologies that consist of time-delay links and a large number of nodes. We demonstrate how we realize networks with periodic, chaotic, and excitable dynamics and study their properties. Field-programmable gate arrays define a new experimental paradigm that holds great potential to test a large body of theoretical results on the dynamics of complex networks, which has been beyond reach of traditional experimental approaches.
Collapse
Affiliation(s)
- David P Rosin
- Duke University, Department of Physics, Science Drive, Durham, North Carolina 27708, USA
| | | | | | | |
Collapse
|
32
|
Martinez E, Trevino V. Modelling gene expression profiles related to prostate tumor progression using binary states. Theor Biol Med Model 2013; 10:37. [PMID: 23721350 PMCID: PMC3691825 DOI: 10.1186/1742-4682-10-37] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 05/21/2013] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Cancer is a complex disease commonly characterized by the disrupted activity of several cancer-related genes such as oncogenes and tumor-suppressor genes. Previous studies suggest that the process of tumor progression to malignancy is dynamic and can be traced by changes in gene expression. Despite the enormous efforts made for differential expression detection and biomarker discovery, few methods have been designed to model the gene expression level to tumor stage during malignancy progression. Such models could help us understand the dynamics and simplify or reveal the complexity of tumor progression. METHODS We have modeled an on-off state of gene activation per sample then per stage to select gene expression profiles associated to tumor progression. The selection is guided by statistical significance of profiles based on random permutated datasets. RESULTS We show that our method identifies expected profiles corresponding to oncogenes and tumor suppressor genes in a prostate tumor progression dataset. Comparisons with other methods support our findings and indicate that a considerable proportion of significant profiles is not found by other statistical tests commonly used to detect differential expression between tumor stages nor found by other tailored methods. Ontology and pathway analysis concurred with these findings. CONCLUSIONS Results suggest that our methodology may be a valuable tool to study tumor malignancy progression, which might reveal novel cancer therapies.
Collapse
Affiliation(s)
- Emmanuel Martinez
- Tecnológico de Monterrey, Campus Monterrey, Cátedra de Bioinformática, Monterrey, Nuevo León 64849, México
| | - Victor Trevino
- Tecnológico de Monterrey, Campus Monterrey, Cátedra de Bioinformática, Monterrey, Nuevo León 64849, México
| |
Collapse
|
33
|
Rinkevich Y, Mori T, Sahoo D, Xu PX, Bermingham JR, Weissman IL. Identification and prospective isolation of a mesothelial precursor lineage giving rise to smooth muscle cells and fibroblasts for mammalian internal organs, and their vasculature. Nat Cell Biol 2012; 14:1251-60. [PMID: 23143399 DOI: 10.1038/ncb2610] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 10/02/2012] [Indexed: 01/14/2023]
Abstract
Fibroblasts and smooth muscle cells (FSMCs) are principal cell types of connective and adventitial tissues that participate in the development, physiology and pathology of internal organs, with incompletely defined cellular origins. Here, we identify and prospectively isolate from the mesothelium a mouse cell lineage that is committed to FSMCs. The mesothelium is an epithelial monolayer covering the vertebrate thoracic and abdominal cavities and internal organs. Time-lapse imaging and transplantation experiments reveal robust generation of FSMCs from the mesothelium. By targeting mesothelin (MSLN), a surface marker expressed on mesothelial cells, we identify and isolate precursors capable of clonally generating FSMCs. Using a genetic lineage tracing approach, we show that embryonic and adult mesothelium represents a common lineage to trunk FSMCs, and trunk vasculature, with minimal contributions from neural crest, or circulating cells. The isolation of FSMC precursors enables the examination of multiple aspects of smooth muscle and fibroblast biology as well as the prospective isolation of these precursors for potential regenerative medicine purposes.
Collapse
Affiliation(s)
- Yuval Rinkevich
- Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Human diseases have been investigated in the context of single genes as well as complex networks of genes. Though single gene approaches have been extremely successful in the past, most human diseases are complex and better characterized by multiple interacting genes commonly known as networks or pathways. With the advent of high-throughput technologies, a recent trend has been to apply network-based analysis to the huge amount of biological data. Analysis on Boolean implication network is one such technique that distinguishes itself based on its simplicity and robustness. Unlike traditional analyses, Boolean implication networks have the power to break into the mechanistic insights of human diseases. A Boolean implication network is a collection of simple Boolean relationships such as “if A is high then B is low.” So far, Boolean implication networks have been employed not only to discover novel markers of differentiation in both normal and cancer tissues, but also to develop robust treatment decisions for cancer patients. Therefore, analyses based on Boolean implication networks have potential to accelerate discoveries in human diseases, suggest therapeutics, and provide robust risk-adapted clinical strategies.
Collapse
Affiliation(s)
- Debashis Sahoo
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University Stanford, CA, USA
| |
Collapse
|
35
|
Guo NL, Wan YW. Pathway-based identification of a smoking associated 6-gene signature predictive of lung cancer risk and survival. Artif Intell Med 2012; 55:97-105. [PMID: 22326768 DOI: 10.1016/j.artmed.2012.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 11/07/2011] [Accepted: 01/17/2012] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Smoking is a prominent risk factor for lung cancer. However, it is not an established prognostic factor for lung cancer in clinics. To date, no gene test is available for diagnostic screening of lung cancer risk or prognostication of clinical outcome in smokers. This study sought to identify a smoking associated gene signature in order to provide a more precise diagnosis and prognosis of lung cancer in smokers. METHODS AND MATERIALS An implication network based methodology was used to identify biomarkers by modeling crosstalk with major lung cancer signaling pathways. Specifically, the methodology contains the following steps: (1) identifying genes significantly associated with lung cancer survival; (2) selecting candidate genes which are differentially expressed in smokers versus non-smokers from the survival genes identified in Step 1; (3) from these candidate genes, constructing gene coexpression networks based on prediction logic for the smoker group and the non-smoker group, respectively; (4) identifying smoking-mediated differential components, i.e., the unique gene coexpression patterns specific to each group; and (5) from the differential components, identifying genes directly co-expressed with major lung cancer signaling hallmarks. RESULTS A smoking-associated 6-gene signature was identified for prognosis of lung cancer from a training cohort (n=256). The 6-gene signature could separate lung cancer patients into two risk groups with distinct post-operative survival (log-rank P<0.04, Kaplan-Meier analyses) in three independent cohorts (n=427). The expression-defined prognostic prediction is strongly related to smoking association and smoking cessation (P<0.02; Pearson's Chi-squared tests). The 6-gene signature is an accurate prognostic factor (hazard ratio=1.89, 95% CI: [1.04, 3.43]) compared to common clinical covariates in multivariate Cox analysis. The 6-gene signature also provides an accurate diagnosis of lung cancer with an overall accuracy of 73% in a cohort of smokers (n=164). The coexpression patterns derived from the implication networks were validated with interactions reported in the literature retrieved with STRING8, Ingenuity Pathway Analysis, and Pathway Studio. CONCLUSIONS The pathway-based approach identified a smoking-associated 6-gene signature that predicts lung cancer risk and survival. This gene signature has potential clinical implications in the diagnosis and prognosis of lung cancer in smokers.
Collapse
Affiliation(s)
- Nancy Lan Guo
- Department of Community Medicine, West Virginia University, Morgantown, WV 26506, USA.
| | | |
Collapse
|
36
|
Three differentiation states risk-stratify bladder cancer into distinct subtypes. Proc Natl Acad Sci U S A 2012; 109:2078-83. [PMID: 22308455 DOI: 10.1073/pnas.1120605109] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Current clinical judgment in bladder cancer (BC) relies primarily on pathological stage and grade. We investigated whether a molecular classification of tumor cell differentiation, based on a developmental biology approach, can provide additional prognostic information. Exploiting large preexisting gene-expression databases, we developed a biologically supervised computational model to predict markers that correspond with BC differentiation. To provide mechanistic insight, we assessed relative tumorigenicity and differentiation potential via xenotransplantation. We then correlated the prognostic utility of the identified markers to outcomes within gene expression and formalin-fixed paraffin-embedded (FFPE) tissue datasets. Our data indicate that BC can be subclassified into three subtypes, on the basis of their differentiation states: basal, intermediate, and differentiated, where only the most primitive tumor cell subpopulation within each subtype is capable of generating xenograft tumors and recapitulating downstream populations. We found that keratin 14 (KRT14) marks the most primitive differentiation state that precedes KRT5 and KRT20 expression. Furthermore, KRT14 expression is consistently associated with worse prognosis in both univariate and multivariate analyses. We identify here three distinct BC subtypes on the basis of their differentiation states, each harboring a unique tumor-initiating population.
Collapse
|
37
|
Affiliation(s)
- Nancy Lan Guo
- Mary Babb Randolph Cancer Center/Department of Community Medicine, School of Medicine, West Virginia University, Morgantown, WV 26506-9300
| |
Collapse
|
38
|
Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age. Proc Natl Acad Sci U S A 2011; 108:20012-7. [PMID: 22123971 DOI: 10.1073/pnas.1116110108] [Citation(s) in RCA: 638] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In the human hematopoietic system, aging is associated with decreased bone marrow cellularity, decreased adaptive immune system function, and increased incidence of anemia and other hematological disorders and malignancies. Recent studies in mice suggest that changes within the hematopoietic stem cell (HSC) population during aging contribute significantly to the manifestation of these age-associated hematopoietic pathologies. Though the mouse HSC population has been shown to change both quantitatively and functionally with age, changes in the human HSC and progenitor cell populations during aging have been incompletely characterized. To elucidate the properties of an aged human hematopoietic system that may predispose to age-associated hematopoietic dysfunction, we evaluated immunophenotypic HSC and other hematopoietic progenitor populations from healthy, hematologically normal young and elderly human bone marrow samples. We found that aged immunophenotypic human HSC increase in frequency, are less quiescent, and exhibit myeloid-biased differentiation potential compared with young HSC. Gene expression profiling revealed that aged immunophenotypic human HSC transcriptionally up-regulate genes associated with cell cycle, myeloid lineage specification, and myeloid malignancies. These age-associated alterations in the frequency, developmental potential, and gene expression profile of human HSC are similar to those changes observed in mouse HSC, suggesting that hematopoietic aging is an evolutionarily conserved process.
Collapse
|
39
|
Ghanbarnejad F, Klemm K. Stability of Boolean and continuous dynamics. PHYSICAL REVIEW LETTERS 2011; 107:188701. [PMID: 22107682 DOI: 10.1103/physrevlett.107.188701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Indexed: 05/31/2023]
Abstract
Regulatory dynamics in biology is often described by continuous rate equations for continuously varying chemical concentrations. Binary discretization of state space and time leads to Boolean dynamics. In the latter, the dynamics has been called unstable if flip perturbations lead to damage spreading. Here, we find that this stability classification strongly differs from the stability properties of the original continuous dynamics under small perturbations of the state vector. In particular, random networks of nodes with large sensitivity yield stable dynamics under small perturbations.
Collapse
Affiliation(s)
- Fakhteh Ghanbarnejad
- Bioinformatics Group, Institute for Computer Science, University of Leipzig, Härtelstraße 16-18, D-04107 Leipzig, Germany.
| | | |
Collapse
|
40
|
An antibody against SSEA-5 glycan on human pluripotent stem cells enables removal of teratoma-forming cells. Nat Biotechnol 2011; 29:829-34. [PMID: 21841799 DOI: 10.1038/nbt.1947] [Citation(s) in RCA: 287] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 07/18/2011] [Indexed: 12/18/2022]
Abstract
An important risk in the clinical application of human pluripotent stem cells (hPSCs), including human embryonic and induced pluripotent stem cells (hESCs and hiPSCs), is teratoma formation by residual undifferentiated cells. We raised a monoclonal antibody against hESCs, designated anti-stage-specific embryonic antigen (SSEA)-5, which binds a previously unidentified antigen highly and specifically expressed on hPSCs--the H type-1 glycan. Separation based on SSEA-5 expression through fluorescence-activated cell sorting (FACS) greatly reduced teratoma-formation potential of heterogeneously differentiated cultures. To ensure complete removal of teratoma-forming cells, we identified additional pluripotency surface markers (PSMs) exhibiting a large dynamic expression range during differentiation: CD9, CD30, CD50, CD90 and CD200. Immunohistochemistry studies of human fetal tissues and bioinformatics analysis of a microarray database revealed that concurrent expression of these markers is both common and specific to hPSCs. Immunodepletion with antibodies against SSEA-5 and two additional PSMs completely removed teratoma-formation potential from incompletely differentiated hESC cultures.
Collapse
|
41
|
Reijo Pera RA. Non-invasive imaging of human embryos to predict developmental competence. Placenta 2011; 32 Suppl 3:S264-7. [PMID: 21802136 DOI: 10.1016/j.placenta.2011.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 07/05/2011] [Accepted: 07/06/2011] [Indexed: 10/17/2022]
Abstract
Although some aspects of human embryo development are conserved with those of other species, including the mouse, many aspects such as the timing of reprogramming and occurrence in the absence of transcription, duration of transcriptional silence and identity of genes with modulated expression in the oocyte to embryo transition, appear to be unique. Yet, frequently, the only data available for understanding the programs of early embryo development is that derived from model or agricultural species. We suggest that a specific understanding of basic aspects of human embryo development can affect a two-fold positive impact: 1) We can improve the health of a substantial subset of patients who seek assisted reproduction by improving diagnostics of viable embryo development in the clinic and, 2) we can use the information we gather to improve derivation and diagnosis of pluripotent stem cell lines (including reference or gold-standard human embryonic stem cell (hESC) lines and closely-related induced pluripotent stem cell (iPSC) lines) and their fates in novel basic and clinical applications.
Collapse
Affiliation(s)
- R A Reijo Pera
- Institute for Stem Cell Biology and Regenerative Medicine, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305-5463, USA.
| |
Collapse
|