1
|
Jin J, Yang YR, Gong Q, Wang JN, Ni WJ, Wen JG, Meng XM. Role of epigenetically regulated inflammation in renal diseases. Semin Cell Dev Biol 2024; 154:295-304. [PMID: 36328897 DOI: 10.1016/j.semcdb.2022.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/01/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
Abstract
In recent decades, renal disease research has witnessed remarkable advances. Experimental evidence in this field has highlighted the role of inflammation in kidney disease. Epigenetic dynamics and immunometabolic reprogramming underlie the alterations in cellular responses to intrinsic and extrinsic stimuli; these factors determine cell identity and cell fate decisions and represent current research hotspots. This review focuses on recent findings and emerging concepts in epigenetics and inflammatory regulation and their effect on renal diseases. This review aims to summarize the role and mechanisms of different epigenetic modifications in renal inflammation and injury and provide new avenues for future research on inflammation-related renal disease and drug development.
Collapse
Affiliation(s)
- Juan Jin
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China; School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Ya-Ru Yang
- Department of Clinical Pharmacology, Second Hospital of Anhui Medical University, Hefei, China
| | - Qian Gong
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Jia-Nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Wei-Jian Ni
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China.
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
2
|
Palshikar MG, Palli R, Tyrell A, Maggirwar S, Schifitto G, Singh MV, Thakar J. Executable models of immune signaling pathways in HIV-associated atherosclerosis. NPJ Syst Biol Appl 2022; 8:35. [PMID: 36131068 PMCID: PMC9492768 DOI: 10.1038/s41540-022-00246-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022] Open
Abstract
Atherosclerosis (AS)-associated cardiovascular disease is an important cause of mortality in an aging population of people living with HIV (PLWH). This elevated risk has been attributed to viral infection, anti-retroviral therapy, chronic inflammation, and lifestyle factors. However, the rates at which PLWH develop AS vary even after controlling for length of infection, treatment duration, and for lifestyle factors. To investigate the molecular signaling underlying this variation, we sequenced 9368 peripheral blood mononuclear cells (PBMCs) from eight PLWH, four of whom have atherosclerosis (AS+). Additionally, a publicly available dataset of PBMCs from persons before and after HIV infection was used to investigate the effect of acute HIV infection. To characterize dysregulation of pathways rather than just measuring enrichment, we developed the single-cell Boolean Omics Network Invariant Time Analysis (scBONITA) algorithm. scBONITA infers executable dynamic pathway models and performs a perturbation analysis to identify high impact genes. These dynamic models are used for pathway analysis and to map sequenced cells to characteristic signaling states (attractor analysis). scBONITA revealed that lipid signaling regulates cell migration into the vascular endothelium in AS+ PLWH. Pathways implicated included AGE-RAGE and PI3K-AKT signaling in CD8+ T cells, and glucagon and cAMP signaling pathways in monocytes. Attractor analysis with scBONITA facilitated the pathway-based characterization of cellular states in CD8+ T cells and monocytes. In this manner, we identify critical cell-type specific molecular mechanisms underlying HIV-associated atherosclerosis using a novel computational method.
Collapse
Affiliation(s)
- Mukta G Palshikar
- Biophysics, Structural, and Computational Biology Program, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Rohith Palli
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Alicia Tyrell
- University of Rochester Clinical & Translational Science Institute, Rochester, USA
| | - Sanjay Maggirwar
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Giovanni Schifitto
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, USA
- Department of Imaging Sciences, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Meera V Singh
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Juilee Thakar
- Biophysics, Structural, and Computational Biology Program, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, USA.
| |
Collapse
|
3
|
Yang L, Semmes EC, Ovies C, Megli C, Permar S, Gilner JB, Coyne CB. Innate immune signaling in trophoblast and decidua organoids defines differential antiviral defenses at the maternal-fetal interface. eLife 2022; 11:e79794. [PMID: 35975985 PMCID: PMC9470165 DOI: 10.7554/elife.79794] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Infections at the maternal-fetal interface can directly harm the fetus and induce complications that adversely impact pregnancy outcomes. Innate immune signaling by both fetal-derived placental trophoblasts and the maternal decidua must provide antimicrobial defenses at this critical interface without compromising its integrity. Here, we developed matched trophoblast (TO) and decidua organoids (DO) from human placentas to define the relative contributions of these cells to antiviral defenses at the maternal-fetal interface. We demonstrate that TO and DO basally secrete distinct immunomodulatory factors, including the constitutive release of the antiviral type III interferon IFN-λ2 from TOs, and differentially respond to viral infections through the induction of organoid-specific factors. Finally, we define the differential susceptibility and innate immune signaling of TO and DO to human cytomegalovirus (HCMV) and develop a co-culture model of TO and DO which showed that trophoblast-derived factors protect decidual cells from HCMV infection. Our findings establish matched TO and DO as ex vivo models to study vertically transmitted infections and highlight differences in innate immune signaling by fetal-derived trophoblasts and the maternal decidua.
Collapse
Affiliation(s)
- Liheng Yang
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
| | - Eleanor C Semmes
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
- Duke Human Vaccine Institute, Duke UniversityDurhamUnited States
| | - Cristian Ovies
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
| | - Christina Megli
- Division of Maternal-Fetal Medicine, Division of Reproductive Infectious Disease, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh Medical Center (UPMC)PittsburghUnited States
- Magee Womens Research InstitutePittsburghUnited States
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medical Center, Duke University Medical CenterDurhamUnited States
| | - Jennifer B Gilner
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Duke University Medical CenterDurhamUnited States
| | - Carolyn B Coyne
- Department of Molecular Genetics and Microbiology, Duke University School of MedicineDurhamUnited States
- Duke Human Vaccine Institute, Duke UniversityDurhamUnited States
| |
Collapse
|
4
|
McCall MN, Chu CY, Wang L, Benoodt L, Thakar J, Corbett A, Holden-Wiltse J, Slaunwhite C, Grier A, Gill SR, Falsey AR, Topham DJ, Caserta MT, Walsh EE, Qiu X, Mariani TJ. A systems genomics approach uncovers molecular associates of RSV severity. PLoS Comput Biol 2021; 17:e1009617. [PMID: 34962914 PMCID: PMC8746750 DOI: 10.1371/journal.pcbi.1009617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 01/10/2022] [Accepted: 11/05/2021] [Indexed: 01/06/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection results in millions of hospitalizations and thousands of deaths each year. Variations in the adaptive and innate immune response appear to be associated with RSV severity. To investigate the host response to RSV infection in infants, we performed a systems-level study of RSV pathophysiology, incorporating high-throughput measurements of the peripheral innate and adaptive immune systems and the airway epithelium and microbiota. We implemented a novel multi-omic data integration method based on multilayered principal component analysis, penalized regression, and feature weight back-propagation, which enabled us to identify cellular pathways associated with RSV severity. In both airway and immune cells, we found an association between RSV severity and activation of pathways controlling Th17 and acute phase response signaling, as well as inhibition of B cell receptor signaling. Dysregulation of both the humoral and mucosal response to RSV may play a critical role in determining illness severity.
Collapse
Affiliation(s)
- Matthew N. McCall
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester New York, United States of America
| | - Chin-Yi Chu
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Pediatrics, University of Rochester Medical Center, Rochester New York, United States of America
| | - Lu Wang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Lauren Benoodt
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester New York, United States of America
| | - Juilee Thakar
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Anthony Corbett
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
- Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester New York, United States of America
| | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
- Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester New York, United States of America
| | - Christopher Slaunwhite
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Pediatrics, University of Rochester Medical Center, Rochester New York, United States of America
| | - Alex Grier
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Steven R. Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Ann R. Falsey
- Department of Medicine, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Medicine, Rochester General Hospital, Rochester New York, United States of America
| | - David J. Topham
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Mary T. Caserta
- Department of Pediatrics, University of Rochester Medical Center, Rochester New York, United States of America
| | - Edward E. Walsh
- Department of Medicine, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Medicine, Rochester General Hospital, Rochester New York, United States of America
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Thomas J. Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Pediatrics, University of Rochester Medical Center, Rochester New York, United States of America
| |
Collapse
|
5
|
Cornwell A, Palli R, Singh MV, Benoodt L, Tyrell A, Abe JI, Schifitto G, Maggirwar SB, Thakar J. Molecular characterization of atherosclerosis in HIV positive persons. Sci Rep 2021; 11:3232. [PMID: 33547350 PMCID: PMC7865026 DOI: 10.1038/s41598-021-82429-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 12/30/2020] [Indexed: 01/30/2023] Open
Abstract
People living with HIV are at higher risk of atherosclerosis (AS). The pathogenesis of this risk is not fully understood. To assess the regulatory networks involved in AS we sequenced mRNA of the peripheral blood mononuclear cells (PBMCs) and measured cytokine and chemokine levels in the plasma of 13 persons living with HIV and 12 matched HIV-negative persons with and without AS. microRNAs (miRNAs) are known to play a role in HIV infection and may modulate gene regulation to drive AS. Hence, we further assessed miRNA expression in PBMCs of a subset of 12 HIV+ people with and without atherosclerosis. We identified 12 miRNAs differentially expressed between HIV+ AS+ and HIV+ , and validated 5 of those by RT-qPCR. While a few of these miRNAs have been implicated in HIV and atherosclerosis, others are novel. Integrating miRNA measurements with mRNA, we identified 27 target genes including SLC4A7, a critical sodium and bicarbonate transporter, that are potentially dysregulated during atherosclerosis. Additionally, we uncovered that levels of plasma cytokines were associated with transcription factor activity and miRNA expression in PBMCs. For example, BACH2 activity was associated with IL-1β, IL-15, and MIP-1α. IP10 and TNFα levels were associated with miR-124-3p. Finally, integration of all data types into a single network revealed increased importance of miRNAs in network regulation of the HIV+ group in contrast with increased importance of cytokines in the HIV+ AS+ group.
Collapse
Affiliation(s)
- Adam Cornwell
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
| | - Rohith Palli
- Medical Scientist Training Program, University of Rochester, Rochester, NY, USA
- Biophysics, Structural, and Computational Biology PhD Program, University of Rochester, Rochester, NY, USA
| | - Meera V Singh
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Lauren Benoodt
- Biophysics, Structural, and Computational Biology PhD Program, University of Rochester, Rochester, NY, USA
| | - Alicia Tyrell
- Department of Neurology, General Neurology, University of Rochester, Rochester, NY, USA
- Department of Imaging Sciences, University of Rochester, Rochester, NY, USA
| | - Jun-Ichi Abe
- Department of Cardiology-Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Texas A&M Health Science Center Institute of Biosciences and Technology, Houston, TX, USA
| | - Giovanni Schifitto
- Department of Neurology, General Neurology, University of Rochester, Rochester, NY, USA
- Department of Imaging Sciences, University of Rochester, Rochester, NY, USA
| | - Sanjay B Maggirwar
- Department of Microbiology, Immunology, and Tropical Medicine, George Washing University, Washington, DC, USA
| | - Juilee Thakar
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA.
- Department of Biostatistics and Computational Biology, University of Rochester, 601 Elmwood Avenue, , Box 672, Rochester, NY, 14642, USA.
| |
Collapse
|
6
|
Thakar J, Qian Y, Benoodt L, Roumanes D, Qiu X, Laniewski N, Chu C, Slaunwhite C, Wang L, Mandava A, Chang I, Falsey AR, Caserta MT, Mariani TJ, Scheuermann RH, Walsh EE, Topham DJ. Unbiased analysis of peripheral blood mononuclear cells reveals CD4 T cell response to RSV matrix protein. Vaccine X 2020; 5:100065. [PMID: 32529184 PMCID: PMC7280769 DOI: 10.1016/j.jvacx.2020.100065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/13/2020] [Accepted: 04/20/2020] [Indexed: 12/02/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most important cause of respiratory tract illness especially in young infants that develop severe disease requiring hospitalization, and accounting for 74,000-126,000 admissions in the United States (Rezaee et al., 2017; Resch, 2017). Observations of neonatal and infant T cells suggest that they may express different immune markers compared to T-cells from older children. Flow cytometry analysis of cellular responses using "conventional" anti-viral markers (IL2, IFN-γ, TNF, IL10 and IL4) upon RSV-peptide stimulation detected an overall low RSV response in peripheral blood. Therefore we sought an unbiased approach to identify RSV-specific immune markers using RNA-sequencing upon stimulation of infant PBMCs with overlapping peptides representing RSV antigens. To understand the cellular response using transcriptional signatures, transcription factors and cell-type specific signatures were used to investigate breadth of response across peptides. Unexpected from the ICS data, M peptide induced a response equivalent to the F-peptide and was characterized by activation of GATA2, 3, STAT3 and IRF1. This along with upregulation of several unconventional T cell signatures was only observed upon M-peptide stimulation. Moreover, signatures of natural RSV infections were identified from the data available in the public domain to investigate similarities between transcriptional signatures from PBMCs and upon peptide stimulation. This analysis also suggested activation of T cell response upon M-peptide stimulation. Hence, based on transcriptional response, markers were chosen to validate the role of M-peptide in activation of T cells. Indeed, CD4+CXCL9+ cells were identified upon M-peptide stimulation by flow cytometry. Future work using additional markers identified in this study could reveal additional unconventional T cells responding to RSV infections in infants. In conclusion, T cell responses to RSV in infants may not follow the canonical Th1/Th2 patterns of effector responses but include additional functions that may be unique to the neonatal period and correlate with clinical outcomes.
Collapse
Affiliation(s)
- Juilee Thakar
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | - Yu Qian
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Lauren Benoodt
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States
- Biophysics and Computational Biology Graduate Program, University of Rochester, Rochester, NY, United States
| | - David Roumanes
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | - Nathan Laniewski
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - ChinYi Chu
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, United States
| | - Christopher Slaunwhite
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, United States
| | - Lu Wang
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | | | - Ivan Chang
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Ann R Falsey
- Department of Medicine, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, NY, United States
| | - Mary T Caserta
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, United States
| | - Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, United States
| | - Richard H Scheuermann
- J. Craig Venter Institute, La Jolla, CA, United States
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| | - Edward E Walsh
- Department of Medicine, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, NY, United States
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, United States
| | - David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
7
|
Executable pathway analysis using ensemble discrete-state modeling for large-scale data. PLoS Comput Biol 2019; 15:e1007317. [PMID: 31479446 PMCID: PMC6743792 DOI: 10.1371/journal.pcbi.1007317] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 09/13/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022] Open
Abstract
Pathway analysis is widely used to gain mechanistic insights from high-throughput omics data. However, most existing methods do not consider signal integration represented by pathway topology, resulting in enrichment of convergent pathways when downstream genes are modulated. Incorporation of signal flow and integration in pathway analysis could rank the pathways based on modulation in key regulatory genes. This implementation can be facilitated for large-scale data by discrete state network modeling due to simplicity in parameterization. Here, we model cellular heterogeneity using discrete state dynamics and measure pathway activities in cross-sectional data. We introduce a new algorithm, Boolean Omics Network Invariant-Time Analysis (BONITA), for signal propagation, signal integration, and pathway analysis. Our signal propagation approach models heterogeneity in transcriptomic data as arising from intercellular heterogeneity rather than intracellular stochasticity, and propagates binary signals repeatedly across networks. Logic rules defining signal integration are inferred by genetic algorithm and are refined by local search. The rules determine the impact of each node in a pathway, which is used to score the probability of the pathway's modulation by chance. We have comprehensively tested BONITA for application to transcriptomics data from translational studies. Comparison with state-of-the-art pathway analysis methods shows that BONITA has higher sensitivity at lower levels of source node modulation and similar sensitivity at higher levels of source node modulation. Application of BONITA pathway analysis to previously validated RNA-sequencing studies identifies additional relevant pathways in in-vitro human cell line experiments and in-vivo infant studies. Additionally, BONITA successfully detected modulation of disease specific pathways when comparing relevant RNA-sequencing data with healthy controls. Most interestingly, the two highest impact score nodes identified by BONITA included known drug targets. Thus, BONITA is a powerful approach to prioritize not only pathways but also specific mechanistic role of genes compared to existing methods. BONITA is available at: https://github.com/thakar-lab/BONITA.
Collapse
|
8
|
Mariani TJ, Qiu X, Chu C, Wang L, Thakar J, Holden-Wiltse J, Corbett A, Topham DJ, Falsey AR, Caserta MT, Walsh EE. Association of Dynamic Changes in the CD4 T-Cell Transcriptome With Disease Severity During Primary Respiratory Syncytial Virus Infection in Young Infants. J Infect Dis 2017; 216:1027-1037. [PMID: 28962005 PMCID: PMC5853440 DOI: 10.1093/infdis/jix400] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/08/2017] [Indexed: 01/10/2023] Open
Abstract
Background Nearly all children are infected with respiratory syncytial virus (RSV) within the first 2 years of life, with a minority developing severe disease (1%-3% hospitalized). We hypothesized that an assessment of the adaptive immune system, using CD4+ T-lymphocyte transcriptomics, would identify gene expression correlates of disease severity. Methods Infants infected with RSV representing extremes of clinical severity were studied. Mild illness (n = 23) was defined as a respiratory rate (RR) < 55 and room air oxygen saturation (SaO2) ≥ 97%, and severe illness (n = 23) was defined as RR ≥ 65 and SaO2 ≤ 92%. RNA from fresh, sort-purified CD4+ T cells was assessed by RNA sequencing. Results Gestational age, age at illness onset, exposure to environmental tobacco smoke, bacterial colonization, and breastfeeding were associated (adjusted P < .05) with disease severity. RNA sequencing analysis reliably measured approximately 60% of the genome. Severity of RSV illness had the greatest effect size upon CD4 T-cell gene expression. Pathway analysis identified correlates of severity, including JAK/STAT, prolactin, and interleukin 9 signaling. We also identified genes and pathways associated with timing of symptoms and RSV group (A/B). Conclusions These data suggest fundamental changes in adaptive immune cell phenotypes may be associated with RSV clinical severity.
Collapse
Affiliation(s)
- Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program.,Department of Medicine, University of Rochester Medical Center
| | - Xing Qiu
- Department of Biostatistics and Computational Biology
| | - ChinYi Chu
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program.,Department of Medicine, University of Rochester Medical Center
| | - Lu Wang
- Department of Biostatistics and Computational Biology
| | | | | | | | | | - Ann R Falsey
- Department of Medicine, University of Rochester Medical Center.,Department of Medicine, Rochester General Hospital, Rochester, New York
| | | | - Edward E Walsh
- Department of Medicine, University of Rochester Medical Center.,Department of Medicine, Rochester General Hospital, Rochester, New York
| |
Collapse
|