1
|
Elsayed AA, Barghash SM, El-Kattan AM, Wassif IM, Osman WA, Ateya AI. Analysis of potential genes, immunological and antioxidant profiles associated with trypanosomiasis susceptibility in dromedary camels. Vet Parasitol 2024; 331:110264. [PMID: 39059159 DOI: 10.1016/j.vetpar.2024.110264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Trypanosomiasis is associated with tissue damage and may trigger an immunological response. These tissue lesions are linked to metabolic issues and oxidative stress. The current study aimed to investigate the immunological, antioxidant, and metabolic changes that may be connected to camel trypanosomiasis. Blood samples were collected from 54 camels and allocated into two groups: The control group (35 camels) and the infected group (19 camels). The genes TLR2, TLR5, IL-17, MARCHF3, RASGRP1, EPS15L1, PPIE, ASB16, CMPK2, LPCAT1, FPGT, GPHN, TNNI3K, DIO3, keap1, and OXSR1 were significantly up-regulated in trypanosomiasis camels. However, down-regulation was observed for the genes Nrf2, PRDX6, and NDUFS5. PCR-DNA sequencing was used to identify nucleotide sequence polymorphisms in the immune (TLR2, TLR5, IL-17, MARCHF3, RASGRP1, and EPS15L1), metabolic (PPIE, ASB16, CMPK2, LPCAT1, FPGT, GPHN, TNNI3K, and DIO3), and antioxidant (Nrf2, Keap1, PRDX6, NDUFS5, and OXSR1) genes between healthy and trypanosomiasis-affected camels. Exploring the serum profile also showed a significant (P ˂ 0.05) increase in Hp, SAA, Cp, IL-1β, IL-6, IL 10, TNF-α, and MDA, with significant (P ˂ 0.05) reduction in the serum levels of CAT, SOD, GSH, T3, and T4 in diseased camels compared with healthy ones. Our findings confirm the significance of nucleotide variations, gene expression patterns, and the biochemical profile of the investigated markers as indicators for the susceptibility of trypanosomiasis in dromedary camels and may be utilized to create management strategies.
Collapse
Affiliation(s)
- Ahmed A Elsayed
- Department of Animal Health and Poultry, Animal and Poultry Production Division, Desert Research Center (DRC), Cairo, Egypt
| | - Safaa M Barghash
- Department of Animal Health and Poultry, Animal and Poultry Production Division, Desert Research Center (DRC), Cairo, Egypt
| | - Adel M El-Kattan
- Department of Animal Health and Poultry, Animal and Poultry Production Division, Desert Research Center (DRC), Cairo, Egypt
| | - Islam M Wassif
- Department of Animal Health and Poultry, Animal and Poultry Production Division, Desert Research Center (DRC), Cairo, Egypt
| | - Wafaa A Osman
- Department of Animal Health and Poultry, Animal and Poultry Production Division, Desert Research Center (DRC), Cairo, Egypt
| | - Ahmed I Ateya
- Department of Animal Wealth Development, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
2
|
Wu X, Xiao C, Rasooly D, Zhao X, Morton CC, Jiang X, Gallagher CS. A comprehensive genome-wide cross-trait analysis of sexual factors and uterine leiomyoma. PLoS Genet 2024; 20:e1011268. [PMID: 38701081 PMCID: PMC11095738 DOI: 10.1371/journal.pgen.1011268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 05/15/2024] [Accepted: 04/22/2024] [Indexed: 05/05/2024] Open
Abstract
Age at first sexual intercourse (AFS) and lifetime number of sexual partners (NSP) may influence the pathogenesis of uterine leiomyoma (UL) through their associations with hormonal concentrations and uterine infections. Leveraging summary statistics from large-scale genome-wide association studies conducted in European ancestry for each trait (NAFS = 214,547; NNSP = 370,711; NUL = 302,979), we observed a significant negative genomic correlation for UL with AFS (rg = -0.11, P = 7.83×10-4), but not with NSP (rg = 0.01, P = 0.62). Four specific genomic regions were identified as contributing significant local genetic correlations to AFS and UL, including one genomic region further identified for NSP and UL. Partitioning SNP-heritability with cell-type-specific annotations, a close clustering of UL with both AFS and NSP was identified in immune and blood-related components. Cross-trait meta-analysis revealed 15 loci shared between AFS/NSP and UL, including 7 novel SNPs. Univariable two-sample Mendelian randomization (MR) analysis suggested no evidence for a causal association between genetically predicted AFS/NSP and risk of UL, nor vice versa. Multivariable MR adjusting for age at menarche or/and age at natural menopause revealed a significant causal effect of genetically predicted higher AFS on a lower risk of UL. Such effect attenuated to null when age at first birth was further included. Utilizing participant-level data from the UK Biobank, one-sample MR based on genetic risk scores yielded consistent null findings among both pre-menopausal and post-menopausal females. From a genetic perspective, our study demonstrates an intrinsic link underlying sexual factors (AFS and NSP) and UL, highlighting shared biological mechanisms rather than direct causal effects. Future studies are needed to elucidate the specific mechanisms involved in the shared genetic influences and their potential impact on UL development.
Collapse
Affiliation(s)
- Xueyao Wu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
- National Cancer Institute, Rockville, Maryland, United States of America
| | - Changfeng Xiao
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Danielle Rasooly
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xunying Zhao
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cynthia Casson Morton
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Manchester Centre for Audiology and Deafness, Manchester Academic Health Science Center, University of Manchester, Manchester, United Kingdom
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Solna, Stockholm, Sweden
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - C. Scott Gallagher
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
3
|
Wang L, Sun T, Liu X, Wang Y, Qiao X, Chen N, Liu F, Zhou X, Wang H, Shen H. Myocarditis: A multi-omics approach. Clin Chim Acta 2024; 554:117752. [PMID: 38184138 DOI: 10.1016/j.cca.2023.117752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 12/29/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
Myocarditis, an inflammatory condition of weakened heart muscles often triggered by a variety of causes, that can result in heart failure and sudden death. Novel ways to enhance our understanding of myocarditis pathogenesis is available through newer modalities (omics). In this review, we examine the roles of various biomolecules and associated functional pathways across genomics, transcriptomics, proteomics, and metabolomics in the pathogenesis of myocarditis. Our analysis further explores the reproducibility and variability intrinsic to omics studies, underscoring the necessity and significance of employing a multi-omics approach to gain profound insights into myocarditis pathogenesis. This integrated strategy not only enhances our understanding of the disease, but also confirms the critical importance of a holistic multi-omics approach in disease analysis.
Collapse
Affiliation(s)
- Lulu Wang
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Tao Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Xiaolan Liu
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Yan Wang
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Xiaorong Qiao
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Nuo Chen
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Fangqian Liu
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Xiaoxiang Zhou
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Hua Wang
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Hongxing Shen
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China.
| |
Collapse
|
4
|
Jin U, Park SJ, Lee BG, Kim JB, Kim SJ, Joe EH, Woo HG, Park SM. Critical roles of parkin and PINK1 in coxsackievirus B3-induced viral myocarditis. Microbes Infect 2023; 25:105211. [PMID: 37574181 DOI: 10.1016/j.micinf.2023.105211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Viral myocarditis is an inflammatory disease of the myocardium, often leads to cardiac dysfunction and death. PARKIN (PRKN) and PINK1, well known as Parkinson's disease-associated genes, have been reported to be involved in innate immunity and mitochondrial damage control. Therefore, we investigated the role of parkin and PINK1 in coxsackievirus B3 (CVB3)-induced viral myocarditis because the etiology of myocarditis is related to abnormal immune response to viral infection and mitochondrial damage. After viral infection, the survival was significantly lower and myocardial damage was more severe in parkin knockout (KO) and PINK1 KO mice compared to wild-type (WT) mice. Parkin KO and PINK1 KO showed defective immune cell recruitment and impaired production of antiviral cytokines such as interferon-gamma, allowing increased viral replication. In addition, parkin KO and PINK1 KO mice were more susceptible to CVB3-induced mitochondrial damage than WT mice, resulting in susceptibility to viral-induced cardiac damage. Finally, using publicly available RNA-seq data, we found that pathogenic mutants of the PRKN gene are more common in patients with dilated cardiomyopathy and myocarditis than in controls or the general population. This study will help elucidate the molecular mechanism of CVB3-induced viral myocarditis.
Collapse
Affiliation(s)
- Uram Jin
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea; Department of Cardiology, Ajou University School of Medicine, Suwon, South Korea
| | - Soo Jin Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea; Department of Thoracic and Cardiovascular Surgery, Ajou University School of Medicine, Suwon, South Korea
| | - Byoung Gil Lee
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea; Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
| | - Jae-Bong Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea
| | - Soo Jeong Kim
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea
| | - Eun-Hye Joe
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea
| | - Hyun Goo Woo
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea; Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea.
| |
Collapse
|
5
|
Meade RK, Long JE, Jinich A, Rhee KY, Ashbrook DG, Williams RW, Sassetti CM, Smith CM. Genome-wide screen identifies host loci that modulate Mycobacterium tuberculosis fitness in immunodivergent mice. G3 (BETHESDA, MD.) 2023; 13:jkad147. [PMID: 37405387 PMCID: PMC10468300 DOI: 10.1093/g3journal/jkad147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/05/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023]
Abstract
Genetic differences among mammalian hosts and among strains of Mycobacterium tuberculosis (Mtb) are well-established determinants of tuberculosis (TB) patient outcomes. The advent of recombinant inbred mouse panels and next-generation transposon mutagenesis and sequencing approaches has enabled dissection of complex host-pathogen interactions. To identify host and pathogen genetic determinants of Mtb pathogenesis, we infected members of the highly diverse BXD family of strains with a comprehensive library of Mtb transposon mutants (TnSeq). Members of the BXD family segregate for Mtb-resistant C57BL/6J (B6 or B) and Mtb-susceptible DBA/2J (D2 or D) haplotypes. The survival of each bacterial mutant was quantified within each BXD host, and we identified those bacterial genes that were differentially required for Mtb fitness across BXD genotypes. Mutants that varied in survival among the host family of strains were leveraged as reporters of "endophenotypes," each bacterial fitness profile directly probing specific components of the infection microenvironment. We conducted quantitative trait loci (QTL) mapping of these bacterial fitness endophenotypes and identified 140 host-pathogen QTL (hpQTL). We located a QTL hotspot on chromosome 6 (75.97-88.58 Mb) associated with the genetic requirement of multiple Mtb genes: Rv0127 (mak), Rv0359 (rip2), Rv0955 (perM), and Rv3849 (espR). Together, this screen reinforces the utility of bacterial mutant libraries as precise reporters of the host immunological microenvironment during infection and highlights specific host-pathogen genetic interactions for further investigation. To enable downstream follow-up for both bacterial and mammalian genetic research communities, all bacterial fitness profiles have been deposited into GeneNetwork.org and added into the comprehensive collection of TnSeq libraries in MtbTnDB.
Collapse
Affiliation(s)
- Rachel K Meade
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | - Jarukit E Long
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01655, USA
- Research Animal Diagnostic Services, Charles River Laboratories, Wilmington, MA 01887, USA
| | - Adrian Jinich
- Division of Infectious Diseases, Weill Cornell Medical College, New York, NY 10021, USA
| | - Kyu Y Rhee
- Division of Infectious Diseases, Weill Cornell Medical College, New York, NY 10021, USA
| | - David G Ashbrook
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Christopher M Sassetti
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Clare M Smith
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| |
Collapse
|
6
|
Wang J, Zhang X, Yang X, Yu H, Bu M, Fu J, Zhang Z, Xu H, Hu J, Lu J, Zhang H, Zhai Z, Yang W, Wu X, Wang Y, Tong Q. Revitalizing myocarditis treatment through gut microbiota modulation: unveiling a promising therapeutic avenue. Front Cell Infect Microbiol 2023; 13:1191936. [PMID: 37260696 PMCID: PMC10229058 DOI: 10.3389/fcimb.2023.1191936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/24/2023] [Indexed: 06/02/2023] Open
Abstract
Numerous studies have demonstrated that gut microbiota plays an important role in the development and treatment of different cardiovascular diseases, including hypertension, heart failure, myocardial infarction, arrhythmia, and atherosclerosis. Furthermore, evidence from recent studies has shown that gut microbiota contributes to the development of myocarditis. Myocarditis is an inflammatory disease that often results in myocardial damage. Myocarditis is a common cause of sudden cardiac death in young adults. The incidence of myocarditis and its associated dilated cardiomyopathy has been increasing yearly. Myocarditis has gained significant attention on social media due to its association with both COVID-19 and COVID-19 vaccinations. However, the current therapeutic options for myocarditis are limited. In addition, little is known about the potential therapeutic targets of myocarditis. In this study, we review (1) the evidence on the gut-heart axis, (2) the crosslink between gut microbiota and the immune system, (3) the association between myocarditis and the immune system, (4) the impact of gut microbiota and its metabolites on myocarditis, (5) current strategies for modulating gut microbiota, (6) challenges and future directions for targeted gut microbiota in the treatment of myocarditis. The approach of targeting the gut microbiota in myocarditis is still in its infancy, and this is the study to explore the gut microbiota-immune system-myocarditis axis. Our findings are expected to pave the way for the use of gut microbiota as a potential therapeutic target in the treatment of myocarditis.
Collapse
Affiliation(s)
- Jingyue Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Xianfeng Zhang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Xinyu Yang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Hang Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Mengmeng Bu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jie Fu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Zhengwei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Hui Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jiachun Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jinyue Lu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Haojian Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Zhao Zhai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Wei Yang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xiaodan Wu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Qian Tong
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
7
|
Reuter SP, Soonpaa MH, Field D, Simpson E, Rubart-von der Lohe M, Lee HK, Sridhar A, Ware SM, Green N, Li X, Ofner S, Marchuk DA, Wollert KC, Field LJ. Cardiac Troponin I-Interacting Kinase Affects Cardiomyocyte S-Phase Activity but Not Cardiomyocyte Proliferation. Circulation 2023; 147:142-153. [PMID: 36382596 PMCID: PMC9839600 DOI: 10.1161/circulationaha.122.061130] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Identifying genetic variants that affect the level of cell cycle reentry and establishing the degree of cell cycle progression in those variants could help guide development of therapeutic interventions aimed at effecting cardiac regeneration. We observed that C57Bl6/NCR (B6N) mice have a marked increase in cardiomyocyte S-phase activity after permanent coronary artery ligation compared with infarcted DBA/2J (D2J) mice. METHODS Cardiomyocyte cell cycle activity after infarction was monitored in D2J, (D2J×B6N)-F1, and (D2J×B6N)-F1×D2J backcross mice by means of bromodeoxyuridine or 5-ethynyl-2'-deoxyuridine incorporation using a nuclear-localized transgenic reporter to identify cardiomyocyte nuclei. Genome-wide quantitative trait locus analysis, fine scale genetic mapping, whole exome sequencing, and RNA sequencing analyses of the backcross mice were performed to identify the gene responsible for the elevated cardiomyocyte S-phase phenotype. RESULTS (D2J×B6N)-F1 mice exhibited a 14-fold increase in cardiomyocyte S-phase activity in ventricular regions remote from infarct scar compared with D2J mice (0.798±0.09% versus 0.056±0.004%; P<0.001). Quantitative trait locus analysis of (D2J×B6N)-F1×D2J backcross mice revealed that the gene responsible for differential S-phase activity was located on the distal arm of chromosome 3 (logarithm of the odds score=6.38; P<0.001). Additional genetic and molecular analyses identified 3 potential candidates. Of these, Tnni3k (troponin I-interacting kinase) is expressed in B6N hearts but not in D2J hearts. Transgenic expression of TNNI3K in a D2J genetic background results in elevated cardiomyocyte S-phase activity after injury. Cardiomyocyte S-phase activity in both Tnni3k-expressing and Tnni3k-nonexpressing mice results in the formation of polyploid nuclei. CONCLUSIONS These data indicate that Tnni3k expression increases the level of cardiomyocyte S-phase activity after injury.
Collapse
Affiliation(s)
- Sean P. Reuter
- Krannert Cardiovascular Research Center, Indiana University School of Medicine
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Mark H. Soonpaa
- Krannert Cardiovascular Research Center, Indiana University School of Medicine
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Dorothy Field
- Krannert Cardiovascular Research Center, Indiana University School of Medicine
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Ed Simpson
- Center for Computational Biology & Bioinformatics, Indiana University School of Medicine
| | | | - Han Kyu Lee
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine
| | - Arthi Sridhar
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Stephanie M. Ware
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| | - Nick Green
- Center for Computational Biology & Bioinformatics, Indiana University School of Medicine
| | - Xiaochun Li
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine
| | - Susan Ofner
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine
| | - Douglas A. Marchuk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine
| | - Kai C. Wollert
- Department of Cardiology and Angiology, Division of Molecular and Translational Cardiology, Hannover Medical School
| | - Loren J. Field
- Krannert Cardiovascular Research Center, Indiana University School of Medicine
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine
| |
Collapse
|
8
|
Qu H, Zhang Y, Zhang W, Zhu Y, Xu R. Knockout of Cardiac Troponin I-Interacting Kinase leads to cardiac dysfunction and remodeling. Clin Exp Pharmacol Physiol 2022; 49:1169-1178. [PMID: 35781726 DOI: 10.1111/1440-1681.13699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022]
Abstract
Cardiac troponin I-interacting kinase (TNNI3K) is a cardiac-specific kinase that has been identified as a diagnostic marker and a therapeutic target in cardiovascular diseases. However, the biological function of TNNI3K in cardiac dysfunction and remodeling remain elusive. In the present study, a Tnni3k cardiomyocyte-specific knockout (Tnni3k-cKO) mouse model was established. Echocardiography was used to evaluate cardiac function in mice. Heart failure markers were detected using enzyme-linked immunosorbent assay. Hematoxylin and eosin staining, wheat germ agglutinin staining, Masson's trichrome staining, Sirius red staining, and TUNEL staining were used to assess histopathological changes, cardiac hypertrophy, collagen deposition, and myocardial apoptosis, respectively. Expression levels of TNNI3K, apoptosis-related proteins, and p38 mitogen-activated protein kinase were measured using Western blot analysis. Compared to wild-type controls, cardiac dysfunction and cardiac remodeling of Tnni3k-cKO mice increased gradually with age. Tnni3k-cKO mice exhibited cardiac hypertrophy, cardiac fibrosis, and cardiomyocyte apoptosis. Upregulation of cleaved caspase-3 in Tnni3k-cKO mice appeared to be related to phosphorylation and activation of the p38 mitogen-activated protein kinase signaling pathway. In conclusion, this study shows that TNNI3K is essential for cardiac development and function, providing new insights into the development of novel therapeutic strategies for cardiac diseases.
Collapse
Affiliation(s)
- Huilin Qu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Zhang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Zhang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaxin Zhu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruixia Xu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Smatti MK, Alkhatib HA, Al Thani AA, Yassine HM. Will Host Genetics Affect the Response to SARS-CoV-2 Vaccines? Historical Precedents. Front Med (Lausanne) 2022; 9:802312. [PMID: 35360730 PMCID: PMC8962369 DOI: 10.3389/fmed.2022.802312] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/10/2022] [Indexed: 11/25/2022] Open
Abstract
Recent progress in genomics and bioinformatics technologies have allowed for the emergence of immunogenomics field. This intersection of immunology and genetics has broadened our understanding of how the immune system responds to infection and vaccination. While the immunogenetic basis of the huge clinical variability in response to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is currently being extensively studied, the host genetic determinants of SARS-CoV-2 vaccines remain largely unknown. Previous reports evidenced that vaccines may not protect all populations or individuals equally, due to multiple host- and vaccine-specific factors. Several studies on vaccine response to measles, rubella, hepatitis B, smallpox, and influenza highlighted the contribution of genetic mutations or polymorphisms in modulating the innate and adaptive immunity following vaccination. Specifically, genetic variants in genes encoding virus receptors, antigen presentation, cytokine production, or related to immune cells activation and differentiation could influence how an individual responds to vaccination. Although such knowledge could be utilized to generate personalized vaccine strategies to optimize the vaccine response, studies in this filed are still scarce. Here, we briefly summarize the scientific literature related to the immunogenetic determinants of vaccine-induced immunity, highlighting the possible role of host genetics in response to SARS-CoV-2 vaccines as well.
Collapse
Affiliation(s)
- Maria K. Smatti
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | | | - Hadi M. Yassine
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
10
|
Seidman MA, McManus B. Myocarditis. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00005-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
11
|
Pham C, Muñoz-Martín N, Lodder EM. The Diverse Roles of TNNI3K in Cardiac Disease and Potential for Treatment. Int J Mol Sci 2021; 22:6422. [PMID: 34203974 PMCID: PMC8232738 DOI: 10.3390/ijms22126422] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/25/2022] Open
Abstract
In the two decades since the discovery of TNNI3K it has been implicated in multiple cardiac phenotypes and physiological processes. TNNI3K is an understudied kinase, which is mainly expressed in the heart. Human genetic variants in TNNI3K are associated with supraventricular arrhythmias, conduction disease, and cardiomyopathy. Furthermore, studies in mice implicate the gene in cardiac hypertrophy, cardiac regeneration, and recovery after ischemia/reperfusion injury. Several new papers on TNNI3K have been published since the last overview, broadening the clinical perspective of TNNI3K variants and our understanding of the underlying molecular biology. We here provide an overview of the role of TNNI3K in cardiomyopathy and arrhythmia covering both a clinical perspective and basic science advancements. In addition, we review the potential of TNNI3K as a target for clinical treatments in different cardiac diseases.
Collapse
Affiliation(s)
| | | | - Elisabeth M. Lodder
- Department of Clinical and Experimental Cardiology, Heart Center, University of Amsterdam, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands; (C.P.); (N.M.-M.)
| |
Collapse
|
12
|
Singh A, Mehrotra A, Gondro C, Romero ARDS, Pandey AK, Karthikeyan A, Bashir A, Mishra BP, Dutt T, Kumar A. Signatures of Selection in Composite Vrindavani Cattle of India. Front Genet 2020; 11:589496. [PMID: 33391343 PMCID: PMC7775581 DOI: 10.3389/fgene.2020.589496] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
Vrindavani is an Indian composite cattle breed developed by crossbreeding taurine dairy breeds with native indicine cattle. The constituent breeds were selected for higher milk production and adaptation to the tropical climate. However, the selection response for production and adaptation traits in the Vrindavani genome is not explored. In this study, we provide the first overview of the selection signatures in the Vrindavani genome. A total of 96 Vrindavani cattle were genotyped using the BovineSNP50 BeadChip and the SNP genotype data of its constituent breeds were collected from a public database. Within-breed selection signatures in Vrindavani were investigated using the integrated haplotype score (iHS). The Vrindavani breed was also compared to each of its parental breeds to discover between-population signatures of selection using two approaches, cross-population extended haplotype homozygosity (XP-EHH) and fixation index (FST). We identified 11 common regions detected by more than one method harboring genes such as LRP1B, TNNI3K, APOB, CACNA2D1, FAM110B, and SPATA17 associated with production and adaptation. Overall, our results suggested stronger selective pressure on regions responsible for adaptation compared to milk yield.
Collapse
Affiliation(s)
- Akansha Singh
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, India
| | - Arnav Mehrotra
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, India
| | - Cedric Gondro
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | | | - Ashwni Kumar Pandey
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, India
| | - A Karthikeyan
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, India
| | - Aamir Bashir
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, India
| | - B P Mishra
- Animal Biotechnology, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, India
| | - Triveni Dutt
- Livestock Production and Management, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, India
| | - Amit Kumar
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, India
| |
Collapse
|
13
|
Lempainen J, Korhonen LS, Kantojärvi K, Heinonen S, Toivonen L, Räty P, Ramilo O, Mejias A, Laine AP, Vuorinen T, Waris M, Karlsson L, Karlsson H, Paunio T, Peltola V. Associations Between IFI44L Gene Variants and Rates of Respiratory Tract Infections During Early Childhood. J Infect Dis 2020; 223:157-165. [DOI: 10.1093/infdis/jiaa341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
Abstract
Background
Genetic heterogeneity in type I interferon (IFN)–related gene IFI44L may account for variable susceptibility to respiratory tract infections (RTIs) in children.
Methods
In 2 prospective, population-based birth cohorts, the STEPS Study and the FinnBrain Birth Cohort Study, IFI44L genotypes for rs273259 and rs1333969 were determined in relation to the development of RTIs until 1 or 2 years of age, respectively. At age 3 months, whole-blood transcriptional profiles were analyzed and nasal samples were tested for respiratory viruses in a subset of children.
Results
In the STEPS Study (n = 1135), IFI44L minor/minor gene variants were associated with lower rates of acute otitis media episodes (adjusted incidence rate ratio, 0.77 [95% confidence interval, .61–.96] for rs273259 and 0.74 [.55–.99] for rs1333969) and courses of antibiotics for RTIs (0.76 [.62–.95] and 0.73 [.56–.97], respectively. In the FinnBrain cohort (n = 971), IFI44L variants were associated with lower rates of RTIs and courses of antibiotics for RTIs. In respiratory virus–positive 3-month-old children, IFI44L gene variants were associated with decreased expression levels of IFI44L and several other IFN-related genes.
Conclusions
Variant forms of IFI44L gene were protective against early-childhood RTIs or acute otitis media, and they attenuated IFN pathway activation by respiratory viruses.
Collapse
Affiliation(s)
- Johanna Lempainen
- Department of Paediatrics and Adolescent Medicine, University of Turku and Turku University Hospital, Turku, Finland
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Laura S Korhonen
- Department of Paediatrics and Adolescent Medicine, University of Turku and Turku University Hospital, Turku, Finland
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Katri Kantojärvi
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Finnish Institute for Health and Welfare, Genomics and Biobank Unit, Helsinki, Finland
- Department of Psychiatry and SleepWell Research Program, Faculty of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Santtu Heinonen
- New Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Laura Toivonen
- Department of Paediatrics and Adolescent Medicine, University of Turku and Turku University Hospital, Turku, Finland
| | - Panu Räty
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Octavio Ramilo
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Asuncion Mejias
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Antti-Pekka Laine
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tytti Vuorinen
- Institute of Biomedicine, University of Turku and Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Matti Waris
- Institute of Biomedicine, University of Turku and Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Child Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
| | - Tiina Paunio
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Finnish Institute for Health and Welfare, Genomics and Biobank Unit, Helsinki, Finland
- Department of Psychiatry and SleepWell Research Program, Faculty of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Ville Peltola
- Department of Paediatrics and Adolescent Medicine, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
14
|
Sánchez-Molano E, Bay V, Smith RF, Oikonomou G, Banos G. Quantitative Trait Loci Mapping for Lameness Associated Phenotypes in Holstein-Friesian Dairy Cattle. Front Genet 2019; 10:926. [PMID: 31636655 PMCID: PMC6787292 DOI: 10.3389/fgene.2019.00926] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 09/05/2019] [Indexed: 01/08/2023] Open
Abstract
Lameness represents a significant challenge for the dairy cattle industry, resulting in economic losses and reduced animal health and welfare. The existence of underlying genomic variation for lameness associated traits has the potential to improve selection strategies by using genomic markers. Therefore, the aim of this study was to identify genomic regions and potential candidate genes associated with lameness traits. Lameness related lesions and digital cushion thickness were studied using records collected by our research team, farm records, and a combination of both. Genome-wide analyses were performed to identify significant genomic effects, and a combination of single SNP association analysis and regional heritability mapping was used to identify associated genomic regions. Significant genomic effects were identified for several lameness related traits: Two genomic regions were identified on chromosome 3 associated with digital dermatitis and interdigital hyperplasia, one genomic region on chromosome 23 associated with interdigital hyperplasia, and one genomic region on chromosome 2 associated with sole haemorrhage. Candidate genes in those regions are mainly related to immune response and fibroblast proliferation. Quantitative trait loci (QTL) identified in this study could enlighten the understanding of lameness pathogenesis, providing an opportunity to improve health and welfare in dairy cattle with the addition of these regions into selection programs.
Collapse
Affiliation(s)
- Enrique Sánchez-Molano
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Edinburgh, United Kingdom
| | - Veysel Bay
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Bandirma Sheep Research Institute, The Ministry of Agriculture and Forestry, Balikesir, Turkey
| | - Robert F Smith
- Institute of Veterinary Science, University of Liverpool, Leahurst Campus, Liverpool, United Kingdom
| | - Georgios Oikonomou
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Institute of Veterinary Science, University of Liverpool, Leahurst Campus, Liverpool, United Kingdom
| | - Georgios Banos
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Edinburgh, United Kingdom.,The Roslin Institute Building, Scotland's Rural College, Easter Bush, Edinburgh, United Kingdom
| |
Collapse
|
15
|
Hosseini SMJ, Mirhosseini SM, Taghian M, Salehi M, Farahani MM, Bakhtiari F, Ghasemi-Pirbaluti M, Motaghi E. First evidence of the presence of adenovirus type 8 in myocardium of patients with severe idiopathic dilated cardiomyopathy. Arch Virol 2018; 163:2895-2897. [PMID: 30022238 DOI: 10.1007/s00705-018-3942-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 07/02/2018] [Indexed: 01/04/2023]
Abstract
Previous studies have detected adenovirus and cytomegalovirus (CMV) in cardiac tissue of patients with myocarditis. Therefore, in this study, we investigated the frequency of these viruses, which may be involved in the development of severe dilated cardiomyopathy (DCM). Myocardial tissue from of 23 cardiac transplant candidates with acute idiopathic DCM below the age of 40 years were analyzed by amplification of adenovirus and CMV DNA and subsequent sequencing. Adenovirus was detected in four (17.4%) and CMV in one (4.3%) of the patients. All controls were negative for the presence of both viruses. Our study shows that myocardial infection with adenovirus may play an important role in the pathogenesis of severe DCM and suggests that vaccination against adenovirus might be helpful in decreasing the prevalence of severe idiopathic DCM. This is the first study in which adenovirus type 8 has been detected in the hearts of patients with DCM.
Collapse
Affiliation(s)
| | | | - Mehdi Taghian
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | - Mehrdad Salehi
- Cardiac Surgery Ward, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Fariba Bakhtiari
- Department of Microbiology, Faculty of Advanced Science and Technologies, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | | | - Ehsan Motaghi
- Department of Physiology and Pharmacology, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
16
|
Role of gene polymorphisms/haplotypes and serum levels of interleukin-17A in susceptibility to viral myocarditis. Exp Mol Pathol 2018. [PMID: 29530464 DOI: 10.1016/j.yexmp.2018.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interleukin-17A (IL-17A) has been implicated in the pathogenesis of viral myocarditis (VMC). However, the role of IL-17A polymorphisms in susceptibility to VMC has not been reported to date. The aim of this study was to explore the association between IL-17A variants as well as serum IL-17 levels with VMC. Three single-nucleotide polymorphisms (SNPs) (rs2275913, rs3819025, and rs3748067) were analyzed by the polymerase chain reaction-restriction fragment length polymorphism method in 236 VMC patients and 259 controls from China. Serum IL-17A levels were measured by enzyme-linked immunosorbent assay kits. Multivariable logistic regression analysis that the rs2275913 AA genotype and the haplotype -197A/+45G/+1249G (AGG) were associated with an increased risk of VMC (all P < 0.05). Consistent with these findings, the rs2275913 AA genotype was linked to higher serum IL-17A compared to GG/AG genotype (all P < 0.001). We observed no associations between the other two SNPs and risk of VMC. Serum IL-17A levels were significantly higher in the VMC group than controls (P < 0.001) and gradually increased with the increase of New York Heart Association grade in VMC patients (P < 0.05). Spearman correlation test revealed that the serum IL-17A level was correlated with the cardiac damage and left ventricular systolic functions among VMC patients (all P < 0.05). Our study reveals that IL-17A expression may contribute to the development and severity of VMC. The SNP rs2275913 in the IL-17A gene might exert influence on susceptibility to VMC via linking with the serum IL-17A level.
Collapse
|
17
|
Autoimmune-Disease-Prone NOD Mice Help To Reveal a New Genetic Locus for Reducing Pulmonary Disease Caused by Mycoplasma pulmonis. Infect Immun 2018; 86:IAI.00812-17. [PMID: 29263105 DOI: 10.1128/iai.00812-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 12/07/2017] [Indexed: 11/20/2022] Open
Abstract
Mycoplasmas are bacterial pathogens of a range of animals, including humans, and are a common cause of respiratory disease. However, the host genetic factors that affect resistance to infection or regulate the resulting pulmonary inflammation are not well defined. We and others have previously demonstrated that nonobese diabetic (NOD) mice can be used to investigate disease loci that affect bacterial infection and autoimmune diabetes. Here we show that NOD mice are more susceptible than C57BL/6 (B6) mice to infection with Mycoplasma pulmonis, a natural model of pulmonary mycoplasmosis. The lungs of infected NOD mice had higher loads of M. pulmonis and more severe inflammatory lesions. Moreover, congenic NOD mice that harbored different B6-derived chromosomal intervals enabled identification and localization of a new mycoplasmosis locus, termed Mpr2, on chromosome 13. These congenic NOD mice demonstrated that the B6 allele for Mpr2 reduced the severity of pulmonary inflammation caused by infection with M. pulmonis and that this was associated with altered cytokine and chemokine concentrations in the infected lungs. Mpr2 also colocalizes to the same genomic interval as Listr2 and Idd14, genetic loci linked to listeriosis resistance and autoimmune diabetes susceptibility, respectively, suggesting that allelic variation within these loci may affect the development of both infectious and autoimmune disease.
Collapse
|
18
|
Neupane B, Zhou Q, Gawaz M, Gramlich M. Personalized medicine in inflammatory cardiomyopathy. Per Med 2018; 15:127-136. [PMID: 29714129 DOI: 10.2217/pme-2017-0074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Inflammatory cardiomyopathy is the result of persistent myocardial inflammation which can arise from both infectious or noninfectious causes. While most patients with acute myocarditis recover, up to 20% develop inflammatory cardiomyopathy with chronic heart failure. The interaction between host factors and factors of the agent that triggered myocardial inflammation must be considered to fully understand the individual mechanism of disease. Several inflammatory biomarkers, histology, immunohistochemistry, advanced imaging technologies as well as molecular high-throughput sequencing techniques help to identify disease pathways and to establish a comprehensive, individualized treatment approach, which can include anti-inflammatory medication, antiviral drugs and heart failure therapy. This might help to prevent transition from acute inflammation to persistent heart failure and to restore cardiac function.
Collapse
Affiliation(s)
- Balram Neupane
- Department of Cardiology & Cardiovascular Diseases, Eberhard Karls University, Tübingen, Germany
| | - Qifeng Zhou
- Department of Cardiology & Cardiovascular Diseases, Eberhard Karls University, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology & Cardiovascular Diseases, Eberhard Karls University, Tübingen, Germany
| | - Michael Gramlich
- Department of Cardiology & Cardiovascular Diseases, Eberhard Karls University, Tübingen, Germany
| |
Collapse
|
19
|
Autosomal Recessive Cardiomyopathy Presenting as Acute Myocarditis. J Am Coll Cardiol 2017; 69:1653-1665. [PMID: 28359509 DOI: 10.1016/j.jacc.2017.01.043] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/11/2016] [Accepted: 01/19/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Myocarditis is inflammation of the heart muscle that can follow various viral infections. Why children only rarely develop life-threatening acute viral myocarditis (AVM), given that the causal viral infections are common, is unknown. Genetic lesions might underlie such susceptibilities. Mouse genetic studies demonstrated that interferon (IFN)-α/β immunity defects increased susceptibility to virus-induced myocarditis. Moreover, variations in human TLR3, a potent inducer of IFNs, were proposed to underlie AVM. OBJECTIVES This study sought to evaluate the hypothesis that human genetic factors may underlie AVM in previously healthy children. METHODS We tested the role of TLR3-IFN immunity using human induced pluripotent stem cell-derived cardiomyocytes. We then performed whole-exome sequencing of 42 unrelated children with acute myocarditis (AM), some with proven viral causes. RESULTS We found that TLR3- and STAT1-deficient cardiomyocytes were not more susceptible to Coxsackie virus B3 (CVB3) infection than control cells. Moreover, CVB3 did not induce IFN-α/β and IFN-α/β-stimulated genes in control cardiomyocytes. Finally, exogenous IFN-α did not substantially protect cardiomyocytes against CVB3. We did not observe a significant enrichment of rare variations in TLR3- or IFN-α/β-related genes. Surprisingly, we found that homozygous but not heterozygous rare variants in genes associated with inherited cardiomyopathies were significantly enriched in AM-AVM patients compared with healthy individuals (p = 2.22E-03) or patients with other diseases (p = 1.08E-04). Seven of 42 patients (16.7%) carried rare biallelic (homozygous or compound heterozygous) nonsynonymous or splice-site variations in 6 cardiomyopathy-associated genes (BAG3, DSP, PKP2, RYR2, SCN5A, or TNNI3). CONCLUSIONS Previously silent recessive defects of the myocardium may predispose to acute heart failure presenting as AM, notably after common viral infections in children.
Collapse
|
20
|
Genome-wide associations of CD46 and IFI44L genetic variants with neutralizing antibody response to measles vaccine. Hum Genet 2017; 136:421-435. [PMID: 28289848 DOI: 10.1007/s00439-017-1768-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/14/2017] [Indexed: 12/27/2022]
Abstract
Population-based studies have revealed 2-10% measles vaccine failure rate even after two vaccine doses. While the mechanisms behind this remain unknown, we hypothesized that host genetic factors are likely to be involved. We performed a genome-wide association study of measles specific neutralizing antibody and IFNγ ELISPOT response in a combined sample of 2872 subjects. We identified two distinct chromosome 1 regions (previously associated with MMR-related febrile seizures), associated with vaccine-induced measles neutralizing antibody titers. The 1q32 region contained 20 significant SNPs in/around the measles virus receptor-encoding CD46 gene, including the intronic rs2724384 (p value = 2.64 × 10-09) and rs2724374 (p value = 3.16 × 10-09) SNPs. The 1q31.1 region contained nine significant SNPs in/around IFI44L, including the intronic rs1333973 (p value = 1.41 × 10-10) and the missense rs273259 (His73Arg, p value = 2.87 × 10-10) SNPs. Analysis of differential exon usage with mRNA-Seq data and RT-PCR suggests the involvement of rs2724374 minor G allele in the CD46 STP region exon B skipping, resulting in shorter CD46 isoforms. Our study reveals common CD46 and IFI44L SNPs associated with measles-specific humoral immunity, and highlights the importance of alternative splicing/virus cellular receptor isoform usage as a mechanism explaining inter-individual variation in immune response after live measles vaccine.
Collapse
|
21
|
Podliesna S, Bezzina CR, Lodder EM. Complex Genetics of Cardiovascular Traits in Mice: F2-Mapping of QTLs and Their Underlying Genes. Methods Mol Biol 2017; 1488:431-454. [PMID: 27933537 DOI: 10.1007/978-1-4939-6427-7_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In this chapter, we will use the example of the identification of Tnni3k as a modulator of cardiac conduction to introduce you to the use of a murine F2-generation intercross as a powerful method for the identification of novel genes relevant for cardiovascular traits. Murine F2-progeny is a genetically diverse panel of mice with differences in phenotype manifestations, e.g. cardiovascular traits such as cardiomyopathy and ECG parameters. This chapter discusses the best strategies for using F2-mice for genetic mapping. Moreover, we provide an example of the feasibility of identification of new genes modulating cardiac function utilizing the technique of mapping quantitative trait loci (QTLs) and a systems genetics integration of available genetic, gene expression, and phenotypic data.
Collapse
Affiliation(s)
- Svitlana Podliesna
- Department of Clinical and Experimental Cardiology, Academic Medical Centre (AMC), University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Academic Medical Centre (AMC), University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Elisabeth M Lodder
- Department of Clinical and Experimental Cardiology, Academic Medical Centre (AMC), University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Tsunoda I, Sato F, Omura S, Fujita M, Sakiyama N, Park AM. Three immune-mediated disease models induced by Theiler's virus: Multiple sclerosis, seizures and myocarditis. CLINICAL & EXPERIMENTAL NEUROIMMUNOLOGY 2016; 7:330-345. [PMID: 28603559 PMCID: PMC5464738 DOI: 10.1111/cen3.12341] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Theiler's murine encephalomyelitis virus (TMEV) infection has been used as a viral model for multiple sclerosis (MS), as TMEV can induce chronic inflammatory demyelinating lesions with viral persistence in the spinal cord of SJL/J mice. In contrast, when C57BL/6 mice are infected with TMEV, the mice can clear the virus from the central nervous system (CNS), without viral persistence or demyelination, but develop seizures and hippocampal sclerosis, which has been used as a viral model for seizures/epilepsy. In the two TMEV-induced CNS disease models, not only viral infection, but also immune responses contribute to the pathogenesis. Interestingly, acquired immunity plays an effector role in the MS model, whereas innate immunity appears to contribute to the development of seizures. Recently, we have established the third TMEV-induced disease model, a mouse model for viral myocarditis, using C3H mice. TMEV-induced myocarditis is a triphasic disease, which mimics human myocarditis; phase I, mediated by viral replication in the heart and innate immunity; phase II, mediated by acquired immunity; and phase III, resulted from cardiac fibrosis. The genetic susceptibility to the aforementioned three models (MS, seizures and myocarditis) differs among mouse strains. We have compared and contrasted the three models induced by one single pathogen, TMEV, particularly in regard to the roles of T helper cells and natural killer T cells, which will give an insight into how interactions between the immune system and the host's genetic background determine the tissue tropism of virus and the development of virus-induced organ-specific immunopathology.
Collapse
Affiliation(s)
- Ikuo Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Fumitaka Sato
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Seiichi Omura
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Mitsugu Fujita
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Namie Sakiyama
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Ah-Mee Park
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| |
Collapse
|
23
|
Abstract
As a greater proportion of patients survive their initial cardiac insult, medical systems worldwide are being faced with an ever-growing need to understand the mechanisms behind the pathogenesis of chronic heart failure (HF). There is a wealth of information about the role of inflammatory cells and pathways during acute injury and the reparative processes that are subsequently activated. We discuss the different causes that lead to chronic HF development and how the sum of initial inflammatory and reparative responses only sets the trajectory for disease progression. Unfortunately, comparatively little is known about the contribution of the immune system once the trajectory has been set, and chronic HF has been established—which clinically represents the majority of patients. It is known that chronic HF is associated with circulating inflammatory cytokines that can predict clinical outcomes, yet the causative role inflammation plays in disease progression is not well defined, and the majority of clinical trials that target aspects of inflammation in patients with chronic HF have largely been negative. This review will present what is currently known about inflammation in chronic HF in both humans and animal models as a means to highlight the gap in our knowledge base that requires further examination.
Collapse
Affiliation(s)
- Sarah A. Dick
- From the Division of Cardiology, Department of Medicine, University Health Network, Toronto, Ontario, Canada (S.A.D, S.E.); University of Toronto, Toronto, Ontario, Canada (S.E); Peter Munk Cardiac Centre, Toronto, Ontario, Canada (S.A.D, S.E.); and Toronto General Hospital Research Institute, Toronto, Ontario, Canada (S.A.D, S.E.)
| | - Slava Epelman
- From the Division of Cardiology, Department of Medicine, University Health Network, Toronto, Ontario, Canada (S.A.D, S.E.); University of Toronto, Toronto, Ontario, Canada (S.E); Peter Munk Cardiac Centre, Toronto, Ontario, Canada (S.A.D, S.E.); and Toronto General Hospital Research Institute, Toronto, Ontario, Canada (S.A.D, S.E.)
| |
Collapse
|
24
|
Marton J, Albert D, Wiltshire SA, Park R, Bergen A, Qureshi S, Malo D, Burelle Y, Vidal SM. Cyclosporine A Treatment Inhibits Abcc6-Dependent Cardiac Necrosis and Calcification following Coxsackievirus B3 Infection in Mice. PLoS One 2015; 10:e0138222. [PMID: 26375467 PMCID: PMC4574283 DOI: 10.1371/journal.pone.0138222] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/26/2015] [Indexed: 11/18/2022] Open
Abstract
Coxsackievirus type B3 (CVB3) is a cardiotropic enterovirus. Infection causes cardiomyocyte necrosis and myocardial inflammation. The damaged tissue that results is replaced with fibrotic or calcified tissue, which can lead to permanently altered cardiac function. The extent of pathogenesis among individuals exposed to CVB3 is dictated by a combination of host genetics, viral virulence, and the environment. Here, we aimed to identify genes that modulate cardiopathology following CVB3 infection. 129S1 mice infected with CVB3 developed increased cardiac pathology compared to 129X1 substrain mice despite no difference in viral burden. Linkage analysis identified a major locus on chromosome 7 (LOD: 8.307, P<0.0001) that controlled the severity of cardiac calcification and necrosis following infection. Sub-phenotyping and genetic complementation assays identified Abcc6 as the underlying gene. Microarray expression profiling identified genotype-dependent regulation of genes associated with mitochondria. Electron microscopy examination showed elevated deposition of hydroxyapatite-like material in the mitochondrial matrices of infected Abcc6 knockout (Abcc6-/-) mice but not in wildtype littermates. Cyclosporine A (CsA) inhibits mitochondrial permeability transition pore opening by inhibiting cyclophilin D (CypD). Treatment of Abcc6 -/- mice with CsA reduced cardiac necrosis and calcification by more than half. Furthermore, CsA had no effect on the CVB3-induced phenotype of doubly deficient CypD-/-Abcc6-/- mice. Altogether, our work demonstrates that mutations in Abcc6 render mice more susceptible to cardiac calcification following CVB3 infection. Moreover, we implicate CypD in the control of cardiac necrosis and calcification in Abcc6-deficient mice, whereby CypD inhibition is required for cardioprotection.
Collapse
Affiliation(s)
- Jennifer Marton
- Department of Human Genetics and Complex Traits Group, McGill University, Montreal, Canada
| | - Danica Albert
- Department of Human Genetics and Complex Traits Group, McGill University, Montreal, Canada
| | - Sean A. Wiltshire
- Department of Human Genetics and Complex Traits Group, McGill University, Montreal, Canada
| | - Robin Park
- Department of Human Genetics and Complex Traits Group, McGill University, Montreal, Canada
| | - Arthur Bergen
- Department of Ophthalmogenetics, The Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Salman Qureshi
- The Center for Host Resistance and the Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Danielle Malo
- Department of Human Genetics and Complex Traits Group, McGill University, Montreal, Canada
| | - Yan Burelle
- Faculty of Pharmacy, University of Montreal, Montreal, Canada
| | - Silvia M. Vidal
- Department of Human Genetics and Complex Traits Group, McGill University, Montreal, Canada
| |
Collapse
|
25
|
Activation and function of murine primary microglia in the absence of the prion protein. J Neuroimmunol 2015; 286:25-32. [PMID: 26298321 DOI: 10.1016/j.jneuroim.2015.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/02/2015] [Accepted: 07/04/2015] [Indexed: 11/21/2022]
Abstract
The prion protein (PrP(C)) is predominantly expressed in the nervous and immune systems and is involved in relevant cell signaling. Microglia participate in neuroimmune interactions, and their regulatory mechanisms are critical for both health and disease. Despite recent reports with a microglial cell line, little is known about the relevance of PrP(C) in brain microglia. We investigated the role of PrP(C) in mouse primary microglia, and found no differences between wild type and Prnp-null cells in cell morphology or the expression of a microglial marker. Translocation of NF-κB to the nucleus also did not differ, nor did cytokine production. The levels of iNOS were also similar and, finally, microglia of either genotype showed no differences in either rates of phagocytosis or migration, even following activation. Thus, functional roles of PrP(C) in primary microglial cells are - if present - much more subtle than in transformed microglial cell lines.
Collapse
|
26
|
Wiltshire SA, Marton J, Leiva-Torres GA, Vidal SM. Mapping of a quantitative trait locus controlling susceptibility to Coxsackievirus B3-induced viral hepatitis. Genes Immun 2015; 16:261-7. [PMID: 25790079 DOI: 10.1038/gene.2015.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 12/23/2014] [Accepted: 01/05/2015] [Indexed: 11/09/2022]
Abstract
The pathogenesis of coxsackieviral infection is a multifactorial process involving host genetics, viral genetics and the environment in which they interact. We have used a mouse model of Coxsackievirus B3 infection to characterize the contribution of host genetics to infection survival and to viral hepatitis. Twenty-five AcB/BcA recombinant congenic mouse strains were screened. One, BcA86, was found to be particularly susceptible to early mortality; 100% of BcA86 mice died by day 6 compared with 0% of B6 mice (P=0.0012). This increased mortality was accompanied by an increased hepatic necrosis as measured by serum alanine aminotransferase (ALT) levels (19547±10556 vs 769±109, P=0.0055). This occurred despite a predominantly resistant (C57BL/6) genetic background. Linkage analysis in a cohort (n=210) of (BcA86x C56Bl/10)F2 animals revealed a new locus on chromosome 13 (peak linkage 101.2 Mbp, lod 4.50 and P=0.003). This locus controlled serum ALT levels as early as 48 h following the infection, and led to an elevated expression of type I interferon. Another locus on chromosome 17 (peak linkage 57.2 Mbp) was significantly linked to heart viral titer (lod 3.4 and P=0.046). These results provide new evidence for the presence of genetic loci contributing to the susceptibility of mice to viral hepatitis.
Collapse
Affiliation(s)
- S A Wiltshire
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - J Marton
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - G A Leiva-Torres
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - S M Vidal
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| |
Collapse
|
27
|
Reduced MCMV Δm157 viral clearance in the absence of TSAd. Sci Rep 2015; 5:9219. [PMID: 25783199 PMCID: PMC4363830 DOI: 10.1038/srep09219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/19/2015] [Indexed: 12/14/2022] Open
Abstract
The T cell specific adapter protein (TSAd) is expressed in activated T cells and NK cells. While TSAd is beginning to emerge as a critical regulator of Lck and Itk activity in T cells, its role in NK cells has not yet been explored. Here we have examined susceptibility to virus infections in a murine model using various viral infection models. We report that TSAd-deficient mice display reduced clearance of murine cytomegalovirus (MCMV) that lack the viral MHC class I homologue m157, which is critical for Ly49H-mediated NK cell recognition of infected cells. In this infection model, NK cells contribute in the early stages of the disease, whereas CD8+ T cells are critical for viral clearance. We found that mice infected with MCMV Δm157 displayed reduced viral clearance in the spleen as well as reduced proliferation in spleen NK cells and CD8+ T cells in the absence of TSAd. Though no other immunophenotype was detected in the infection models tested, these data suggests that in the absence of the Ly49H ligand activation, NK cell and CD8+ T cell responses may be compromised in TSAd-deficient mice.
Collapse
|
28
|
Milano A, Lodder EM, Bezzina CR. TNNI3K in cardiovascular disease and prospects for therapy. J Mol Cell Cardiol 2015; 82:167-73. [PMID: 25787061 DOI: 10.1016/j.yjmcc.2015.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 02/23/2015] [Accepted: 03/09/2015] [Indexed: 12/26/2022]
Abstract
Cardiovascular diseases are an important cause of morbidity and mortality worldwide and the global burden of these diseases continues to grow. Therefore new therapies are urgently needed. The role of protein kinases in disease, including cardiac disease, is long recognized, making kinases important therapeutic targets. We here review the knowledge gathered in the last decade about troponin I-interacting kinase (TNNI3K), a kinase with cardiac-restricted expression that has been implicated in various cardiac phenotypes and diseases including heart failure, cardiomyopathy, ischemia/reperfusion injury and conduction of the cardiac electrical impulse.
Collapse
Affiliation(s)
- Annalisa Milano
- Department of Clinical and Experimental Cardiology, Academic Medical Centre, Amsterdam, The Netherlands
| | - Elisabeth M Lodder
- Department of Clinical and Experimental Cardiology, Academic Medical Centre, Amsterdam, The Netherlands
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Academic Medical Centre, Amsterdam, The Netherlands.
| |
Collapse
|
29
|
Lafferty EI, Wiltshire SA, Angers I, Vidal SM, Qureshi ST. Unc93b1 -Dependent Endosomal Toll-Like Receptor Signaling Regulates Inflammation and Mortality during Coxsackievirus B3 Infection. J Innate Immun 2015; 7:315-30. [PMID: 25675947 DOI: 10.1159/000369342] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 10/26/2014] [Indexed: 12/24/2022] Open
Abstract
Coxsackievirus strain B serotype 3 (CVB3)-induced myocarditis is an important human disease that causes permanent tissue damage and can lead to death from acute infection or long-term morbidity caused by chronic inflammation. The timing and magnitude of immune activation following CVB3 infection can mediate a positive host outcome or increase tissue pathology. To better elucidate the role of endosomal Toll-like receptor (TLR) signaling in acute CVB3 infection, we studied mice with a loss-of-function mutation, known as Letr for 'loss of endosomal TLR response', in Unc93b1, which is a chaperone protein for TLR3, TLR7 and TLR9. Using Unc93b1(Letr/)(Letr) mice, we determined that Unc93b1-dependent TLR activation was essential for the survival of acute CVB3-induced myocarditis. We also determined that a lack of endosomal TLR signaling was associated with a higher viral load in target organs and that it increased inflammation, necrosis and fibrosis in cardiac tissue. Loss of Unc93b1 function was also associated with increased cardiac expression of Ifn-b and markers of tissue injury and fibrosis including Lcn2 and Serpina3n early after CVB3 infection. These observations establish a significant role for Unc93b1 in the regulation of the host inflammatory response to CVB3 infection and also reveal potential mediators of host tissue damage that merit further investigation in acute viral myocarditis.
Collapse
Affiliation(s)
- Erin I Lafferty
- Meakins-Christie Laboratories, McGill University, Montréal, Qué., Canada
| | | | | | | | | |
Collapse
|
30
|
Mouse ENU Mutagenesis to Understand Immunity to Infection: Methods, Selected Examples, and Perspectives. Genes (Basel) 2014; 5:887-925. [PMID: 25268389 PMCID: PMC4276919 DOI: 10.3390/genes5040887] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/19/2014] [Accepted: 08/21/2014] [Indexed: 12/30/2022] Open
Abstract
Infectious diseases are responsible for over 25% of deaths globally, but many more individuals are exposed to deadly pathogens. The outcome of infection results from a set of diverse factors including pathogen virulence factors, the environment, and the genetic make-up of the host. The completion of the human reference genome sequence in 2004 along with technological advances have tremendously accelerated and renovated the tools to study the genetic etiology of infectious diseases in humans and its best characterized mammalian model, the mouse. Advancements in mouse genomic resources have accelerated genome-wide functional approaches, such as gene-driven and phenotype-driven mutagenesis, bringing to the fore the use of mouse models that reproduce accurately many aspects of the pathogenesis of human infectious diseases. Treatment with the mutagen N-ethyl-N-nitrosourea (ENU) has become the most popular phenotype-driven approach. Our team and others have employed mouse ENU mutagenesis to identify host genes that directly impact susceptibility to pathogens of global significance. In this review, we first describe the strategies and tools used in mouse genetics to understand immunity to infection with special emphasis on chemical mutagenesis of the mouse germ-line together with current strategies to efficiently identify functional mutations using next generation sequencing. Then, we highlight illustrative examples of genes, proteins, and cellular signatures that have been revealed by ENU screens and have been shown to be involved in susceptibility or resistance to infectious diseases caused by parasites, bacteria, and viruses.
Collapse
|
31
|
Abraham DM, Marchuk DA. Inhibition of the cardiomyocyte-specific troponin I-interacting kinase limits oxidative stress, injury, and adverse remodeling due to ischemic heart disease. Circ Res 2014; 114:938-40. [PMID: 24625723 DOI: 10.1161/circresaha.113.303238] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ischemia–reperfusion injury is strongly associated with increased oxidative stress, mitochondrial dysfunction, and cell death. These processes are diminished in an animal model of ischemia–reperfusion by the genetic loss or pharmacological inhibition of troponin I–interacting kinase.
Collapse
Affiliation(s)
- Dennis M Abraham
- From the Department of Medicine, Division of Cardiology (D.M.A.) and Department of Molecular Genetics and Microbiology (D.A.M.), Duke University School of Medicine, Durham, NC
| | | |
Collapse
|
32
|
Lal H, Ahmad F, Parikh S, Force T. Troponin I-interacting protein kinase: a novel cardiac-specific kinase, emerging as a molecular target for the treatment of cardiac disease. Circ J 2014; 78:1514-1519. [PMID: 24899531 PMCID: PMC4151348 DOI: 10.1253/circj.cj-14-0543] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Coronary artery disease is the leading cause of death and disability worldwide. In patients with acute coronary syndromes, timely and effective myocardial reperfusion by percutaneous coronary intervention is the primary treatment of choice to minimize the ischemic injury and limit the size of the myocardial infarction (MI). However, reperfusion can itself promote cardiomyocyte death, which leads to cardiac dysfunction via reperfusion injury. The molecular mechanisms of ischemia-reperfusion (IR) injury are not completely understood and new drug targets are needed. Recently, we reported that cardiac troponin I-interacting protein kinase (TNNI3K), a cardiomyocyte-specific kinase, promotes IR injury via profound oxidative stress, thereby promoting cardiomyocyte death. By using novel genetic animal models and newly developed small-molecule TNNI3K inhibitors, we demonstrated that TNNI3K-mediated IR injury occurs through impaired mitochondrial function and is in part dependent on p38 MAPK. Here we discuss the emerging role of TNNI3K as a promising new drug target to limit IR-induced myocardial injury. We will also examine the underlying mechanisms that drive the profoundly reduced infarct size in mice in whichTNNI3Kis specifically deleted in cardiomyocytes. Because TNNI3K is a cardiac-specific kinase, it could be an ideal molecular target, as inhibiting it would have little or no effect on other organ systems, a serious problem associated with the use of kinase inhibitors targeting kinases that are more widely expressed.
Collapse
Affiliation(s)
- Hind Lal
- Center for Translational Medicine and Cardiology Division, Temple University School of Medicine, 3500 N. Broad St., MERB 943, Philadelphia, PA19140
| | - Firdos Ahmad
- Center for Translational Medicine and Cardiology Division, Temple University School of Medicine, 3500 N. Broad St., MERB 943, Philadelphia, PA19140
| | - Shan Parikh
- Center for Translational Medicine and Cardiology Division, Temple University School of Medicine, 3500 N. Broad St., MERB 943, Philadelphia, PA19140
| | - Thomas Force
- Center for Translational Medicine and Cardiology Division, Temple University School of Medicine, 3500 N. Broad St., MERB 943, Philadelphia, PA19140
| |
Collapse
|
33
|
Song Y, Ge W, Cai H, Zhang H. Curcumin Protects Mice From Coxsackievirus B3-Induced Myocarditis by Inhibiting the Phosphatidylinositol 3 kinase/Akt/Nuclear Factor-κB Pathway. J Cardiovasc Pharmacol Ther 2013; 18:560-9. [PMID: 24057864 DOI: 10.1177/1074248413503044] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Viral myocarditis is an inflammation of the myocardium, and coxsackievirus B3 (CVB3) is one of the most important etiologic agents. Curcumin is an active ingredient of Curcumin longa, which has been used as a traditional Chinese herb for the treatment of various inflammatory diseases. The aim of this study was to explore the therapeutic effect of curcumin on CVB3-induced myocarditis and the underlying mechanism. Our results showed that treatment with curcumin could significantly attenuate CVB3-induced myocarditis, as demonstrated by improved weight loss, increased survival rate, reduced serological level cardiac enzymes, and improved heart histopathology. Of importance, curcumin administration was revealed to significantly reduce the systemic and local myocardial expression of proinflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL) 6, and IL-1β, in the CVB3-infected mice. Further study showed that curcumin treatment significantly inhibited the CVB3-induced activation of nuclear factor-κB (NF-κB), a key transcription factor in the pathogenesis of inflammation, in a phosphatidylinositol 3 kinase (PI3K)/Akt pathway-dependent manner. These data indicate that curcumin has protective effect against CVB3-induced myocarditis by inhibiting PI3K/Akt/NF-κB signaling pathway and thus reducing the inflammatory response.
Collapse
Affiliation(s)
- Yunxiao Song
- Department of Clinical Laboratory, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Wen Ge
- Department of Clinical Laboratory, Shanghai Xuhui Central Hospital, Shanghai, China
| | - HaiBing Cai
- Department of Clinical Laboratory, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Haichen Zhang
- Department of Clinical Laboratory, Shanghai Xuhui Central Hospital, Shanghai, China
| |
Collapse
|
34
|
Trifonov VA, Dementyeva PV, Larkin DM, O'Brien PCM, Perelman PL, Yang F, Ferguson-Smith MA, Graphodatsky AS. Transcription of a protein-coding gene on B chromosomes of the Siberian roe deer (Capreolus pygargus). BMC Biol 2013; 11:90. [PMID: 23915065 PMCID: PMC3751663 DOI: 10.1186/1741-7007-11-90] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/16/2013] [Indexed: 11/25/2022] Open
Abstract
Background Most eukaryotic species represent stable karyotypes with a particular diploid number. B chromosomes are additional to standard karyotypes and may vary in size, number and morphology even between cells of the same individual. For many years it was generally believed that B chromosomes found in some plant, animal and fungi species lacked active genes. Recently, molecular cytogenetic studies showed the presence of additional copies of protein-coding genes on B chromosomes. However, the transcriptional activity of these genes remained elusive. We studied karyotypes of the Siberian roe deer (Capreolus pygargus) that possess up to 14 B chromosomes to investigate the presence and expression of genes on supernumerary chromosomes. Results Here, we describe a 2 Mbp region homologous to cattle chromosome 3 and containing TNNI3K (partial), FPGT, LRRIQ3 and a large gene-sparse segment on B chromosomes of the Siberian roe deer. The presence of the copy of the autosomal region was demonstrated by B-specific cDNA analysis, PCR assisted mapping, cattle bacterial artificial chromosome (BAC) clone localization and quantitative polymerase chain reaction (qPCR). By comparative analysis of B-specific and non-B chromosomal sequences we discovered some B chromosome-specific mutations in protein-coding genes, which further enabled the detection of a FPGT-TNNI3K transcript expressed from duplicated genes located on B chromosomes in roe deer fibroblasts. Conclusions Discovery of a large autosomal segment in all B chromosomes of the Siberian roe deer further corroborates the view of an autosomal origin for these elements. Detection of a B-derived transcript in fibroblasts implies that the protein coding sequences located on Bs are not fully inactivated. The origin, evolution and effect on host of B chromosomal genes seem to be similar to autosomal segmental duplications, which reinforces the view that supernumerary chromosomal elements might play an important role in genome evolution.
Collapse
|
35
|
Chevenon M, Naccache M, Eva MM, Khan RT, Malo D. Functional validation of the genetic architecture of Salmonella Enteritidis persistence in 129S6 mice. Mamm Genome 2013; 24:218-27. [PMID: 23588612 DOI: 10.1007/s00335-013-9453-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/18/2013] [Indexed: 11/26/2022]
Abstract
The Gram-negative bacteria, Salmonella, cause a broad spectrum of clinical diseases in humans, ranging from asymptomatic carriage to life-threatening sepsis. We have designed an experimental model to study the contribution of genetic factors to the persistence of Salmonella Enteritidis during the late phase of infection in 129S6/SvEvTac and C57BL/6J mice. C57BL/6J mice cleared the bacteria from their reticuloendothelial system within a period of 42 days, whereas the 129S6 mice still presented a high bacterial load. Using this model, we have identified ten Salmonella Enteritidis susceptibility loci (Ses1, Ses1.1, and Ses3-Ses10) associated with bacterial persistence in target organs of 129S6/SvEvTac mice using a two-locus epistasis QTL linkage mapping approach. Significant statistical interactions were detected between Ses1 on chromosome 1 and Ses5 on chromosome 7 and between Ses1 and Ses4 on chromosome X. In this study, we functionally validated the genetic architecture of Salmonella persistence in 129S6 mice using single- (129S6.B6-Ses1.2 that combines Ses1 and Ses1.1 loci, 129S6.B6-Ses4, and 129S6.B6-Ses5) and double-congenic mice (129S6.B6-Ses1.2/Ses4 and 129S6.B6-Ses1.2/Ses5). These experiments demonstrate functional interactions between Ses1.2 and Ses4 or Ses5 that improve Salmonella Enteritidis clearance, validating the critical role played by gene-gene interactions in the contribution to bacterial clearance heritability. Improved bacterial clearance in double-congenic mice could be explained by the impact of Ses4 and Ses5 in combination with Ses1.2 on TH polarization since a TH2 bias (decreased Ifng and increased Il4 mRNA levels and reduced IgG2a immunoglobulins in the serum) was observed in 129S6.B6-Ses1.2/Ses5 mice and a TH17 (high Il17 expression) bias in 129S6.B6-Ses1.2/Ses4.
Collapse
Affiliation(s)
- Marie Chevenon
- Department of Medicine, McGill University, Montreal, QC, Canada
| | | | | | | | | |
Collapse
|
36
|
Wang X, Wang J, Su M, Wang C, Chen J, Wang H, Song L, Zou Y, Zhang L, Zhang Y, Hui R. TNNI3K, a cardiac-specific kinase, promotes physiological cardiac hypertrophy in transgenic mice. PLoS One 2013; 8:e58570. [PMID: 23472207 PMCID: PMC3589374 DOI: 10.1371/journal.pone.0058570] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 02/06/2013] [Indexed: 01/09/2023] Open
Abstract
Purpose Protein kinase plays an essential role in controlling cardiac growth and hypertrophic remodeling. The cardiac troponin I-interacting kinase (TNNI3K), a novel cardiac specific kinase, is associated with cardiomyocyte hypertrophy. However, the precise function of TNNI3K in regulating cardiac remodeling has remained controversial. Methods and Results In a rat model of cardiac hypertrophy generated by transverse aortic constriction, myocardial TNNI3K expression was significantly increased by 1.62 folds (P<0.05) after constriction for 15 days. To investigate the role of TNNI3K in cardiac hypertrophy, we generated transgenic mouse lines with overexpression of human TNNI3K specifically in the heart. At the age of 3 months, the high-copy-number TNNI3K transgenic mice demonstrated a phenotype of concentric hypertrophy with increased heart weight normalized to body weight (1.31 fold, P<0.01). Echocardiography and non-invasive hemodynamic assessments showed enhanced cardiac function. No necrosis or myocyte disarray was observed in the heart of TNNI3K transgenic mice. This concentric hypertrophy maintained up to 12 months of age without cardiac dysfunction. The phospho amino acid analysis revealed that TNNI3K is a protein-tyrosine kinase. The yeast two-hybrid screen and co-immunoprecipitation assay identified cTnI as a target for TNNI3K. Moreover, TNNI3K overexpression induced cTnI phosphorylation at Ser22/Ser23 in vivo and in vitro, suggesting that TNNI3K is a novel upstream regulator for cTnI phosphorylation. Conclusion TNNI3K promotes a concentric hypertrophy with enhancement of cardiac function via regulating the phosphorylation of cTnI. TNNI3K could be a potential therapeutic target for preventing from heart failure.
Collapse
Affiliation(s)
- Xiaojian Wang
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Jizheng Wang
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Ming Su
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Changxin Wang
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Jingzhou Chen
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Hu Wang
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Lei Song
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Yubao Zou
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People's Republic of China
| | - Youyi Zhang
- Institute of Vascular Medicine, Peking University Third Hospital, Beijing, People's Republic of China
| | - Rutai Hui
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, FuWai Hospital & Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
37
|
Tang H, Xiao K, Mao L, Rockman HA, Marchuk DA. Overexpression of TNNI3K, a cardiac-specific MAPKKK, promotes cardiac dysfunction. J Mol Cell Cardiol 2013; 54:101-11. [PMID: 23085512 PMCID: PMC3535516 DOI: 10.1016/j.yjmcc.2012.10.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 10/01/2012] [Accepted: 10/04/2012] [Indexed: 12/21/2022]
Abstract
Cardiac troponin I-interacting kinase (TNNI3K) is a cardiac-specific kinase whose biological function remains largely unknown. We have recently shown that TNNI3K expression greatly accelerates cardiac dysfunction in mouse models of cardiomyopathy, indicating an important role in modulating disease progression. To further investigate TNNI3K kinase activity in vivo, we have generated transgenic mice expressing both wild-type and kinase-dead versions of the human TNNI3K protein. Importantly, we show that the increased TNNI3K kinase activity induces mouse cardiac remodeling, and its kinase activity promotes accelerated disease progression in a left-ventricular pressure overload model of mouse cardiomyopathy. Using an in vitro kinase assay and proteomics analysis, we show that TNNI3K is a dual-function kinase with Tyr and Ser/Thr kinase activity. TNNI3K expression induces a series of cellular and molecular changes, including a reduction of sarcomere length and changes in titin isoform composition, which are indicative of cardiac remodeling. Using antisera to TNNI3K, we show that TNNI3K protein is located at the sarcomere Z disc. These combined data suggest that TNNI3K mediates cell signaling to modulate cardiac response to stress.
Collapse
Affiliation(s)
- Hao Tang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710
| | - Kunhong Xiao
- Department of Medicine, Duke University, Durham, NC 27710
| | - Lan Mao
- Department of Medicine, Duke University, Durham, NC 27710
| | | | - Douglas A. Marchuk
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710
| |
Collapse
|
38
|
Nadeau JH, Forejt J, Takada T, Shiroishi T. Chromosome substitution strains: gene discovery, functional analysis, and systems studies. Mamm Genome 2012; 23:693-705. [PMID: 22961226 DOI: 10.1007/s00335-012-9426-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2012] [Accepted: 08/02/2012] [Indexed: 12/31/2022]
Abstract
Laboratory mice are valuable in biomedical research in part because of the extraordinary diversity of genetic resources that are available for studies of complex genetic traits and as models for human biology and disease. Chromosome substitution strains (CSSs) are important in this resource portfolio because of their demonstrated use for gene discovery, genetic and epigenetic studies, functional characterizations, and systems analysis. CSSs are made by replacing a single chromosome in a host strain with the corresponding chromosome from a donor strain. A complete CSS panel involves a total of 22 engineered inbred strains, one for each of the 19 autosomes, one each for the X and Y chromosomes, and one for mitochondria. A genome survey simply involves comparing each phenotype for each of the CSSs with the phenotypes of the host strain. The CSS panels that are available for laboratory mice have been used to dissect a remarkable variety of phenotypes and to characterize an impressive array of disease models. These surveys have revealed considerable phenotypic diversity even among closely related progenitor strains, evidence for strong epistasis and for heritable epigenetic changes. Perhaps most importantly, and presumably because of their unique genetic constitution, CSSs, and congenic strains derived from them, the genetic variants underlying quantitative trait loci (QTLs) are readily identified and functionally characterized. Together these studies show that CSSs are important resource for laboratory mice.
Collapse
Affiliation(s)
- Joseph H Nadeau
- Pacific Northwest Research Institute, 720 Broadway, Seattle, WA 98122, USA.
| | | | | | | |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW To present recent findings on the pathogenesis of coxsackievirus B3 (CVB3) myocarditis based on animal models, with a focus on the role of T helper (Th) immune responses in disease progression. RECENT FINDINGS Acute CVB3 myocarditis is known to be increased by Th1 immune responses, but recent findings indicate that Th1-type immunity protects against acute myocarditis by reducing viral replication and prevents the progression to chronic myocarditis and dilated cardiomyopathy (DCM) by inhibiting Th2 responses. Th2 responses reduce acute myocarditis by inhibiting Th1 responses via regulatory T cells and anti-inflammatory cytokines, but can be deleterious when they induce acute cardiac remodeling leading to chronic myocarditis/DCM. Th2-skewed immune responses allow resistant strains of mice to progress from myocarditis to DCM. In contrast, Th17 responses are elevated during acute and chronic myocarditis and have been found to contribute to cardiac remodeling and DCM. SUMMARY Recent data indicate that elevated Th2 and Th17 responses during acute CVB3 myocarditis are critical for the progression from myocarditis to DCM and heart failure because of their ability to induce cardiac remodeling. Th1 responses protect against CVB3 myocarditis by inhibiting Th2 responses and viral replication, but increase acute inflammation.
Collapse
Affiliation(s)
- DeLisa Fairweather
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
| | | | | |
Collapse
|
40
|
New findings and the role of cardiac imaging in myocarditis and related induced cardiomyopathy. J Cardiovasc Echogr 2012. [DOI: 10.1016/j.jcecho.2012.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
41
|
Lai ZF, Chen YZ. Evidence, hypotheses and significance of MAP kinase TNNI3K interacting with its partners. World J Hypertens 2012; 2:22-28. [DOI: 10.5494/wjh.v2.i2.22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
TNNI3K is a cardiac-specific and cardiac troponin I (cTnI)-interacting MAP kinase, known to play important roles in promoting cardiac differentiation, maintenance of beating rhythm and contractual force. The molecular structure of TNNI3K contains three kinds of domain: a seven or ten NH2-terminal ankyrin repeat domain followed by a protein kinase domain and a COOH-terminal serine-rich domain. There are many binding sites in the structure of TNNI3K for binding to ATP, magnesium, nucleotide, protein kinase C, antioxidant protein 1 (AOP-1) and cTnI, indicating TNNI3K has many interacting partners. This review summarizes the evidence, hypothesis and significance of TNNI3K interacting with TNNI3 and its other putative interaction partners. From the literature, the interaction partners of TNNI3K are divided into 2 types following their phenotypic pattern of functions, positive interaction (to increase the cardiac performance) or negative interaction (to suppress the cardiac performance). Following their binding sites, it also can be divided into other 2 types: binding to C-terminal domain (e.g., cTnI) or binding to both ankyrin repeat domain and C-terminal domains (AOP-1). To date, a well understood partner of TNNI3K is cTnI, from the molecular structure, physiological function, mechanisms and its significance in some physiological and pathophysiological conditions. There are many reasons to believe that, with more understanding on the TNNI3K interacting with its partners, we can understand more roles of TNNI3K in some cardiac diseases.
Collapse
|
42
|
Abstract
Modifier genes are an integral part of the genetic landscape in both humans and experimental organisms, but have been less well explored in mammals than other systems. A growing number of modifier genes in mouse models of disease nonetheless illustrate the potential for novel findings, while new technical advances promise many more to come. Modifier genes in mouse models include induced mutations and spontaneous or wild-derived variations captured in inbred strains. Identification of modifiers among wild-derived variants in particular should detect disease modifiers that have been shaped by selection and might therefore be compatible with high fitness and function. Here we review selected examples and argue that modifier genes derived from natural variation may provide a bias for nodes in genetic networks that have greater intrinsic plasticity and whose therapeutic manipulation may therefore be more resilient to side effects than conventional targets.
Collapse
Affiliation(s)
- Bruce A Hamilton
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America.
| | | |
Collapse
|
43
|
Elliott PM, Mohiddin SA. Almanac 2011: Cardiomyopathies. The national society journals present selected research that has driven recent advances in clinical cardiology. Egypt Heart J 2012. [DOI: 10.1016/j.ehj.2012.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
44
|
Elliott PM, Mohiddin SA. Almanac 2011: Cardiomyopathies. The national society journals present selected research that has driven recent advances in clinical cardiology. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2012. [DOI: 10.1016/j.repce.2012.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
45
|
Almanac 2011: Cardiomyopathies. The national society journals present selected research that has driven recent advances in clinical cardiology. Rev Port Cardiol 2012; 31:255-61. [DOI: 10.1016/j.repc.2012.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 01/04/2012] [Indexed: 11/23/2022] Open
|