1
|
Ruiz-Orera J, Miller DC, Greiner J, Genehr C, Grammatikaki A, Blachut S, Mbebi J, Patone G, Myronova A, Adami E, Dewani N, Liang N, Hummel O, Muecke MB, Hildebrandt TB, Fritsch G, Schrade L, Zimmermann WH, Kondova I, Diecke S, van Heesch S, Hübner N. Evolution of translational control and the emergence of genes and open reading frames in human and non-human primate hearts. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1217-1235. [PMID: 39317836 PMCID: PMC11473369 DOI: 10.1038/s44161-024-00544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 08/28/2024] [Indexed: 09/26/2024]
Abstract
Evolutionary innovations can be driven by changes in the rates of RNA translation and the emergence of new genes and small open reading frames (sORFs). In this study, we characterized the transcriptional and translational landscape of the hearts of four primate and two rodent species through integrative ribosome and transcriptomic profiling, including adult left ventricle tissues and induced pluripotent stem cell-derived cardiomyocyte cell cultures. We show here that the translational efficiencies of subunits of the mitochondrial oxidative phosphorylation chain complexes IV and V evolved rapidly across mammalian evolution. Moreover, we discovered hundreds of species-specific and lineage-specific genomic innovations that emerged during primate evolution in the heart, including 551 genes, 504 sORFs and 76 evolutionarily conserved genes displaying human-specific cardiac-enriched expression. Overall, our work describes the evolutionary processes and mechanisms that have shaped cardiac transcription and translation in recent primate evolution and sheds light on how these can contribute to cardiac development and disease.
Collapse
Affiliation(s)
- Jorge Ruiz-Orera
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
| | - Duncan C Miller
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Technology Platform Pluripotent Stem Cells, Berlin, Germany
| | - Johannes Greiner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Carolin Genehr
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Technology Platform Pluripotent Stem Cells, Berlin, Germany
| | - Aliki Grammatikaki
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Susanne Blachut
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Jeanne Mbebi
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Giannino Patone
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anna Myronova
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Eleonora Adami
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Nikita Dewani
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ning Liang
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Oliver Hummel
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Michael B Muecke
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Thomas B Hildebrandt
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
- Freie Universitaet Berlin, Berlin, Germany
| | - Guido Fritsch
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Lisa Schrade
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Wolfram H Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Lower Saxony, Göttingen, Germany
- DZNE (German Center for Neurodegenerative Diseases), Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Göttingen, Germany
| | - Ivanela Kondova
- Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Sebastian Diecke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Technology Platform Pluripotent Stem Cells, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Sebastiaan van Heesch
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
- Charité-Universitätsmedizin, Berlin, Germany.
- Helmholtz Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
2
|
Read DF, Booth GT, Daza RM, Jackson DL, Gladden RG, Srivatsan SR, Ewing B, Franks JM, Spurrell CH, Gomes AR, O'Day D, Gogate AA, Martin BK, Larson H, Pfleger C, Starita L, Lin Y, Shendure J, Lin S, Trapnell C. Single-cell analysis of chromatin and expression reveals age- and sex-associated alterations in the human heart. Commun Biol 2024; 7:1052. [PMID: 39187646 PMCID: PMC11347658 DOI: 10.1038/s42003-024-06582-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/11/2024] [Indexed: 08/28/2024] Open
Abstract
Sex differences and age-related changes in the human heart at the tissue, cell, and molecular level have been well-documented and many may be relevant for cardiovascular disease. However, how molecular programs within individual cell types vary across individuals by age and sex remains poorly characterized. To better understand this variation, we performed single-nucleus combinatorial indexing (sci) ATAC- and RNA-Seq in human heart samples from nine donors. We identify hundreds of differentially expressed genes by age and sex and find epigenetic signatures of variation in ATAC-Seq data in this discovery cohort. We then scale up our single-cell RNA-Seq analysis by combining our data with five recently published single nucleus RNA-Seq datasets of healthy adult hearts. We find variation such as metabolic alterations by sex and immune changes by age in differential expression tests, as well as alterations in abundance of cardiomyocytes by sex and neurons with age. In addition, we compare our adult-derived ATAC-Seq profiles to analogous fetal cell types to identify putative developmental-stage-specific regulatory factors. Finally, we train predictive models of cell-type-specific RNA expression levels utilizing ATAC-Seq profiles to link distal regulatory sequences to promoters, quantifying the predictive value of a simple TF-to-expression regulatory grammar and identifying cell-type-specific TFs. Our analysis represents the largest single-cell analysis of cardiac variation by age and sex to date and provides a resource for further study of healthy cardiac variation and transcriptional regulation at single-cell resolution.
Collapse
Affiliation(s)
- David F Read
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Gregory T Booth
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Riza M Daza
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Dana L Jackson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Rula Green Gladden
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sanjay R Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Brent Ewing
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Jennifer M Franks
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | | | - Diana O'Day
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Aishwarya A Gogate
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Beth K Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Haleigh Larson
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Christian Pfleger
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA, USA
| | - Lea Starita
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Yiing Lin
- Department of Surgery, Washington University, St Louis, MO, USA
| | - Jay Shendure
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Seattle Children's Research Institute, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| | - Shin Lin
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA, USA.
| | - Cole Trapnell
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
| |
Collapse
|
3
|
Zhang B, Zhu Y, Zhang Z, Wu F, Ma X, Sheng W, Dai R, Guo Z, Yan W, Hao L, Huang G, Ma D, Hao B, Ma J. SMC3 contributes to heart development by regulating super-enhancer associated genes. Exp Mol Med 2024; 56:1826-1842. [PMID: 39085358 PMCID: PMC11372143 DOI: 10.1038/s12276-024-01293-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/23/2024] [Accepted: 05/21/2024] [Indexed: 08/02/2024] Open
Abstract
Abnormal cardiac development has been observed in individuals with Cornelia de Lange syndrome (CdLS) due to mutations in genes encoding members of the cohesin complex. However, the precise role of cohesin in heart development remains elusive. In this study, we aimed to elucidate the indispensable role of SMC3, a component of the cohesin complex, in cardiac development and its underlying mechanism. Our investigation revealed that CdLS patients with SMC3 mutations have high rates of congenital heart disease (CHD). We utilized heart-specific Smc3-knockout (SMC3-cKO) mice, which exhibit varying degrees of outflow tract (OFT) abnormalities, to further explore this relationship. Additionally, we identified 16 rare SMC3 variants with potential pathogenicity in individuals with isolated CHD. By employing single-nucleus RNA sequencing and chromosome conformation capture high-throughput genome-wide translocation sequencing, we revealed that Smc3 deletion downregulates the expression of key genes, including Ets2, in OFT cardiac muscle cells by specifically decreasing interactions between super-enhancers (SEs) and promoters. Notably, Ets2-SE-null mice also exhibit delayed OFT development in the heart. Our research revealed a novel role for SMC3 in heart development via the regulation of SE-associated genes, suggesting its potential relevance as a CHD-related gene and providing crucial insights into the molecular basis of cardiac development.
Collapse
Affiliation(s)
- Bowen Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Yongchang Zhu
- Henan Medical Genetics Institute, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China
| | - Zhen Zhang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Feizhen Wu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Xiaojing Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Wei Sheng
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Ranran Dai
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Zhenglong Guo
- Henan Medical Genetics Institute, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China
| | - Weili Yan
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Lili Hao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Guoying Huang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China.
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China.
| | - Bingtao Hao
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- Henan Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450000, China.
| | - Jing Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences; ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital; Institute of Medical Genetics & Genomics; Key Laboratory of Birth Defects, Children's Hospital; Medical Science Data Center at Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
4
|
Palomer X, Salvador JM, Griñán-Ferré C, Barroso E, Pallàs M, Vázquez-Carrera M. GADD45A: With or without you. Med Res Rev 2024; 44:1375-1403. [PMID: 38264852 DOI: 10.1002/med.22015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/11/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024]
Abstract
The growth arrest and DNA damage inducible (GADD)45 family includes three small and ubiquitously distributed proteins (GADD45A, GADD45B, and GADD45G) that regulate numerous cellular processes associated with stress signaling and injury response. Here, we provide a comprehensive review of the current literature investigating GADD45A, the first discovered member of the family. We first depict how its levels are regulated by a myriad of genotoxic and non-genotoxic stressors, and through the combined action of intricate transcriptional, posttranscriptional, and even, posttranslational mechanisms. GADD45A is a recognized tumor suppressor and, for this reason, we next summarize its role in cancer, as well as the different mechanisms by which it regulates cell cycle, DNA repair, and apoptosis. Beyond these most well-known actions, GADD45A may also influence catabolic and anabolic pathways in the liver, adipose tissue and skeletal muscle, among others. Not surprisingly, GADD45A may trigger AMP-activated protein kinase activity, a master regulator of metabolism, and is known to act as a transcriptional coregulator of numerous nuclear receptors. GADD45A has also been reported to display a cytoprotective role by regulating inflammation, fibrosis and oxidative stress in several organs and tissues, and is regarded an important contributor for the development of heart failure. Overall data point to that GADD45A may play an important role in metabolic, neurodegenerative and cardiovascular diseases, and also autoimmune-related disorders. Thus, the potential mechanisms by which dysregulation of GADD45A activity may contribute to the progression of these diseases are also reviewed below.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Jesús M Salvador
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Christian Griñán-Ferré
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona (NeuroUB), Barcelona, Spain
- Spanish Biomedical Research Center in Neurodegenerative Diseases (CIBERNED)-Instituto de Salud Carlos III, Madrid, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Mercè Pallàs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona (NeuroUB), Barcelona, Spain
- Spanish Biomedical Research Center in Neurodegenerative Diseases (CIBERNED)-Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| |
Collapse
|
5
|
Li X, Wang G, Wang X, Li W, Li N, Liu X, Fan W, He S, Han Y, Su G, Cao Q, Yang P, Hou S. OR11H1 Missense Variant Confers the Susceptibility to Vogt-Koyanagi-Harada Disease by Mediating Gadd45g Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306563. [PMID: 38168905 PMCID: PMC10953539 DOI: 10.1002/advs.202306563] [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: 09/12/2023] [Revised: 11/17/2023] [Indexed: 01/05/2024]
Abstract
Vogt-Koyanagi-Harada (VKH) disease is a severe autoimmune disease. Herein, whole-exome sequencing (WES) study are performed on 2,573 controls and 229 VKH patients with follow-up next-generation sequencing (NGS) in a collection of 2,380 controls and 2,278 VKH patients. A rare c.188T>C (p Val63Ala) variant in the olfactory receptor 11H1 (OR11H1) gene is found to be significantly associated with VKH disease (rs71235604, Pcombined = 7.83 × 10-30 , odds ratio = 3.12). Functional study showes that OR11H1-A63 significantly increased inflammatory factors production and exacerbated barrier function damage. Further studies using RNA-sequencing find that OR11H1-A63 markedly increased growth arrest and DNA-damage-inducible gamma (GADD45G) expression. Moreover, OR11H1-A63 activates the MAPK and NF-κB pathways, and accelerates inflammatory cascades. In addition, inhibiting GADD45G alleviates inflammatory factor secretion, likely due to the regulatory effect of GADD45G on the MAPK and NF-κB pathways. Collectively, this study suggests that the OR11H1-A63 missense mutation may increase susceptibility to VKH disease in a GADD45G-dependent manner.
Collapse
Affiliation(s)
- Xingran Li
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
| | - Guoqing Wang
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
| | - Xiaotang Wang
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
| | - Wanqian Li
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
| | - Na Li
- Department of Laboratory MedicineBeijing Tongren Hospital, Capital Medical UniversityBeijing100005China
| | - Xianyang Liu
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
| | - Wei Fan
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
| | - Siyuan He
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
| | - Yue Han
- Beijing Novogene Bioinformatics Technology Co.,LtdBeijing100600China
| | - Guannan Su
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
| | - Qingfeng Cao
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
| | - Peizeng Yang
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
| | - Shengping Hou
- Chongqing Branch of National Clinical Research Center for Ocular Diseases; Chongqing Key Laboratory of Ophthalmology; Chongqing Eye InstituteThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400042China
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijing Tongren HospitalCapital Medical UniversityBeijing100730China
| |
Collapse
|
6
|
Kumagai Y, Kemp MW, Usuda H, Takahashi T, Takahashi Y, Hamada H, Schmidt AF, Hanita T, Watanabe S, Sato S, Ikeda H, Fee EL, Furfaro L, Newnham JP, Jobe AH, Yaegashi N, Saito M. A Reduction in Antenatal Steroid Dose Was Associated with Reduced Cardiac Dysfunction in a Sheep Model of Pregnancy. Reprod Sci 2023; 30:3222-3234. [PMID: 37264260 PMCID: PMC10643432 DOI: 10.1007/s43032-023-01264-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/07/2023] [Indexed: 06/03/2023]
Abstract
Despite widespread use, dosing regimens for antenatal corticosteroid (ACS) therapy are poorly unoptimized. ACS therapy exerts a programming effect on fetal development, which may be associated with an increased risk of cardiovascular disease. Having demonstrated that low-dose steroid therapy is an efficacious means of maturing the preterm lung, we hypothesized that a low-dose steroid exposure would exert fewer adverse functional and transcriptional changes on the fetal heart. We tested this hypothesis using low-dose steroid therapy (10 mg delivered to the ewe over 36 h via constant infusion) and compared cardiac effects with those of a higher dose treatment (30 mg delivered to the ewe over 24 h by intramuscular injection; simulating currently employed clinical ACS regimens). Fetal cardiac function was assessed by ultrasound on the day of ACS treatment initiation. Transcriptomic analyses were performed on fetal myocardial tissue. Relative to saline control, fetuses in the higher-dose clinical treatment group had significantly lower ratios between early diastolic ventricular filling and ventricular filling during atrial systole, and showed the differential expression of myocardial hypertrophy-associated transcripts including βMHC, GADD45γ, and PPARγ. The long-term implications of these changes remain unstudied. Irrespective, optimizing ACS dosing regimens to maximize respiratory benefit while minimizing adverse effects on key organ systems, such as the heart, offers a means of improving the acute and long-term outcomes associated with this important obstetric therapy.
Collapse
Affiliation(s)
- Yusaku Kumagai
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan.
| | - Matthew W Kemp
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
- College of Veterinary and Life Sciences, Murdoch University, Perth, WA, Australia
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Haruo Usuda
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - Tsukasa Takahashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - Yuki Takahashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - Hirotaka Hamada
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | | | - Takushi Hanita
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shimpei Watanabe
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shinichi Sato
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Hideyuki Ikeda
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Erin L Fee
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - Lucy Furfaro
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - John P Newnham
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - Alan H Jobe
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
- Cincinnati Children's Hospital Medical Centre, Cincinnati, OH, USA
| | - Nobuo Yaegashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Masatoshi Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
7
|
Ross JA, Arcos-Villacis N, Battey E, Boogerd C, Orellana CA, Marhuenda E, Swiatlowska P, Hodzic D, Prin F, Mohun T, Catibog N, Tapia O, Gerace L, Iskratsch T, Shah AM, Stroud MJ. Lem2 is essential for cardiac development by maintaining nuclear integrity. Cardiovasc Res 2023; 119:2074-2088. [PMID: 37067297 PMCID: PMC10478753 DOI: 10.1093/cvr/cvad061] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 01/19/2023] [Accepted: 02/08/2023] [Indexed: 04/18/2023] Open
Abstract
AIMS Nuclear envelope integrity is essential for the compartmentalization of the nucleus and cytoplasm. Importantly, mutations in genes encoding nuclear envelope (NE) and associated proteins are the second highest cause of familial dilated cardiomyopathy. One such NE protein that causes cardiomyopathy in humans and affects mouse heart development is Lem2. However, its role in the heart remains poorly understood. METHODS AND RESULTS We generated mice in which Lem2 was specifically ablated either in embryonic cardiomyocytes (Lem2 cKO) or in adult cardiomyocytes (Lem2 iCKO) and carried out detailed physiological, tissue, and cellular analyses. High-resolution episcopic microscopy was used for three-dimensional reconstructions and detailed morphological analyses. RNA-sequencing and immunofluorescence identified altered pathways and cellular phenotypes, and cardiomyocytes were isolated to interrogate nuclear integrity in more detail. In addition, echocardiography provided a physiological assessment of Lem2 iCKO adult mice. We found that Lem2 was essential for cardiac development, and hearts from Lem2 cKO mice were morphologically and transcriptionally underdeveloped. Lem2 cKO hearts displayed high levels of DNA damage, nuclear rupture, and apoptosis. Crucially, we found that these defects were driven by muscle contraction as they were ameliorated by inhibiting myosin contraction and L-type calcium channels. Conversely, reducing Lem2 levels to ∼45% in adult cardiomyocytes did not lead to overt cardiac dysfunction up to 18 months of age. CONCLUSIONS Our data suggest that Lem2 is critical for integrity at the nascent NE in foetal hearts, and protects the nucleus from the mechanical forces of muscle contraction. In contrast, the adult heart is not detectably affected by partial Lem2 depletion, perhaps owing to a more established NE and increased adaptation to mechanical stress. Taken together, these data provide insights into mechanisms underlying cardiomyopathy in patients with mutations in Lem2 and cardio-laminopathies in general.
Collapse
Affiliation(s)
- Jacob A Ross
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Nathaly Arcos-Villacis
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Edmund Battey
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK
| | - Cornelis Boogerd
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht 3584 CT, The Netherlands
| | - Constanza Avalos Orellana
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Emilie Marhuenda
- Division of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
| | - Pamela Swiatlowska
- Division of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
| | - Didier Hodzic
- Department of Developmental Biology, Washington University School of Medicine, 660S. Euclid Avenue, St Louis, MO 63110, USA
| | - Fabrice Prin
- Crick Advanced Light Microscopy Facility, The Francis Crick Institute, London NW1 1AT, UK
| | - Tim Mohun
- Crick Advanced Light Microscopy Facility, The Francis Crick Institute, London NW1 1AT, UK
| | - Norman Catibog
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Olga Tapia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander 39011, Spain
- Department of Basic Medical Sciences, Institute of Biomedical Technologies, University of La Laguna, Tenerife 38200, Spain
| | - Larry Gerace
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
| | - Ajay M Shah
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Matthew J Stroud
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
8
|
Ghosh AK, Kalousdian AA, Shang M, Lux E, Eren M, Keating A, Wilsbacher LD, Vaughan DE. Cardiomyocyte PAI-1 influences the cardiac transcriptome and limits the extent of cardiac fibrosis in response to left ventricular pressure overload. Cell Signal 2023; 104:110555. [PMID: 36584735 DOI: 10.1016/j.cellsig.2022.110555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/26/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is a specific and rapid-acting inhibitor of endogenous plasminogen activators (uPA and tPA). The global PAI-1 knockout mice (PAI-1KO) develop age-dependent cardiac-selective fibrosis, and young global PAI-1KO mice exhibit augmented susceptibility to developing cardiac fibrosis in response to hypertension. Here, we tested the hypothesis that cardiomyocyte PAI-1 is necessary to provide cardioprotective effects in a left ventricular pressure overload-induced murine model of cardiac hypertrophy and fibrosis using cardiomyocyte-specific PAI-1 knockout (cmPAI-1KO) mice. The results revealed that cmPAI-1KO mice display significantly worse cardiac fibrosis than controls. To investigate the molecular mechanisms responsible for these effects, genome-wide cardiac transcriptome analysis was performed. Loss of cardiomyocyte PAI-1 led to differential expression of 978 genes compared to controls in response to left ventricular pressure overload. Pathway enrichment analysis identified the inflammatory response, cell substrate adhesion, regulation of cytokine production, leukocyte migration, extracellular matrix organization, and cytokine-mediated signaling pathways as being significantly upregulated in cmPAI-1KO hearts. Conversely, specific epigenetic repressors, cation transmembrane transport, muscle system processes, and nitric oxide signaling were significantly downregulated in cmPAI-1KO hearts compared to control hearts in response to left ventricular pressure overload. Collectively, the present study provides strong evidence of the impact of cardiomyocyte PAI-1 in regulation of the transcriptome network involved in the cardiac stress response. In response to stress, the deregulatory impact of cardiomyocyte PAI-1 loss on the cardiac transcriptome may be the underlying cause of cardiac-selective accelerated fibrogenesis in global PAI-1-deficient mice.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Anthony A Kalousdian
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Meng Shang
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth Lux
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Mesut Eren
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anna Keating
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lisa D Wilsbacher
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Douglas E Vaughan
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
9
|
Yao D, Shi B, Wang S, Bao L, Tan M, Shen H, Zhang Z, Pan X, Yang Y, Wu Y, Gong K. Isoliquiritigenin Ameliorates Ischemia-Induced Myocardial Injury via Modulating the Nrf2/HO-1 Pathway in Mice. Drug Des Devel Ther 2022; 16:1273-1287. [PMID: 35517984 PMCID: PMC9064455 DOI: 10.2147/dddt.s362754] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/21/2022] [Indexed: 12/30/2022] Open
Abstract
Background Oxidative stress and inflammatory reaction play critical roles in acute myocardial infarction (AMI). Isoliquiritigenin (ISL), a flavonoid monomer extracted from licorice, has been found to have antioxidant and anti-inflammatory effects in cancer studies. Here, we tested the effect and underlying mechanisms of ISL on ischemia-induced myocardial injury in a mouse AMI model. Methods Adult C57BL/6 mice were pre-treated by intraperitoneal injection of ISL and/or a specific nuclear factor E2-related factor 2 (Nrf2) inhibitor ML385 for 3 days, respectively. Then, the AMI model was established by ligating the anterior descending branch of the left coronary artery. Myocardial oxidative stress status, inflammatory response, cardiac function and infarction size were assessed after 7th day of surgery. Results Compared with sham group, the reactive oxygen species (ROS) and malondialdehyde (MDA) level in AMI group were significantly increased. However, the superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) level were dramatically decreased. ISL treatment significantly reduced the myocardial infarction area, improved cardiac function, inhibited the production of ROS and MDA and reduced the consumption of SOD and GSH-Px. Interestingly, ISL could significantly increase nuclear Nrf2 and cytosolic heme oxygenase 1 (HO-1) level in the infarcted myocardium and reduce the oxidative stress after AMI. Also, ISL treatment dramatically inhibited the activation of myocardial NF-κB pathway and reduced the expression of pro-inflammatory factors in the AMI group. However, the administration of ML385 not only suppressed the Nrf2/HO-1 activation, the anti-oxidant and anti-inflammatory effects induced by ISL, but also attenuated the beneficial role of ISL on reducing infarct size and improving cardiac function in the mouse with AMI. Conclusion The results suggested that activation of Nrf2/HO-1 pathway has an essential role in ISL-induced cardiac protection by alleviating myocardial oxidative stress and inflammation response in mice with AMI.
Collapse
Affiliation(s)
- Deshan Yao
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China.,Jiangsu Key Laboratory of Integrative Medicine for the Control of Geriatrics and Institute of Cardiovascular Disease, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Bo Shi
- School of Life Science, Liaoning Normal University, Dalian, 116081, People's Republic of China
| | - Sichuan Wang
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China.,Jiangsu Key Laboratory of Integrative Medicine for the Control of Geriatrics and Institute of Cardiovascular Disease, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Liuxiang Bao
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China.,Jiangsu Key Laboratory of Integrative Medicine for the Control of Geriatrics and Institute of Cardiovascular Disease, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Meng Tan
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China.,Jiangsu Key Laboratory of Integrative Medicine for the Control of Geriatrics and Institute of Cardiovascular Disease, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Hui Shen
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China.,Jiangsu Key Laboratory of Integrative Medicine for the Control of Geriatrics and Institute of Cardiovascular Disease, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Zhengang Zhang
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China.,Jiangsu Key Laboratory of Integrative Medicine for the Control of Geriatrics and Institute of Cardiovascular Disease, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Xin Pan
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China.,Jiangsu Key Laboratory of Integrative Medicine for the Control of Geriatrics and Institute of Cardiovascular Disease, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Yi Yang
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China.,Jiangsu Key Laboratory of Integrative Medicine for the Control of Geriatrics and Institute of Cardiovascular Disease, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Yong Wu
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China.,Jiangsu Key Laboratory of Integrative Medicine for the Control of Geriatrics and Institute of Cardiovascular Disease, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Kaizheng Gong
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, People's Republic of China.,Jiangsu Key Laboratory of Integrative Medicine for the Control of Geriatrics and Institute of Cardiovascular Disease, Yangzhou University, Yangzhou, 225001, People's Republic of China
| |
Collapse
|
10
|
Humayun A, Fornace AJ. GADD45 in Stress Signaling, Cell Cycle Control, and Apoptosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1360:1-22. [PMID: 35505159 DOI: 10.1007/978-3-030-94804-7_1] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
GADD45 is a gene family consisting of GADD45A, GADD45B, and GADD45G that is often induced by DNA damage and other stress signals associated with growth arrest and apoptosis. Many of these roles are carried out via signaling mediated by p38 mitogen-activated protein kinases (MAPKs). The GADD45 proteins can contribute to p38 activation either by activation of upstream kinase(s) or by direct interaction, as well as suppression of p38 activity in certain cases. In vivo, there are important tissue and cell type specific differences in the roles for GADD45 in MAPK signaling. In addition to being p53-regulated, GADD45A has also been found to contribute to p53 activation via p38. Like other stress and signaling proteins, GADD45 proteins show complex regulation and numerous effectors. More recently, aberrant GADD45 expression has been found in several human cancers, but the mechanisms behind these findings largely remain to be understood.
Collapse
Affiliation(s)
- Arslon Humayun
- Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Albert J Fornace
- Lombardi Comprehensive Cancer Center, Washington, DC, USA.
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC, USA.
| |
Collapse
|
11
|
DNMT3B System Dysregulation Contributes to the Hypomethylated State in Ischaemic Human Hearts. Biomedicines 2022; 10:biomedicines10040866. [PMID: 35453616 PMCID: PMC9029641 DOI: 10.3390/biomedicines10040866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 12/10/2022] Open
Abstract
A controversial understanding of the state of the DNA methylation machinery exists in ischaemic cardiomyopathy (ICM). Moreover, its relationship to other epigenetic alterations is incomplete. Therefore, we carried out an in-depth study of the DNA methylation process in human cardiac tissue. We showed a dysregulation of the DNA methylation machinery accordingly with the genome-wide hypomethylation that we observed: specifically, an overexpression of main genes involved in the elimination of methyl groups (TET1, SMUG1), and underexpression of molecules implicated in the maintenance of methylation (MBD2, UHRF1). By contrast, we found DNMT3B upregulation, a key molecule in the addition of methyl residues in DNA, and an underexpression of miR-133a-3p, an inhibitor of DNMT3B transcription. However, we found many relevant alterations that would counteract the upregulation observed, such as the overexpression of TRAF6, responsible for Dnmt3b degradation. Furthermore, we showed that molecules regulating Dnmts activity were altered; specifically, SAM/SAH ratio reduction. All these results are in concordance with the Dnmts normal function that we show. Our analysis revealed genome-wide hypomethylation along with dysregulation in the mechanisms of addition, elimination and maintenance of methyl groups in the DNA of ICM. We describe relevant alterations in the DNMT3B system, which promote a normal Dnmt3b function despite its upregulation.
Collapse
|
12
|
Gan L, Liu D, Xie D, Bond Lau W, Liu J, Christopher TA, Lopez B, Liu L, Hu H, Yao P, He Y, Gao E, Koch WJ, Zhao J, Ma XL, Cao Y, Wang Y. Ischemic Heart-Derived Small Extracellular Vesicles Impair Adipocyte Function. Circ Res 2022; 130:48-66. [PMID: 34763521 DOI: 10.1161/circresaha.121.320157] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Patients with acute myocardial infarction suffer systemic metabolic dysfunction via incompletely understood mechanisms. Adipocytes play critical role in metabolic homeostasis. The impact of acute myocardial infarction upon adipocyte function is unclear. Small extracellular vesicles (sEVs) critically contribute to organ-organ communication. Whether and how small extracellular vesicle mediate post-MI cardiomyocyte/adipocyte communication remain unknown. METHODS Plasma sEVs were isolated from sham control (Pla-sEVSham) or 3 hours after myocardial ischemia/reperfusion (Pla-sEVMI/R) and incubated with adipocytes for 24 hours. Compared with Pla-sEVSham, Pla-sEVMI/R significantly altered expression of genes known to be important in adipocyte function, including a well-known metabolic regulatory/cardioprotective adipokine, APN (adiponectin). Pla-sEVMI/R activated 2 (PERK-CHOP and ATF6 [transcription factor 6]-EDEM [ER degradation enhancing alpha-mannosidase like protein 1] pathways) of the 3 endoplasmic reticulum (ER) stress pathways in adipocytes. These pathological alterations were also observed in adipocytes treated with sEVs isolated from adult cardiomyocytes subjected to in vivo myocardial ischemia/reperfusion (MI/R) (Myo-sEVMI/R). Bioinformatic/RT-qPCR analysis demonstrates that the members of miR-23-27-24 cluster are significantly increased in Pla-sEVMI/R, Myo-sEVMI/R, and adipose tissue of MI/R animals. Administration of cardiomyocyte-specific miR-23-27-24 sponges abolished adipocyte miR-23-27-24 elevation in MI/R animals, supporting the cardiomyocyte origin of adipocyte miR-23-27-24 cluster. In similar fashion to Myo-sEVMI/R, a miR-27a mimic activated PERK-CHOP and ATF6-EDEM-mediated ER stress. Conversely, a miR-27a inhibitor significantly attenuated Myo-sEVMI/R-induced ER stress and restored APN production. RESULTS An unbiased approach identified EDEM3 (ER degradation enhancing alpha-mannosidase like protein 3) as a novel downstream target of miR-27a. Adipocyte EDEM3 deficiency phenocopied multiple pathological alterations caused by Myo-sEVMI/R, whereas EDEM3 overexpression attenuated Myo-sEVMI/R-resulted ER stress. Finally, administration of GW4869 or cardiomyocyte-specific miR-23-27-24 cluster sponges attenuated adipocyte ER stress, improved adipocyte endocrine function, and restored plasma APN levels in MI/R animals. CONCLUSIONS We demonstrate for the first time that MI/R causes significant adipocyte ER stress and endocrine dysfunction by releasing miR-23-27-24 cluster-enriched small extracellular vesicle. Targeting small extracellular vesicle-mediated cardiomyocyte-adipocyte pathological communication may be of therapeutic potential to prevent metabolic dysfunction after MI/R.
Collapse
Affiliation(s)
- Lu Gan
- Laboratory of Emergency Medicine, Department of Emergency Medicine and National Clinical Research Center for Geriatrics, West China Hospital (L.G., L.L., H.H., P.Y., Y.H., Y.C.), Sichuan University, Chengdu, China
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Demin Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China (D.L.)
| | - Dina Xie
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Jing Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Theodore A Christopher
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Bernard Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Lian Liu
- Laboratory of Emergency Medicine, Department of Emergency Medicine and National Clinical Research Center for Geriatrics, West China Hospital (L.G., L.L., H.H., P.Y., Y.H., Y.C.), Sichuan University, Chengdu, China
| | - Hang Hu
- Laboratory of Emergency Medicine, Department of Emergency Medicine and National Clinical Research Center for Geriatrics, West China Hospital (L.G., L.L., H.H., P.Y., Y.H., Y.C.), Sichuan University, Chengdu, China
| | - Peng Yao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Yarong He
- Laboratory of Emergency Medicine, Department of Emergency Medicine and National Clinical Research Center for Geriatrics, West China Hospital (L.G., L.L., H.H., P.Y., Y.H., Y.C.), Sichuan University, Chengdu, China
| | - Erhe Gao
- Laboratory of Emergency Medicine, Department of Emergency Medicine and National Clinical Research Center for Geriatrics, West China Hospital (L.G., L.L., H.H., P.Y., Y.H., Y.C.), Sichuan University, Chengdu, China
- Center for Translational Medicine, Temple University, Philadelphia, PA (E.G., W.J.K.)
| | - Walter J Koch
- Center for Translational Medicine, Temple University, Philadelphia, PA (E.G., W.J.K.)
| | - Jianli Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Yu Cao
- Disaster Medical Center (Y.C.), Sichuan University, Chengdu, China
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| |
Collapse
|
13
|
Boyd R, McMullen H, Beqaj H, Kalfa D. Environmental Exposures and Congenital Heart Disease. Pediatrics 2022; 149:183839. [PMID: 34972224 DOI: 10.1542/peds.2021-052151] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 12/16/2022] Open
Abstract
Congenital heart disease (CHD) is the most common congenital abnormality worldwide, affecting 8 to 12 infants per 1000 births globally and causing >40% of prenatal deaths. However, its causes remain mainly unknown, with only up to 15% of CHD cases having a determined genetic cause. Exploring the complex relationship between genetics and environmental exposures is key in understanding the multifactorial nature of the development of CHD. Multiple population-level association studies have been conducted on maternal environmental exposures and their association with CHD, including evaluating the effect of maternal disease, medication exposure, environmental pollution, and tobacco and alcohol use on the incidence of CHD. However, these studies have been done in a siloed manner, with few examining the interplay between multiple environmental exposures. Here, we broadly and qualitatively review the current literature on maternal and paternal prenatal exposures and their association with CHD. We propose using the framework of the emerging field of the exposome, the environmental complement to the genome, to review all internal and external prenatal environmental exposures and identify potentiating or alleviating synergy between exposures. Finally, we propose mechanistic pathways through which susceptibility to development of CHD may be induced via the totality of prenatal environmental exposures, including the interplay between placental and cardiac development and the internal vasculature and placental morphology in early stages of pregnancy.
Collapse
|
14
|
Xu J, Liu X, Dai Q. Integration of transcriptomic data identifies key hallmark genes in hypertrophic cardiomyopathy. BMC Cardiovasc Disord 2021; 21:330. [PMID: 34225646 PMCID: PMC8259117 DOI: 10.1186/s12872-021-02147-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 06/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) represents one of the most common inherited heart diseases. To identify key molecules involved in the development of HCM, gene expression patterns of the heart tissue samples in HCM patients from multiple microarray and RNA-seq platforms were investigated. METHODS The significant genes were obtained through the intersection of two gene sets, corresponding to the identified differentially expressed genes (DEGs) within the microarray data and within the RNA-Seq data. Those genes were further ranked using minimum-Redundancy Maximum-Relevance feature selection algorithm. Moreover, the genes were assessed by three different machine learning methods for classification, including support vector machines, random forest and k-Nearest Neighbor. RESULTS Outstanding results were achieved by taking exclusively the top eight genes of the ranking into consideration. Since the eight genes were identified as candidate HCM hallmark genes, the interactions between them and known HCM disease genes were explored through the protein-protein interaction (PPI) network. Most candidate HCM hallmark genes were found to have direct or indirect interactions with known HCM diseases genes in the PPI network, particularly the hub genes JAK2 and GADD45A. CONCLUSIONS This study highlights the transcriptomic data integration, in combination with machine learning methods, in providing insight into the key hallmark genes in the genetic etiology of HCM.
Collapse
Affiliation(s)
- Jing Xu
- Department of Clinical Laboratory, ZhongDa Hospital, Southeast University, Nanjing, China
| | - Xiangdong Liu
- Institute of Life Science, Southeast University, Nanjing, China
| | - Qiming Dai
- Department of Cardiology, ZhongDa Hospital, Southeast University, Nanjing, China.
| |
Collapse
|
15
|
Song Y, Choi JE, Kwon YJ, Chang HJ, Kim JO, Park DH, Park JM, Kim SJ, Lee JW, Hong KW. Identification of susceptibility loci for cardiovascular disease in adults with hypertension, diabetes, and dyslipidemia. J Transl Med 2021; 19:85. [PMID: 33632238 PMCID: PMC7905883 DOI: 10.1186/s12967-021-02751-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/11/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Hypertension (HTN), diabetes mellitus (DM), and dyslipidemia (DL) are well-known risk factors of cardiovascular disease (CVD), but not all patients develop CVDs. Studies have been limited investigating genetic risk of CVDs specific to individuals with metabolic diseases. This study aimed to identify disease-specific and/or common genetic loci associated with CVD susceptibility in chronic metabolic disease patients. METHODS We conducted a genome-wide association study (GWAS) of a multiple case-control design with data from the City Cohort within Health EXAminees subcohort of the Korean Genome and Epidemiology Study (KoGES_HEXA). KoGES_HEXA is a population-based prospective cohort of 173,357 urban Korean adults that had health examinations at medical centers. 42,393 participants (16,309 HTN; 5,314 DM; 20,770 DL) were analyzed, and each metabolic disease group was divided into three CVD case-controls: coronary artery disease (CAD), ischemic stroke (IS), and cardio-cerebrovascular disease (CCD). GWASs were conducted for each case-control group with 7,975,321 imputed single nucleotide polymorphisms using the Phase 3 Asian panel from 1000 Genomes Project, by logistic regression and controlled for confounding variables. Genome-wide significant levels were implemented to identify important susceptibility loci. RESULTS Totaling 42,393 individuals, this study included 16,309 HTN (mean age [SD], 57.28 [7.45]; 816 CAD, 398 IS, and 1,185 CCD cases), 5,314 DM (57.79 [7.39]; 361 CAD, 153 IS, and 497 CCD cases), and 20,770 DL patients (55.34 [7.63]; 768 CAD, 295 IS, and 1,039 CCD cases). Six genome-wide significant CVD risk loci were identified, with relatively large effect sizes: 1 locus in HTN (HTN-CAD: 17q25.3/CBX8-CBX4 [OR, 2.607; P = 6.37 × 10-9]), 2 in DM (DM-IS: 4q32.3/MARCH1-LINC01207 [OR, 5.587; P = 1.34 × 10-8], and DM-CCD: 17q25.3/RPTOR [OR, 3.511; P = 1.99 × 10-8]), and 3 in DL (DL-CAD: 9q22.2/UNQ6494-LOC101927847 [OR, 2.282; P = 7.78 × 10-9], DL-IS: 3p22.1/ULK4 [OR, 2.162; P = 2.97 × 10-8], and DL-CCD: 2p22.2/CYP1B1-CYP1B1-AS1 [OR, 2.027; P = 4.24 × 10-8]). CONCLUSIONS This study identified 6 susceptibility loci and positional candidate genes for CVDs in HTN, DM, and DL patients using an unprecedented study design. 1 locus (17q25.3) was commonly associated with CAD. These associations warrant validation in additional studies for potential therapeutic applications.
Collapse
Affiliation(s)
- Youhyun Song
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211, Eonju-ro, Gangnam-gu, Seoul, 06273, Korea
| | - Ja-Eun Choi
- Healthcare R&D Division, Theragen Bio Co., Ltd., Gwanggyo-ro 145, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Yu-Jin Kwon
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, 363, Dongbaekjukjeon-daero, Giheung-gu, Yongin-si, 16995, Gyeonggi-do, Korea
| | - Hyuk-Jae Chang
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jung Oh Kim
- Healthcare R&D Division, Theragen Bio Co., Ltd., Gwanggyo-ro 145, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Da-Hyun Park
- Healthcare R&D Division, Theragen Bio Co., Ltd., Gwanggyo-ro 145, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Jae-Min Park
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211, Eonju-ro, Gangnam-gu, Seoul, 06273, Korea
| | - Seong-Jin Kim
- Healthcare R&D Division, Theragen Bio Co., Ltd., Gwanggyo-ro 145, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Ji Won Lee
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211, Eonju-ro, Gangnam-gu, Seoul, 06273, Korea.
| | - Kyung-Won Hong
- Healthcare R&D Division, Theragen Bio Co., Ltd., Gwanggyo-ro 145, Suwon-si, Gyeonggi-do, 16229, Republic of Korea.
| |
Collapse
|
16
|
David H, Ughetto A, Gaudard P, Plawecki M, Paiyabhroma N, Zub E, Colson P, Richard S, Marchi N, Sicard P. Experimental Myocardial Infarction Elicits Time-Dependent Patterns of Vascular Hypoxia in Peripheral Organs and in the Brain. Front Cardiovasc Med 2021; 7:615507. [PMID: 33585582 PMCID: PMC7873295 DOI: 10.3389/fcvm.2020.615507] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
Aims: Microvascular alterations occurring after myocardial infarction (MI) may represent a risk factor for multi-organ failure. Here we used in vivo photoacoustic (PA) imaging to track and define the changes in vascular oxygen saturation (sO2) occurring over time after experimental MI in multiple peripheral organs and in the brain. Methods and Results: Experimental MI was obtained in BALB/c mice by permanent ligation of the left anterior descending artery. PA imaging (Vevo LAZR-X) allowed tracking mouse-specific sO2 kinetics in the cardiac left ventricular (LV) anterior wall, brain, kidney, and liver at 4 h, 1 day, and 7 days post-MI. Here we reported a correlation between LV sO2 and longitudinal anterior myocardial strain after MI (r = −0.44, p < 0.0001, n = 96). Acute LV dysfunction was associated with global hypoxia, specifically a decrease in sO2 level in the brain (−5.9%), kidney (−6.4%), and liver (−7.3%) at 4 and 24 h post-MI. Concomitantly, a preliminary examination of capillary NG2DsRed pericytes indicated cell rarefication in the heart and kidney. While the cardiac tissue was persistently impacted, sO2 levels returned to pre-MI levels in the brain and in peripheral organs 7 days after MI. Conclusions: Collectively, our data indicate that experimental MI elicits precise trajectories of vascular hypoxia in peripheral organs and in the brain. PA imaging enabled the synchronous tracking of oxygenation in multiple organs and occurring post-MI, potentially enabling a translational diagnostic modality for the identification of vascular modifications in this disease setting.
Collapse
Affiliation(s)
- Hélène David
- INSERM, CNRS, Université de Montpellier, PHYMEDEXP, Montpellier, France.,Department of Anesthesiology and Critical Care Medicine, Arnaud de Villeneuve Hospital, CHU Montpellier, Montpellier, France
| | - Aurore Ughetto
- INSERM, CNRS, Université de Montpellier, PHYMEDEXP, Montpellier, France.,Department of Anesthesiology and Critical Care Medicine, Arnaud de Villeneuve Hospital, CHU Montpellier, Montpellier, France
| | - Philippe Gaudard
- INSERM, CNRS, Université de Montpellier, PHYMEDEXP, Montpellier, France.,Department of Anesthesiology and Critical Care Medicine, Arnaud de Villeneuve Hospital, CHU Montpellier, Montpellier, France
| | - Maëlle Plawecki
- INSERM, CNRS, Université de Montpellier, PHYMEDEXP, Montpellier, France.,CHU Lapeyronie, Département de Biochimie, Montpellier, France
| | | | - Emma Zub
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Pascal Colson
- Department of Anesthesiology and Critical Care Medicine, Arnaud de Villeneuve Hospital, CHU Montpellier, Montpellier, France.,Montpellier University, INSERM, CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Sylvain Richard
- INSERM, CNRS, Université de Montpellier, PHYMEDEXP, Montpellier, France
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Pierre Sicard
- INSERM, CNRS, Université de Montpellier, PHYMEDEXP, Montpellier, France.,IPAM, BioCampus Montpellier, CNRS, INSERM, Université de Montpellier, Montpellier, France
| |
Collapse
|
17
|
Gong L, Cai L, Li G, Cai J, Yi X. GADD45B Facilitates Metastasis of Ovarian Cancer Through Epithelial-Mesenchymal Transition. Onco Targets Ther 2021; 14:255-269. [PMID: 33469306 PMCID: PMC7811469 DOI: 10.2147/ott.s281450] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/11/2020] [Indexed: 12/22/2022] Open
Abstract
Background Growth arrest and DNA-damage-inducible 45 beta (GADD45B) is overexpressed and is associated with poor clinical outcomes in many human cancers, but the clinical implication of GADD45B in epithelial ovarian cancer (EOC) remains unclear. Methods Bioinformatics analysis of The Cancer Genome Atlas (TCGA) and gene expression omnibus (GEO) cohorts was used to illustrate the relationship between GADD45B expression and metastasis, as well as the survival time of EOC. GADD45B was downregulated by siRNAs in EOC cells, and migration ability was determined by a transwell assay and wound-healing assay. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis and gene set enrichment analysis (GSEA) were conducted to discover the downstream pathway of GADD45B. The regulation of epithelial–mesenchymal transition (EMT) by GADD45B was verified by Western blotting and qRT-PCR. Finally, the correlation of GADD45B expression with EOC metastasis was investigated in EOC tissues by immunohistochemistry. Results Overexpression of GADD45B indicates shorter overall survival time and progression-free survival time, and it is an independent risk factor for poor survival in EOC patients. Elevated GADD45B is related to venous invasion, lymphatic invasion and peritoneal carcinomatosis. Downregulation of GADD45B decreases the migration of ES2 and SKOV3 cells. Further KEGG enrichment analysis and GSEA revealed that EMT may be the downstream pathway of GADD45B. In addition, reduced GADD45B increases the expression of E-cadherin and decreases that of N-cadherin and vimentin. Finally, immunohistochemical analysis of GADD45B expression revealed that the expression of GADD45B in omental metastatic tissues was higher than that in matched primary ovarian cancer tissues. These results suggest that elevated GADD45B promotes the motility of ovarian cancer cells through EMT and is associated with EOC metastasis. Conclusion GADD45B can promote the motility of ovarian cancer cells through EMT, is associated with EOC metastasis, and may be a new biomarker of metastasis and prognosis.
Collapse
Affiliation(s)
- Lanqing Gong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Liqiong Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Guodong Li
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Xiaoqing Yi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| |
Collapse
|
18
|
Simats A, Ramiro L, García-Berrocoso T, Briansó F, Gonzalo R, Martín L, Sabé A, Gill N, Penalba A, Colomé N, Sánchez A, Canals F, Bustamante A, Rosell A, Montaner J. A Mouse Brain-based Multi-omics Integrative Approach Reveals Potential Blood Biomarkers for Ischemic Stroke. Mol Cell Proteomics 2020; 19:1921-1936. [PMID: 32868372 PMCID: PMC7710142 DOI: 10.1074/mcp.ra120.002283] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke remains a leading cause of death and disability worldwide. Despite continuous advances, the identification of key molecular signatures in the hyper-acute phase of ischemic stroke is still a primary interest for translational research on stroke diagnosis, prognosis, and treatment. Data integration from high-throughput -omics techniques has become crucial to unraveling key interactions among different molecular elements in complex biological contexts, such as ischemic stroke. Thus, we used advanced data integration methods for a multi-level joint analysis of transcriptomics and proteomics data sets obtained from mouse brains at 2 h after cerebral ischemia. By modeling net-like correlation structures, we identified an integrated network of genes and proteins that are differentially expressed at a very early stage after stroke. We validated 10 of these deregulated elements in acute stroke, and changes in their expression pattern over time after cerebral ischemia were described. Of these, CLDN20, GADD45G, RGS2, BAG5, and CTNND2 were next evaluated as blood biomarkers of cerebral ischemia in mice and human blood samples, which were obtained from stroke patients and patients presenting stroke-mimicking conditions. Our findings indicate that CTNND2 levels in blood might potentially be useful for distinguishing ischemic strokes from stroke-mimicking conditions in the hyper-acute phase of the disease. Furthermore, circulating GADD45G content within the first 6 h after stroke could also play a key role in predicting poor outcomes in stroke patients. For the first time, we have used an integrative biostatistical approach to elucidate key molecules in the initial stages of stroke pathophysiology and highlight new notable molecules that might be further considered as blood biomarkers of ischemic stroke.
Collapse
Affiliation(s)
- Alba Simats
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Ramiro
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Teresa García-Berrocoso
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ferran Briansó
- Bioinformatics and Biostatistics Unit, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Genetics, Microbiology and Statistics Dept., Universitat de Barcelona, Barcelona, Spain
| | - Ricardo Gonzalo
- Bioinformatics and Biostatistics Unit, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Luna Martín
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Sabé
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Natalia Gill
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Penalba
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nuria Colomé
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Sánchez
- Bioinformatics and Biostatistics Unit, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Genetics, Microbiology and Statistics Dept., Universitat de Barcelona, Barcelona, Spain
| | - Francesc Canals
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
19
|
Yang Y, Li J, Geng Y, Liu L, Li D. Azacitidine regulates DNA methylation of GADD45γ in myelodysplastic syndromes. J Clin Lab Anal 2020; 35:e23597. [PMID: 33080073 PMCID: PMC7891504 DOI: 10.1002/jcla.23597] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/30/2020] [Accepted: 09/04/2020] [Indexed: 12/19/2022] Open
Abstract
Background Myelodysplastic syndrome (MDS) is a heterogeneous clonal disease originated from hematopoietic stem cells. Epigenetic studies had demonstrated that DNA methylation and histone acetylation were abnormal in MDS. Azacitidine is an effective drug in the treatment of demethylation. Methods RT‐PCR was performed to determine GADD45γ in 15 MDS clinical samples. Myelodysplastic syndrome cell lines SKM‐1 and HS‐5 were transfected with GADD45γ eukaryotic expression vector and/or GADD45γ shRNA interference plasmid, and treated with azacitidine. Proliferation and apoptosis were examined by CCK‐8 and Western blot analysis to confirm the function role of GADD45γ and azacitidine. The methylation level of GADD45γ gene was detected by bisulfite conversion and PCR. Results This study found that GADD45γ gene was down‐expressed in MDS patients' bone marrow and MDS cell lines, and the down‐regulation of GADD45γ in MDS could inhibit MDS cell apoptosis and promote proliferation. Azacitidine, a demethylation drug, could restore the expression of GADD45γ in MDS cells and inhibit the proliferation of MDS cells by inducing apoptosis, which was related to prognosis and transformation. Conclusion This study indicated that GADD45γ was expected to become a new target of MDS‐targeted therapy. The findings of this study provided a new direction for the research and development of new MDS clinical drugs, and gave a new idea for the development of MDS demethylation drug to realize precise treatment.
Collapse
Affiliation(s)
- Yanli Yang
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Jun Li
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Yinghua Geng
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Lin Liu
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Dianming Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
20
|
Iwahana T, Okada S, Kanda M, Oshima M, Iwama A, Matsumiya G, Kobayashi Y. Novel myocardial markers GADD45G and NDUFS5 identified by RNA-sequencing predicts left ventricular reverse remodeling in advanced non-ischemic heart failure: a retrospective cohort study. BMC Cardiovasc Disord 2020; 20:116. [PMID: 32138671 PMCID: PMC7059273 DOI: 10.1186/s12872-020-01396-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/21/2020] [Indexed: 12/17/2022] Open
Abstract
Background Left ventricular reverse remodeling (LVRR) has been detected in non-ischemic dilated cardiomyopathy (NIDCM) patients following optimal treatment. However, its prediction with only conventional modalities is often difficult. This study sought to examine whether RNA sequencing (RNA-seq) of myocardium tissue samples could predict LVRR in NIDCM. Methods A total of 17 advanced NIDCM patients with left ventricular ejection fraction (LVEF) below 30% who underwent cardiac biopsy from Left ventricle (LV) were prospectively recruited. They received optimal treatment and followed with echocardiogram every 6 months. Based on LVRR status after 12 months of treatment, patients were divided into the reverse remodeling (RR) or non-RR group. Tissue samples were analyzed by RNA-seq, and a functional analysis of differentially expressed genes was carried out. Results There were eight and nine patients in the RR and non-RR groups, respectively. No difference was found in age, sex, disease duration, LV end-diastolic diameter, and LVEF between the two groups. There were 155 genes that were differentially expressed between the two groups. Nicotinamide adenine dinucleotide ubiquinone oxidoreductase subunit (NDUF)S5 and Growth arrest and DNA-damage-inducible protein (GADD)45G, along with several genes related to the mitochondrial respiratory chain and ribosome, were significantly downregulated in the RR as compared to the non-RR group. Conclusion GADD45G and NDUFS5 are potential biomarkers for LVRR in patients with advanced NIDCM.
Collapse
Affiliation(s)
- Togo Iwahana
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Sho Okada
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| | - Masato Kanda
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Motohiko Oshima
- Department of Cellular and Molecular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Atsushi Iwama
- Department of Cellular and Molecular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Goro Matsumiya
- Department of Cardiovascular Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| |
Collapse
|
21
|
Lu Y, Liang M, Zhang Q, Liu Z, Song Y, Lai L, Li Z. Mutations of GADD45G in rabbits cause cleft lip by the disorder of proliferation, apoptosis and epithelial-mesenchymal transition (EMT). Biochim Biophys Acta Mol Basis Dis 2019; 1865:2356-2367. [PMID: 31150757 DOI: 10.1016/j.bbadis.2019.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/17/2019] [Accepted: 05/27/2019] [Indexed: 12/19/2022]
Abstract
The cleft lip with or without cleft palate (CL/P) is one of the most common congenital defects in humans. Genome-wide association studies (GWAS) have been widely used for identifying candidate genes, and different genes or chromosomal regions have shown strong evidence for the presence of causal genes in CL/P. To date, two independent GWAS have identified GADD45G as influencing risk for CL/P. However, there is no animal model evidence about GADD45G related to CL/P. Here, we reported the generation of a novel GADD45G mutated rabbit model by CRISPR/Cas9 and CRISPR-based BE4-Gam systems. The homozygous (GADD45G-/-) while not heterozygous (GADD45G+/-) pups died after birth due to severe craniofacial defects of unilateral or bilateral cleft lip (CL). Moreover, the disorder of proliferation, apoptosis and epithelial-mesenchymal transition (EMT) were also determined in the medial and lateral nasal processes (MNP and LNP) of the embryonic day 13 (E13) GADD45G-/- rabbits, which compared with the normal wild type (WT) rabbits. Thus, our study confirmed for the first time that loss of GADD45G lead to CL at the animal level and provided new insights into the crucial role of GADD45G for upper lip formation and fusion.
Collapse
Affiliation(s)
- Yi Lu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Mingming Liang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Quanjun Zhang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Zhiquan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuning Song
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Liangxue Lai
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China; CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Zhanjun Li
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
22
|
Finan A, Demion M, Sicard P, Guisiano M, Bideaux P, Monceaux K, Thireau J, Richard S. Prolonged elevated levels of c-kit+ progenitor cells after a myocardial infarction by beta 2 adrenergic receptor priming. J Cell Physiol 2019; 234:18283-18296. [PMID: 30912139 DOI: 10.1002/jcp.28461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 12/23/2022]
Abstract
Endogenous progenitor cells may participate in cardiac repair after a myocardial infarction (MI). The beta 2 adrenergic receptor (ß2-AR) pathway induces proliferation of c-kit+ cardiac progenitor cells (CPC) in vitro. We investigated if ß2-AR pharmacological stimulation could ameliorate endogenous CPC-mediated regeneration after a MI. C-kit+ CPC ß1-AR and ß2-AR expression was evaluated in vivo and in vitro. A significant increase in the percentage of CPCs expressing ß1-AR and ß2-AR was measured 7 days post-MI. Accordingly, 24 hrs of low serum and hypoxia in vitro significantly increased CPC ß2-AR expression. Cell viability and differentiation assays validated a functional role of CPC ß2-AR. The effect of pharmacological activation of ß2-AR was studied in C57 mice using fenoterol administered in the drinking water 1 week before MI or sham surgery or at the time of the surgery. MI induced a significant increase in the percentage of c-kit+ progenitor cells at 7 days, whereas pretreatment with fenoterol prolonged this response resulting in a significant elevated number of CPC up to 21 days post-MI. This increased number of CPC correlated with a decrease in infarct size. The immunofluorescence analysis of the heart tissue for proliferation, apoptosis, macrophage infiltration, cardiomyocytes surface area, and vessel density showed significant changes on the basis of surgery but no benefit due to fenoterol treatment. Cardiac function was not ameliorated by fenoterol administration when evaluated by echocardiography. Our results suggest that ß2-AR stimulation may improve the cardiac repair process by supporting an endogenous progenitor cell response but is not sufficient to improve the cardiac function.
Collapse
Affiliation(s)
- Amanda Finan
- Physiology & Experimental Medicine of the Heart and Muscles (PhyMedExp), INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Marie Demion
- Physiology & Experimental Medicine of the Heart and Muscles (PhyMedExp), INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Pierre Sicard
- Physiology & Experimental Medicine of the Heart and Muscles (PhyMedExp), INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Morgane Guisiano
- Physiology & Experimental Medicine of the Heart and Muscles (PhyMedExp), INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Patrice Bideaux
- Physiology & Experimental Medicine of the Heart and Muscles (PhyMedExp), INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Kevin Monceaux
- Physiology & Experimental Medicine of the Heart and Muscles (PhyMedExp), INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Jérôme Thireau
- Physiology & Experimental Medicine of the Heart and Muscles (PhyMedExp), INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - Sylvain Richard
- Physiology & Experimental Medicine of the Heart and Muscles (PhyMedExp), INSERM U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| |
Collapse
|
23
|
Shin GT, Lee HJ, Park JE. Growth arrest and DNA damage 45γ is required for caspase-dependent renal tubular cell apoptosis. PLoS One 2019; 14:e0212818. [PMID: 30794682 PMCID: PMC6386268 DOI: 10.1371/journal.pone.0212818] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/08/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Growth Arrest and DNA Damage 45γ (GADD45γ) is a member of the DNA damage-inducible gene family which responds to environmental stresses. Apoptosis is a critical mode of renal tubular cell death in nephrotoxin-induced acute kidney injury. In this study, we investigated the role of GADD45γ in renal tubular cell apoptosis induced by nephrotoxic drugs. METHODS Primary human renal tubular epithelial (HRE) cells were used in this study. To derive stable cell lines in which GADD45γ expression was silenced, HRE cells were transduced with a plasmid encoding GADD45γ-specific shRNA. The recombinant adenovirus containing the GADD45γ gene was synthesized to overexpress GADD45γ protein. Cell death was induced by cisplatin and cyclosporine A (CsA). To prevent apoptotic cell death, pan-caspase inhibitor ZVAD-FMK was used. To prevent non-apoptotic cell death, necrostatin-1 and ferrostatin-1 were used. The degree of apoptosis and necrosis of cultured cells were evaluated by flow cytometry. RESULTS Expression of the GADD45γ gene was significantly upregulated in response to treatment with CsA and cisplatin. Apoptosis and necrosis induced by these drugs were significantly reduced by silencing of GADD45γ, and significantly augmented by the overexpression of GADD45γ. The activation of caspase-3 and caspase-7 as well as caspase-9 induced by cisplatin or CsA was reduced by silencing of GADD45γ, and was augmented by the overexpression of GADD45γ, indicating that caspase activation is dependent on the expression of GADD45γ. ZVAD-FMK significantly inhibited apoptosis induced by cisplatin or CsA, indicating a role of caspases in mediating apoptotic cell death. ZVAD-FMK was effective to prevent necrosis as well, indicating that the observed necrosis was a secondary event following apoptosis at least in part. CONCLUSIONS To our knowledge, this is the first study to show that GADD45γ is required for the caspase-dependent apoptosis of renal tubular cells induced by nephrotoxic drugs.
Collapse
Affiliation(s)
- Gyu-Tae Shin
- Department of Nephrology, Ajou University School of Medicine, Suwon, Korea
| | - Hwa Joung Lee
- Department of Nephrology, Ajou University School of Medicine, Suwon, Korea
| | - Ji Eun Park
- Department of Nephrology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
24
|
Sicard P, Jouitteau T, Andrade-Martins T, Massad A, Rodrigues de Araujo G, David H, Miquerol L, Colson P, Richard S. Right coronary artery ligation in mice: a novel method to investigate right ventricular dysfunction and biventricular interaction. Am J Physiol Heart Circ Physiol 2018; 316:H684-H692. [PMID: 30575433 DOI: 10.1152/ajpheart.00573.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Right ventricular (RV) dysfunction can lead to complications after acute inferior myocardial infarction (MI). However, it is unclear how RV failure after MI contributes to left-sided dysfunction. The aim of the present study was to investigate the consequences of right coronary artery (RCA) ligation in mice. RCA ligation was performed in C57BL/6JRj mice ( n = 38). The cardiac phenotypes were characterized using high-resolution echocardiography performed up to 4 wk post-RCA ligation. Infarct size was measured using 2,3,5-triphenyltetrazolium chloride staining 24 h post-RCA ligation, and the extent of the fibrotic area was determined 4 wk after MI. RV dysfunction was confirmed 24 h post-RCA ligation by a decrease in the tricuspid annular plane systolic excursion ( P < 0.001) and RV longitudinal strain analysis ( P < 0.001). Infarct size measured ex vivo represented 45.1 ± 9.1% of the RV free wall. RCA permanent ligation increased the RV-to-left ventricular (LV) area ratio ( P < 0.01). Septum hypertrophy ( P < 0.01) was associated with diastolic septal flattening. During the 4-wk post-RCA ligation, LV ejection fraction was preserved, yet it was associated with impaired LV diastolic parameters ( E/ E', global strain rate during early diastole). Histological staining after 4 wk confirmed the remodeling process with a thin and fibrotic RV. This study validates that RCA ligation in mice is feasible and induces RV heart failure associated with the development of LV diastolic dysfunction. Our model offers a new opportunity to study mechanisms and treatments of RV/LV dysfunction after MI. NEW & NOTEWORTHY Right ventricular (RV) dysfunction frequently causes complications after acute inferior myocardial infarction. How RV failure contributes to left-sided dysfunction is elusive because of the lack of models to study molecular mechanisms. Here, we created a new model of myocardial infarction by permanently tying the right coronary artery in mice. This model offers a new opportunity to unravel mechanisms underlying RV/left ventricular dysfunction and evaluate drug therapy.
Collapse
Affiliation(s)
- Pierre Sicard
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, PhyMedExp, Montpellier , France
| | - Timothée Jouitteau
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, PhyMedExp, Montpellier , France.,Department of Anaesthesiology and Critical Care Medicine, Arnaud de Villeneuve Academic Hospital , Montpellier , France
| | - Thales Andrade-Martins
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, PhyMedExp, Montpellier , France.,Department of Anaesthesiology and Critical Care Medicine, Arnaud de Villeneuve Academic Hospital , Montpellier , France.,CiPharma, Escola de Farmácia, Universidade Federal de Ouro Preto , Minas Gerais , Brazil
| | - Abdallah Massad
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, PhyMedExp, Montpellier , France
| | | | - Hélène David
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, PhyMedExp, Montpellier , France.,Department of Anaesthesiology and Critical Care Medicine, Arnaud de Villeneuve Academic Hospital , Montpellier , France
| | - Lucile Miquerol
- Aix-Marseille University, CNRS, Institut de Biologie du Développement de Marseille, Marseille , France
| | - Pascal Colson
- Department of Anaesthesiology and Critical Care Medicine, Arnaud de Villeneuve Academic Hospital , Montpellier , France
| | - Sylvain Richard
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, PhyMedExp, Montpellier , France
| |
Collapse
|
25
|
Warr N, May J, Teboul L, Suzuki T, Asami M, Perry ACF, Wells S, Greenfield A. Characterisation and use of a functional Gadd45g bacterial artificial chromosome. Sci Rep 2018; 8:17318. [PMID: 30470800 PMCID: PMC6251886 DOI: 10.1038/s41598-018-35458-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/05/2018] [Indexed: 01/23/2023] Open
Abstract
Bacterial artificial chromosomes (BACs) offer a means of manipulating gene expression and tagging gene products in the mammalian genome without the need to alter endogenous gene structure and risk deleterious phenotypic consequences. However, for a BAC clone to be useful for such purposes it must be shown to contain all the regulatory elements required for normal gene expression and allow phenotypic rescue in the absence of an endogenous gene. Here, we report identification of a functional BAC containing Gadd45g, a gene implicated in DNA repair, DNA demethylation and testis determination in mice and exhibiting a broad pattern of embryonic expression. Mouse fetuses lacking the endogenous Gadd45g gene undergo normal testis development in the presence of the Gadd45g BAC transgene. Moreover, a survey of embryonic Gadd45g expression from the BAC reveals that all reported sites of expression are maintained. This functional BAC can now be used for subsequent manipulation of the Gadd45g gene with the confidence that regulatory elements required for embryonic expression, including testis determination, are present. We describe the generation and characterisation of a Gadd45g-mCherry fluorescent reporter exhibiting strong expression in developing gonads and neural tissue, recapitulating endogenous gene expression, as evidence of this.
Collapse
Affiliation(s)
- Nick Warr
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Joel May
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
- Boston University School of Medicine, Vascular Biology Section, 650 Albany St, X720, Boston, MA, 02118, USA
| | - Lydia Teboul
- The Mary Lyon Centre, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Toru Suzuki
- Laboratory of Mammalian Molecular Embryology, Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK
| | - Maki Asami
- Laboratory of Mammalian Molecular Embryology, Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK
| | - Anthony C F Perry
- Laboratory of Mammalian Molecular Embryology, Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK
| | - Sara Wells
- The Mary Lyon Centre, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Andy Greenfield
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK.
| |
Collapse
|
26
|
Liu J, Yan W, Zhao X, Jia Q, Wang J, Zhang H, Liu C, He K, Sun Z. Sirt3 attenuates post-infarction cardiac injury via inhibiting mitochondrial fission and normalization of AMPK-Drp1 pathways. Cell Signal 2018; 53:1-13. [PMID: 30219671 DOI: 10.1016/j.cellsig.2018.09.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 01/15/2023]
Abstract
Mitochondrial damage is involved in the pathogenesis of post-infarction cardiac injury. However, the upstream regulators of mitochondrial damage have not yet been identified. The aim of our study is to explore the role of Sirt3 in post-infarction cardiac injury with a particular focus on mitochondrial fission and AMPK-Drp1 pathways. Our results indicated that Sirt3 was downregulated in the progression of post-infarction cardiac injury. Overexpression of Sirt3 attenuated cardiac fibrosis, sustained myocardial function, inhibited the inflammatory response, and reduced cardiomyocyte death. Functional studies illustrated that chronic post-infarction cardiac injury was characterized by increased mitochondrial fission, which triggered mitochondrial oxidative stress, metabolic disorders, mitochondrial potential reduction and caspase-9 apoptosis in cardiomyocytes. However, Sirt3 overexpression attenuated mitochondrial fission and thus preserved mitochondrial homeostasis and cardiomyocyte viability. Furthermore, our results confirmed that Sirt3 repressed mitochondrial fission via normalizing AMPK-Drp1 pathways. Inhibition of AMPK activity re-activated Drp1 and thus abrogated the inhibitory effect of Sirt3 on mitochondrial fission. Altogether, our results indicate that Sirt3 enhancement could be an effective approach to retard the development of post-infarction cardiac injury via disrupting mitochondrial fission and normalizing the AMPK-Drp1 axis.
Collapse
Affiliation(s)
- Jixuan Liu
- Department of Cardiovascular, Chinese PLA General Hospital, Beijing 100853, China
| | - Wei Yan
- Department of Geriatric Medicine, The First Affiliated Hospital of Soochow University, Soochow 215000, China
| | - Xiaojing Zhao
- Transformation Medicine Centre, Chinese PLA General Hospital, Beijing 100853, China; Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, 100853, China
| | - Qian Jia
- Transformation Medicine Centre, Chinese PLA General Hospital, Beijing 100853, China; Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, 100853, China
| | - Jinda Wang
- Department of Cardiovascular, Chinese PLA General Hospital, Beijing 100853, China
| | - Huawei Zhang
- Department of Cardiovascular, Chinese PLA General Hospital, Beijing 100853, China
| | - Chunlei Liu
- Transformation Medicine Centre, Chinese PLA General Hospital, Beijing 100853, China; Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, 100853, China
| | - Kunlun He
- Transformation Medicine Centre, Chinese PLA General Hospital, Beijing 100853, China; Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, 100853, China.
| | - Zhijun Sun
- Department of Cardiovascular, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
27
|
Courtney JA, Cnota JF, Jones HN. The Role of Abnormal Placentation in Congenital Heart Disease; Cause, Correlate, or Consequence? Front Physiol 2018; 9:1045. [PMID: 30131711 PMCID: PMC6091057 DOI: 10.3389/fphys.2018.01045] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/13/2018] [Indexed: 01/11/2023] Open
Abstract
Congenital heart disease (CHD) is the most common birth defect, affecting ~1% of all live births (van der Linde et al., 2011). Despite improvements in clinical care, it is the leading cause of infant mortality related to birth defects (Yang et al., 2006) and burdens survivors with significant morbidity (Gilboa et al., 2016). Furthermore, CHD accounts for the largest proportion (26.7%) of birth defect-associated hospitalization costs—up to $6.1 billion in 2013 (Arth et al., 2017). Yet after decades of research with a primary focus on genetic etiology, the underlying cause of these defects remains unknown in the majority of cases (Zaidi and Brueckner, 2017). Unexplained CHD may be secondary to undiscovered roles of noncoding genetic, epigenetic, and environmental factors, among others (Russell et al., 2018). Population studies have recently demonstrated that pregnancies complicated by CHD also carry a higher risk of developing pathologies associated with an abnormal placenta including growth disturbances (Puri et al., 2017), preeclampsia (Auger et al., 2015; Brodwall et al., 2016), preterm birth (Laas et al., 2012), and stillbirth (Jorgensen et al., 2014). Both the heart and placenta are vascular organs and develop concurrently; therefore, shared pathways almost certainly direct the development of both. The involvement of placental abnormalities in congenital heart disease, whether causal, commensurate or reactive, is under investigated and given the common developmental window and shared developmental pathways of the heart and placenta and concurrent vasculature development, we propose that further investigation combining clinical data, in vitro, in vivo, and computer modeling is fundamental to our understanding and the potential to develop therapeutics.
Collapse
Affiliation(s)
- Jennifer A Courtney
- Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Division of General Pediatric and Thoracic Surgery, Center for Fetal and Placental Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - James F Cnota
- Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Helen N Jones
- Division of General Pediatric and Thoracic Surgery, Center for Fetal and Placental Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
28
|
Li C, Sun W, Gu C, Yang Z, Quan N, Yang J, Shi Z, Yu L, Ma H. Targeting ALDH2 for Therapeutic Interventions in Chronic Pain-Related Myocardial Ischemic Susceptibility. Am J Cancer Res 2018; 8:1027-1041. [PMID: 29463997 PMCID: PMC5817108 DOI: 10.7150/thno.22414] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/09/2017] [Indexed: 01/14/2023] Open
Abstract
Clinical observations have demonstrated a link between chronic pain and increased ischemic heart disease mortality, but the mechanisms remain elusive. Reactive aldehydes have recently been confirmed as a new player in pain pathologies, while our previous study demonstrated that reactive aldehydes (4-HNE) induced carbonyl stress contributing to myocardial ischemic intolerance. The aim of this study was to explore whether chronic pain increases susceptibility to myocardial ischemia/reperfusion (MI/R) injury and to investigate the underlying mechanisms focusing on toxic aldehyde and carbonyl stress. Methods: Chronic pain was induced by chronic compression of the dorsal root ganglion (CCD). After 2 weeks CCD, aldehyde dehydrogenase (ALDH2) KO or wild-type (WT) littermate mice were then subjected to in vivo MI/R. Results: In CCD-WT mice, heightened nociception paralleled circulating aldehyde (4-HNE) accumulation and cardiac protein carbonylation. Mechanistically, CCD-induced 4-HNE overload provoked cardiac Sirtuin 1 (SIRT1) carbonylative inactivation and inhibited Liver kinase B1 (LKB1) - AMP-activated protein kinase (LKB1-AMPK) interaction, which resulted in exacerbated MI/R injury and higher mortality compared with non-CCD WT mice. ALDH2 deficiency further aggravated CCD-induced susceptibility to MI/R injury. Exogenous 4-HNE exposure in peripheral tissue mimicked chronic pain-induced aldehyde overload, elicited sustained allodynia and increased MI/R injury. However, cardiac-specific ALDH2 upregulation by AAV9-cTNT-mediated gene delivery significantly ameliorated chronic pain-induced SIRT1 carbonylative inactivation and decreased MI/R injury (minor infarct size, less apoptosis, and improved cardiac function). Conclusion: Collectively, chronic pain-enhanced carbonyl stress promotes myocardial ischemic intolerance by SIRT1 carbonylative inactivation and impairment of LKB1-AMPK interaction. ALDH2 activation and prevention of protein carbonylation may be a potential therapeutic target for myocardial ischemic vulnerability in chronic pain patients. Our results newly provided overlapping cellular mechanisms of chronic pain and myocardial dysfunction interplay.
Collapse
|
29
|
Gal D, Sipido KR, Vandevelde W. Editorial highlights from Cardiovascular Research. Cardiovasc Res 2017; 113:e64-e68. [PMID: 29186440 DOI: 10.1093/cvr/cvx210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Diane Gal
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, Campus Gasthuisberg, KU Leuven, Belgium
| | - Karin R Sipido
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, Campus Gasthuisberg, KU Leuven, Belgium
| | - Wouter Vandevelde
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, Campus Gasthuisberg, KU Leuven, Belgium
| |
Collapse
|
30
|
Renaud-Gabardos E, Tatin F, Hantelys F, Lebas B, Calise D, Kunduzova O, Masri B, Pujol F, Sicard P, Valet P, Roncalli J, Chaufour X, Garmy-Susini B, Parini A, Prats AC. Therapeutic Benefit and Gene Network Regulation by Combined Gene Transfer of Apelin, FGF2, and SERCA2a into Ischemic Heart. Mol Ther 2017; 26:902-916. [PMID: 29249393 DOI: 10.1016/j.ymthe.2017.11.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/06/2017] [Accepted: 11/10/2017] [Indexed: 01/16/2023] Open
Abstract
Despite considerable advances in cardiovascular disease treatment, heart failure remains a public health challenge. In this context, gene therapy appears as an attractive approach, but clinical trials using single therapeutic molecules result in moderate benefit. With the objective of improving ischemic heart failure therapy, we designed a combined treatment, aimed to simultaneously stimulate angiogenesis, prevent cardiac remodeling, and restore contractile function. We have previously validated IRES-based vectors as powerful tools to co-express genes of interest. Mono- and multicistronic lentivectors expressing fibroblast growth factor 2 (angiogenesis), apelin (cardioprotection), and/or SERCA2a (contractile function) were produced and administrated by intramyocardial injection into a mouse model of myocardial infarction. Data reveal that combined treatment simultaneously improves vessel number, heart function parameters, and fibrosis prevention, due to FGF2, SERCA2a, and apelin, respectively. Furthermore, addition of SERCA2a in the combination decreases cardiomyocyte hypertrophy. Large-scale transcriptome analysis reveals that the triple treatment is the most efficient in restoring angiogenic balance as well as expression of genes involved in cardiac function and remodeling. Our study validates the concept of combined treatment of ischemic heart disease with apelin, FGF2, and SERCA2a and shows that such therapeutic benefit is mediated by a more effective recovery of gene network regulation.
Collapse
Affiliation(s)
| | - Florence Tatin
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Fransky Hantelys
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Benoît Lebas
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France; Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France
| | - Denis Calise
- UMS 006, Université de Toulouse, INSERM, 31432 Toulouse, France
| | - Oksana Kunduzova
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Bernard Masri
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Françoise Pujol
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Pierre Sicard
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Philippe Valet
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Jérôme Roncalli
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France; Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France
| | - Xavier Chaufour
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France; Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France
| | - Barbara Garmy-Susini
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Angelo Parini
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Anne-Catherine Prats
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France.
| |
Collapse
|
31
|
Heinig M, Adriaens ME, Schafer S, van Deutekom HWM, Lodder EM, Ware JS, Schneider V, Felkin LE, Creemers EE, Meder B, Katus HA, Rühle F, Stoll M, Cambien F, Villard E, Charron P, Varro A, Bishopric NH, George AL, Dos Remedios C, Moreno-Moral A, Pesce F, Bauerfeind A, Rüschendorf F, Rintisch C, Petretto E, Barton PJ, Cook SA, Pinto YM, Bezzina CR, Hubner N. Natural genetic variation of the cardiac transcriptome in non-diseased donors and patients with dilated cardiomyopathy. Genome Biol 2017; 18:170. [PMID: 28903782 PMCID: PMC5598015 DOI: 10.1186/s13059-017-1286-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 07/19/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Genetic variation is an important determinant of RNA transcription and splicing, which in turn contributes to variation in human traits, including cardiovascular diseases. RESULTS Here we report the first in-depth survey of heart transcriptome variation using RNA-sequencing in 97 patients with dilated cardiomyopathy and 108 non-diseased controls. We reveal extensive differences of gene expression and splicing between dilated cardiomyopathy patients and controls, affecting known as well as novel dilated cardiomyopathy genes. Moreover, we show a widespread effect of genetic variation on the regulation of transcription, isoform usage, and allele-specific expression. Systematic annotation of genome-wide association SNPs identifies 60 functional candidate genes for heart phenotypes, representing 20% of all published heart genome-wide association loci. Focusing on the dilated cardiomyopathy phenotype we found that eQTL variants are also enriched for dilated cardiomyopathy genome-wide association signals in two independent cohorts. CONCLUSIONS RNA transcription, splicing, and allele-specific expression are each important determinants of the dilated cardiomyopathy phenotype and are controlled by genetic factors. Our results represent a powerful resource for the field of cardiovascular genetics.
Collapse
Affiliation(s)
- Matthias Heinig
- Institute of Computational Biology, Helmholtz Zentrum München, München, Germany.,Department of Informatics, Technical University of Munich, Munich, Germany
| | - Michiel E Adriaens
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105AZ, The Netherlands.,Maastricht Centre for Systems Biology, Maastricht University, Maastricht, The Netherlands
| | - Sebastian Schafer
- National Heart Research Institute Singapore, National Heart Centre Singapore, 168752, Singapore, Singapore.,Division of Cardiovascular & Metabolic Disorders, Duke-National University of Singapore, 169857, Singapore, Singapore
| | - Hanneke W M van Deutekom
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105AZ, The Netherlands
| | - Elisabeth M Lodder
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105AZ, The Netherlands
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit at Royal Brompton and Harefield Hospitals and Imperial College London, London, UK.,Medical Research Council (MRC) London Institute of Medical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Valentin Schneider
- Cardiovascular and Metabolic Sciences, Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Leanne E Felkin
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit at Royal Brompton and Harefield Hospitals and Imperial College London, London, UK
| | - Esther E Creemers
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105AZ, The Netherlands
| | - Benjamin Meder
- Institute for Cardiomyopathies Heidelberg & Department of Cardiology, Angiology and Pneumology, University Heidelberg, Heidelberg, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung, Heidelberg/Mannheim, Germany
| | - Hugo A Katus
- Institute for Cardiomyopathies Heidelberg & Department of Cardiology, Angiology and Pneumology, University Heidelberg, Heidelberg, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung, Heidelberg/Mannheim, Germany
| | - Frank Rühle
- Institute of Human Genetics, Genetic Epidemiology, University of Münster, Münster, Germany
| | - Monika Stoll
- Institute of Human Genetics, Genetic Epidemiology, University of Münster, Münster, Germany.,Department of Biochemistry, Genetic Epidemiology and Statistical Genetics, CARIM School for Cardiovascular Diseases, Maastricht Center for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands
| | - François Cambien
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS 1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, F-75013, Paris, France.,ICAN Institute for Cardiometabolism and Nutrition, F-75013, Paris, France
| | - Eric Villard
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS 1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, F-75013, Paris, France.,ICAN Institute for Cardiometabolism and Nutrition, F-75013, Paris, France
| | - Philippe Charron
- ICAN Institute for Cardiometabolism and Nutrition, F-75013, Paris, France.,Université Versailles Saint Quentin, AP-HP, CESP, INSERM U1018, Hôpital Ambroise Paré, Boulogne-Billancourt, France
| | - Andras Varro
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Nanette H Bishopric
- Department of Medicine, University of Miami School of Medicine, Miami, FL, USA.,Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, FL, USA
| | - Alfred L George
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN, USA.,Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Cristobal Dos Remedios
- Sydney Heart Bank, Department of Anatomy, Bosch Institute, The University of Sydney, Sydney, Australia
| | - Aida Moreno-Moral
- Program in Cardiovascular and Metabolic Disorders, Center for Computational Biology, DUKE-NUS Medical School, Singapore, 169857, Singapore
| | - Francesco Pesce
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit at Royal Brompton and Harefield Hospitals and Imperial College London, London, UK
| | - Anja Bauerfeind
- Cardiovascular and Metabolic Sciences, Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Franz Rüschendorf
- Cardiovascular and Metabolic Sciences, Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Carola Rintisch
- Cardiovascular and Metabolic Sciences, Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Enrico Petretto
- Program in Cardiovascular and Metabolic Disorders, Center for Computational Biology, DUKE-NUS Medical School, Singapore, 169857, Singapore
| | - Paul J Barton
- National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit at Royal Brompton and Harefield Hospitals and Imperial College London, London, UK
| | - Stuart A Cook
- National Heart Research Institute Singapore, National Heart Centre Singapore, 168752, Singapore, Singapore.,Division of Cardiovascular & Metabolic Disorders, Duke-National University of Singapore, 169857, Singapore, Singapore.,National Heart and Lung Institute, Imperial College London, London, UK.,NIHR Cardiovascular Biomedical Research Unit at Royal Brompton and Harefield Hospitals and Imperial College London, London, UK.,Medical Research Council (MRC) London Institute of Medical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Yigal M Pinto
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105AZ, The Netherlands
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105AZ, The Netherlands.
| | - Norbert Hubner
- Cardiovascular and Metabolic Sciences, Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle-Str. 10, 13125, Berlin, Germany. .,Deutsches Zentrum für Herz-Kreislauf-Forschung, Heidelberg/Mannheim, Germany. .,Charité-Universitätsmedizin, Berlin, Germany. .,Deutsches Zentrum für Herz-Kreislauf-Forschung, Berlin, Germany.
| |
Collapse
|
32
|
Liu Y, Baumgardt SL, Fang J, Shi Y, Qiao S, Bosnjak ZJ, Vásquez-Vivar J, Xia Z, Warltier DC, Kersten JR, Ge ZD. Transgenic overexpression of GTP cyclohydrolase 1 in cardiomyocytes ameliorates post-infarction cardiac remodeling. Sci Rep 2017; 7:3093. [PMID: 28596578 PMCID: PMC5465102 DOI: 10.1038/s41598-017-03234-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022] Open
Abstract
GTP cyclohydrolase 1 (GCH1) and its product tetrahydrobiopterin play crucial roles in cardiovascular health and disease, yet the exact regulation and role of GCH1 in adverse cardiac remodeling after myocardial infarction are still enigmatic. Here we report that cardiac GCH1 is degraded in remodeled hearts after myocardial infarction, concomitant with increases in the thickness of interventricular septum, interstitial fibrosis, and phosphorylated p38 mitogen-activated protein kinase and decreases in left ventricular anterior wall thickness, cardiac contractility, tetrahydrobiopterin, the dimers of nitric oxide synthase, sarcoplasmic reticulum Ca2+ release, and the expression of sarcoplasmic reticulum Ca2+ handling proteins. Intriguingly, transgenic overexpression of GCH1 in cardiomyocytes reduces the thickness of interventricular septum and interstitial fibrosis and increases anterior wall thickness and cardiac contractility after infarction. Moreover, we show that GCH1 overexpression decreases phosphorylated p38 mitogen-activated protein kinase and elevates tetrahydrobiopterin levels, the dimerization and phosphorylation of neuronal nitric oxide synthase, sarcoplasmic reticulum Ca2+ release, and sarcoplasmic reticulum Ca2+ handling proteins in post-infarction remodeled hearts. Our results indicate that the pivotal role of GCH1 overexpression in post-infarction cardiac remodeling is attributable to preservation of neuronal nitric oxide synthase and sarcoplasmic reticulum Ca2+ handling proteins, and identify a new therapeutic target for cardiac remodeling after infarction.
Collapse
Affiliation(s)
- Yanan Liu
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.,Department of Medicine, Columbia University, 630 W. 168th Street, New York, New York, 10032, USA
| | - Shelley L Baumgardt
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Yang Shi
- Aurora Research Institute, Aurora Health Care, 750 W. Virginia Street, Milwaukee, Wisconsin, 53234, USA
| | - Shigang Qiao
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zeljko J Bosnjak
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.,Department of Physiology, Medical College of Wiscosin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Jeannette Vásquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, People's Republic of China
| | - David C Warltier
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Judy R Kersten
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA
| | - Zhi-Dong Ge
- Departments of Anesthesiology, Medical College of Wisconsin, Milwaukee, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226, USA.
| |
Collapse
|
33
|
Favero G, Franceschetti L, Buffoli B, Moghadasian MH, Reiter RJ, Rodella LF, Rezzani R. Melatonin: Protection against age-related cardiac pathology. Ageing Res Rev 2017; 35:336-349. [PMID: 27884595 DOI: 10.1016/j.arr.2016.11.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/04/2016] [Accepted: 11/18/2016] [Indexed: 12/14/2022]
Abstract
Aging is a complex and progressive process that involves physiological and metabolic deterioration in every organ and system. Cardiovascular diseases are one of the most common causes of mortality and morbidity among elderly subjects worldwide. Most age-related cardiovascular disorders can be influenced by modifiable behaviours such as a healthy diet rich in fruit and vegetables, avoidance of smoking, increased physical activity and reduced stress. The role of diet in prevention of various disorders is a well-established factor, which has an even more important role in the geriatric population. Melatonin, an indoleamine with multiple actions including antioxidant properties, has been identified in a very large number of plant species, including edible plant products and medical herbs. Among products where melatonin has been identified include wine, olive oil, tomato, beer, and others. Interestingly, consumed melatonin in plant foods or melatonin supplementation may promote health benefits by virtue of its multiple properties and it may counteract pathological conditions also related to cardiovascular disorders, carcinogenesis, neurological diseases and aging. In the present review, we summarized melatonin effects against age-related cardiac alterations and abnormalities with a special focus on heart ischemia/reperfusion (IR) injury and myocardial infarction.
Collapse
Affiliation(s)
- Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Lorenzo Franceschetti
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Barbara Buffoli
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Mohammed H Moghadasian
- Department of Human Nutritional Sciences, University of Manitoba and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Luigi F Rodella
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| |
Collapse
|
34
|
Arora D, Sharma PK, Siddiqui MH, Shukla Y. Necroptosis: Modules and molecular switches with therapeutic implications. Biochimie 2017; 137:35-45. [PMID: 28263777 DOI: 10.1016/j.biochi.2017.02.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 02/07/2017] [Accepted: 02/27/2017] [Indexed: 12/24/2022]
Abstract
Among the various programmed cell death (PCD) pathways, "Necroptosis" has gained much importance as a novel paradigm of cell death. This pathway has emerged as a backup mechanism when physiologically conserved PCD (apoptosis) is non-functional either genetically or pathogenically. The expanding spectrum of necroptosis from physiological development to diverse patho-physiological disorders, including xenobiotics-mediated toxicity has now grabbed the attention worldwide. The efficient role of necroptosis regulators in disease development and management are under constant examination. In fact, few regulators (e.g. MLKL) have already paved their way towards clinical trials and others are in queue. In this review, emphasis has been paid to the various contributing factors and molecular switches that can regulate necroptosis. Here we linked the overview of current knowledge of this enigmatic signaling with magnitude of therapeutics that may underpin the opportunities for novel therapeutic approaches to suppress the pathogenesis of necroptosis-driven disorders.
Collapse
Affiliation(s)
- Deepika Arora
- Environmental Carcinogenesis & Proteomics Laboratory, Food, Drug & Chemical Toxicology Group, VishvigyanBhawan 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, 226026, Uttar Pradesh, India
| | - Pradeep Kumar Sharma
- Environmental Carcinogenesis & Proteomics Laboratory, Food, Drug & Chemical Toxicology Group, VishvigyanBhawan 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
| | - Mohammed Haris Siddiqui
- Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, 226026, Uttar Pradesh, India
| | - Yogeshwer Shukla
- Environmental Carcinogenesis & Proteomics Laboratory, Food, Drug & Chemical Toxicology Group, VishvigyanBhawan 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India.
| |
Collapse
|
35
|
Plasma miRNA Profiles in Pregnant Women Predict Infant Outcomes following Prenatal Alcohol Exposure. PLoS One 2016; 11:e0165081. [PMID: 27828986 PMCID: PMC5102408 DOI: 10.1371/journal.pone.0165081] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 10/05/2016] [Indexed: 12/04/2022] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are difficult to diagnose since many heavily exposed infants, at risk for intellectual disability, do not exhibit craniofacial dysmorphology or growth deficits. Consequently, there is a need for biomarkers that predict disability. In both animal models and human studies, alcohol exposure during pregnancy resulted in significant alterations in circulating microRNAs (miRNAs) in maternal blood. In the current study, we asked if changes in plasma miRNAs in alcohol-exposed pregnant mothers, either alone or in conjunction with other clinical variables, could predict infant outcomes. Sixty-eight pregnant women at two perinatal care clinics in western Ukraine were recruited into the study. Detailed health and alcohol consumption histories, and 2nd and 3rd trimester blood samples were obtained. Birth cohort infants were assessed by a geneticist and classified as unexposed (UE), heavily prenatally exposed and affected (HEa) or heavily exposed but apparently unaffected (HEua). MiRNAs were assessed in plasma samples using qRT-PCR arrays. ANOVA models identified 11 miRNAs that were all significantly elevated in maternal plasma from the HEa group relative to HEua and UE groups. In a random forest analysis classification model, a combination of high variance miRNAs, smoking history and socioeconomic status classified membership in HEa and UE groups, with a misclassification rate of 13%. The RFA model also classified 17% of the HEua group as UE-like, whereas 83% were HEa-like, at least at one stage of pregnancy. Collectively our data indicate that maternal plasma miRNAs predict infant outcomes, and may be useful to classify difficult-to-diagnose FASD subpopulations.
Collapse
|
36
|
Studies on the role of apoptosis after transient myocardial ischemia: genetic deletion of the executioner caspases-3 and -7 does not limit infarct size and ventricular remodeling. Basic Res Cardiol 2016; 111:18. [PMID: 26924441 DOI: 10.1007/s00395-016-0537-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 02/02/2016] [Indexed: 12/13/2022]
Abstract
Although it is widely accepted that apoptosis may contribute to cell death in myocardial infarction, experimental evidence suggests that adult cardiomyocytes repress the expression of the caspase-dependent apoptotic pathway. The aim of this study was to analyze the contribution of caspase-mediated apoptosis to myocardial ischemia-reperfusion injury. Cardiac-specific caspase-3 deficient/full caspase-7-deficient mice (Casp3/7DKO) and wild type control mice (WT) were subjected to in situ ischemia by left anterior coronary artery ligation for 45 min followed by 24 h or 28 days of reperfusion. Heart function was assessed using M-mode echocardiography. Deletion of caspases did not modify neither infarct size determined by triphenyltetrazolium staining after 24 h of reperfusion (40.0 ± 5.1 % in WT vs. 36.2 ± 3.6 % in Casp3/7DKO), nor the scar area measured by pricosirius red staining after 28 days of reperfusion (41.1 ± 5.4 % in WT vs. 44.6 ± 8.7 % in Casp3/7DKO). Morphometric and echocardiographic studies performed 28 days after the ischemic insult revealed left ventricular dilation and severe cardiac dysfunction without statistically significant differences between WT and Casp3/7DKO groups. These data demonstrate that the executioner caspases-3 and -7 do not significantly contribute to cardiomyocyte death induced by transient coronary occlusion and provide the first evidence obtained in an in vivo model that argues against a relevant role of apoptosis through the canonical caspase pathway in this context.
Collapse
|