1
|
Liu H, Liu J, Guan X, Zhao Z, Cheng P, Chen H, Jiang Z, Wang X. Titin gene mutations enhance radiotherapy efficacy via modulation of tumour immune microenvironment in rectum adenocarcinoma. Clin Transl Med 2025; 15:e70123. [PMID: 39748197 PMCID: PMC11695211 DOI: 10.1002/ctm2.70123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/11/2024] [Accepted: 11/24/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE This study investigates the impact of Titin (TTN) gene mutations on radiotherapy sensitivity in rectum adenocarcinoma (READ) by examining changes in the tumour immune microenvironment. METHODS Data on gene expression and mutations in READ were obtained from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. Bioinformatics analysis explored the correlation between TTN mutations and immune cell infiltration. In vitro, lentiviral vectors were used to assess TTN mutations' effects on ANKRD1 expression in two READ cell lines. ANKRD1 was overexpressed, and clonogenic assays evaluated radiotherapy sensitivity. Flow cytometry, immunofluorescence, and comet assays examined mutations' impact on cell cycle, apoptosis, and DNA damage response (DDR). An in vivo mouse model and formalin-fixed paraffin-embedded samples from locally advanced rectal cancer (LARC) patients before and after radiotherapy were analyzed, followed by prognostic evaluation. RESULTS Bioinformatics revealed that TTN mutations increase radiation sensitivity in LARC by slowing cell proliferation, promoting apoptosis, and reducing DDR. TTN mutations also inhibit ANKRD1 expression via JUN disruption and enhance CD4/CD8 T-cell infiltration, improving anti-tumour immunity and outcomes. Observations from the clinical study showed a substantial decline in ANKRD1 expression levels alongside a notable surge in the counts of CD4+ and CD8+ T cells after undergoing radiotherapy. Patients with TTN mutations, low ANKRD1 expression, and high densities of CD4+ and CD8+ T cells had longer 3-year disease-free survival in READ. CONCLUSION Our findings reveal that TTN mutations can serve as biomarkers for enhanced radiotherapy sensitivity in READ. By altering the tumour's immune microenvironment, these mutations may provide a novel target for personalized radiotherapy strategies, potentially improving therapeutic outcomes in patients with READ. HIGHLIGHTS The association between TTN mutations and tumour mutation burden, as well as immune cell infiltration in READ, is examined. TTN mutations enhance the radiation sensitivity of READ cells and weaken DNA damage repair in response to radiation. TTN mutations increase the radiation sensitivity of READ cells by inhibiting ANKRD1. The infiltration of CD8+ and CD4+ T cells induced by TTN mutations is essential for anti-tumour immunity. TTN mutations serve as a biomarker for the pathological response to preoperative radiotherapy in READ.
Collapse
Affiliation(s)
- Hengchang Liu
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jialiang Liu
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xu Guan
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhixun Zhao
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Pu Cheng
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Haipeng Chen
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zheng Jiang
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xishan Wang
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Chen S, Sun J, Wen W, Chen Z, Yu Z. Integrative multi-omics summary-based mendelian randomization identifies key oxidative stress-related genes as therapeutic targets for atrial fibrillation and flutter. Front Genet 2024; 15:1447872. [PMID: 39359474 PMCID: PMC11445139 DOI: 10.3389/fgene.2024.1447872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Background Atrial fibrillation (AF) is a prevalent cardiac arrhythmia associated with substantial morbidity and mortality. Oxidative stress (OS) has been implicated in the pathogenesis of AF, suggesting that targeting OS-related genes could offer novel therapeutic opportunities. This study aimed to identify causal OS-related genes contributing to AF through a comprehensive multi-omics Summary-based Mendelian Randomization (SMR) approach. Methods This study integrated data from genome-wide association studies (GWAS) with methylation quantitative trait loci (mQTL), expression QTL (eQTL), and protein QTL (pQTL) to explore the relationships between oxidative stress-related (OS-related) genes and AF risk. Genes associated with oxidative stress and AF were obtained from the Nielsen et al. study (discovery) and the FinnGen study (replication). The SMR analysis and HEIDI test were utilized to assess causal associations, followed by Bayesian co-localization analysis (PPH4 > 0.5) to confirm shared causal variants. Multi-omics data were employed to analyze the associations within mQTL-eQTL pathways. A two-sample MR analysis was conducted for sensitivity verification. The significance of findings was determined using a false discovery rate (FDR) < 0.05 and p_HEIDI > 0.01. Results At the DNA methylation level, 19 CpG sites near 7 unique genes were found to have causal effects on AF and strong co-localization evidence support (PPH4 > 0.70). At the gene expression level, six oxidative stress-related genes from eQTLGen and three from GTEx (v8), including TNFSF10, CDKN1A, ALOX15, TTN, PTK2, ALB, KCNJ5, and CASQ2, were found to have causal effects on AF in the sensitivity and co-localization analyses (PPH4 > 0.50). At the circulating protein level, both ALAD (OR 0.898, 95% CI 0.845-0.954, PPH4 = 0.67) and APOH (OR 0.896, 95% CI 0.844-0.952, PPH4 = 0.93) were associated with a lower risk of AF, and APOH was validated in the replication group. After integrating the multi-omics data between mQTL and eQTL, we identified two oxidative stress-related genes, TTN and CASQ2. The methylation of cg09915519 and cg10087519 in TTN was associated with higher expression of TTN and a lower risk of AF, which aligns with the negative effect of TTN gene expression on AF risk. TTN may play a protective role in AF. Conclusion This study identified several OS-related genes, particularly TTN, as having causal roles in AF, which were verified across three-omics pathways. The findings underscore the importance of these genes in AF pathogenesis and highlight their potential as therapeutic targets. The integration of multi-omics data provides a comprehensive understanding of the molecular mechanisms underlying AF, paving the way for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Shijian Chen
- Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
| | - Junlong Sun
- Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
| | - Wen Wen
- Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
| | - Zhenfeng Chen
- Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
| | - Ziheng Yu
- Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
| |
Collapse
|
3
|
Risato G, Brañas Casas R, Cason M, Bueno Marinas M, Pinci S, De Gaspari M, Visentin S, Rizzo S, Thiene G, Basso C, Pilichou K, Tiso N, Celeghin R. In Vivo Approaches to Understand Arrhythmogenic Cardiomyopathy: Perspectives on Animal Models. Cells 2024; 13:1264. [PMID: 39120296 PMCID: PMC11311808 DOI: 10.3390/cells13151264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a hereditary cardiac disorder characterized by the gradual replacement of cardiomyocytes with fibrous and adipose tissue, leading to ventricular wall thinning, chamber dilation, arrhythmias, and sudden cardiac death. Despite advances in treatment, disease management remains challenging. Animal models, particularly mice and zebrafish, have become invaluable tools for understanding AC's pathophysiology and testing potential therapies. Mice models, although useful for scientific research, cannot fully replicate the complexity of the human AC. However, they have provided valuable insights into gene involvement, signalling pathways, and disease progression. Zebrafish offer a promising alternative to mammalian models, despite the phylogenetic distance, due to their economic and genetic advantages. By combining animal models with in vitro studies, researchers can comprehensively understand AC, paving the way for more effective treatments and interventions for patients and improving their quality of life and prognosis.
Collapse
Affiliation(s)
- Giovanni Risato
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
- Department of Biology, University of Padua, I-35131 Padua, Italy;
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | | | - Marco Cason
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Maria Bueno Marinas
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Serena Pinci
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Monica De Gaspari
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Silvia Visentin
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | - Stefania Rizzo
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Natascia Tiso
- Department of Biology, University of Padua, I-35131 Padua, Italy;
| | - Rudy Celeghin
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| |
Collapse
|
4
|
Sun W, Liu X, Song L, Tao L, Lai K, Jiang H, Xiao H. The TTN p. Tyr4418Ter mutation causes cardiomyopathy in human and mice. PLoS One 2024; 19:e0296802. [PMID: 38381767 PMCID: PMC10880961 DOI: 10.1371/journal.pone.0296802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/19/2023] [Indexed: 02/23/2024] Open
Abstract
OBJECTIVE To generate a mouse model carrying TTNtv Y4370* simulating the newly discovered human heterozygous nonsense TTNtv c.13254T>G (p.Tyr4418Ter) to supplement and improve the functional evidence of pathogenic mutation TTNtv c.13254T>G on the pathogenic type of dilated cardiomyopathy. METHODS We generated 4 mice carrying TTNtv p. Y4370* through CRISPR/Cas-mediated genome engineering. Monthly serological detection, bimonthly echocardiography, and histology evaluation were carried out to observe and compare alterations of cardiac structure and function between 4 TTN+/- mice and 4 wild-type (WT) mice. RESULTS For the two-month-old TTN+/- mice, serum glutamic-oxalacetic transaminase (AST), lactic dehydrogenase (LDH), and creatine kinase (CK) were significantly increased, the diastolic Left Ventricular Systolic Anterior Wall (LVAW), and the LV mass markedly rose, with the left ventricular volume displaying an increasing trend and Ejection Fraction (EF) and Fractional Shortening (FS) showing a decreasing trend. Besides, the histological evaluation showed that cardiac fibrosis level and positive rate of cardiac mast cell of TTN+/- mice were obviously increased compared with WT mice. CONCLUSIONS TTNtv Y4370* could lead to cardiac structure and function alterations in mice, supplementing the evidence of TTNtv c.13254T>G pathogenicity in human.
Collapse
Affiliation(s)
- Wenqiang Sun
- Division of Cardiac Surgery & Wuhan Clinical Research Center for Cardiomyopathy, Wuhan Asia Heart Hospital Affiliated with Wuhan University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Xiaohui Liu
- Department of Clinical Laboratory, Wuhan Asia Heart Hospital Affiliated with the Wuhan University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Laichun Song
- Division of Cardiac Surgery & Wuhan Clinical Research Center for Cardiomyopathy, Wuhan Asia Heart Hospital Affiliated with Wuhan University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Liang Tao
- Division of Cardiac Surgery & Wuhan Clinical Research Center for Cardiomyopathy, Wuhan Asia Heart Hospital Affiliated with Wuhan University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Kaisheng Lai
- Department of Science Research Centre, BestNovo (Beijing) Medical Laboratory, Beijing, P.R. China
| | - Hui Jiang
- Department of Science Research Centre, BestNovo (Beijing) Medical Laboratory, Beijing, P.R. China
| | - Hongyan Xiao
- Division of Cardiac Surgery & Wuhan Clinical Research Center for Cardiomyopathy, Wuhan Asia Heart Hospital Affiliated with Wuhan University of Science and Technology, Wuhan, Hubei, P.R. China
| |
Collapse
|
5
|
Marcello M, Cetrangolo V, Morotti I, Squarci C, Caremani M, Reconditi M, Savarese M, Bianco P, Piazzesi G, Lombardi V, Udd B, Conte I, Nigro V, Linari M. Sarcomere level mechanics of the fast skeletal muscle of the medaka fish larva. Am J Physiol Cell Physiol 2024; 326:C632-C644. [PMID: 38145303 DOI: 10.1152/ajpcell.00530.2023] [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: 10/12/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
The medaka fish (Oryzias latipes) is a vertebrate model used in developmental biology and genetics. Here we explore its suitability as a model for investigating the molecular mechanisms of human myopathies caused by mutations in sarcomeric proteins. To this end, the relevant mechanical parameters of the intact skeletal muscle of wild-type medaka are determined using the transparent tail at larval stage 40. Tails were mounted at sarcomere length of 2.1 μm in a thermoregulated trough containing physiological solution. Tetanic contractions were elicited at physiological temperature (10°C-30°C) by electrical stimulation, and sarcomere length changes were recorded with nanometer-microsecond resolution during both isometric and isotonic contractions with a striation follower. The force output has been normalized for the actual fraction of the cross section of the tail occupied by the myofilament lattice, as established with transmission electron microscopy (TEM), and then for the actual density of myofilaments, as established with X-ray diffraction. Under these conditions, the mechanical performance of the contracting muscle of the wild-type larva can be defined at the level of the half-thick filament, where ∼300 myosin motors work in parallel as a collective motor, allowing a detailed comparison with the established performance of the skeletal muscle of different vertebrates. The results of this study point out that the medaka fish larva is a suitable model for the investigation of the genotype/phenotype correlations and therapeutic possibilities in skeletal muscle diseases caused by mutations in sarcomeric proteins.NEW & NOTEWORTHY The suitability of the medaka fish as a model for investigating the molecular mechanisms of human myopathies caused by mutations of sarcomeric proteins is tested by combining structural analysis and sarcomere-level mechanics of the skeletal muscle of the tail of medaka larva. The mechanical performance of the medaka muscle, scaled at the level of the myosin-containing thick filament, together with its reduced genome duplication makes this model unique for investigations of the genotype/phenotype correlations in human myopathies.
Collapse
Affiliation(s)
| | - Viviana Cetrangolo
- PhysioLab, University of Florence, Florence, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | | | | | | | | | - Marco Savarese
- Folkhälsan Research Center, Helsinki University, Helsinki, Finland
| | | | | | | | - Bjarne Udd
- Folkhälsan Research Center, Helsinki University, Helsinki, Finland
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Department of Biology, University of Naples "Federico II", Naples, Italy
| | - Vincenzo Nigro
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Department of Precision Medicine, University of Campania, Naples, Italy
| | - Marco Linari
- PhysioLab, University of Florence, Florence, Italy
| |
Collapse
|
6
|
Kellermayer D, Tordai H, Kiss B, Török G, Péter DM, Sayour AA, Pólos M, Hartyánszky I, Szilveszter B, Labeit S, Gángó A, Bedics G, Bödör C, Radovits T, Merkely B, Kellermayer MS. Truncated titin is structurally integrated into the human dilated cardiomyopathic sarcomere. J Clin Invest 2024; 134:e169753. [PMID: 37962957 PMCID: PMC10763722 DOI: 10.1172/jci169753] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 11/08/2023] [Indexed: 11/16/2023] Open
Abstract
Heterozygous (HET) truncating variant mutations in the TTN gene (TTNtvs), encoding the giant titin protein, are the most common genetic cause of dilated cardiomyopathy (DCM). However, the molecular mechanisms by which TTNtv mutations induce DCM are controversial. Here, we studied 127 clinically identified DCM human cardiac samples with next-generation sequencing (NGS), high-resolution gel electrophoresis, Western blot analysis, and super-resolution microscopy in order to dissect the structural and functional consequences of TTNtv mutations. The occurrence of TTNtv was found to be 15% in the DCM cohort. Truncated titin proteins matching, by molecular weight, the gene sequence predictions were detected in the majority of the TTNtv+ samples. Full-length titin was reduced in TTNtv+ compared with TTNtv- samples. Proteomics analysis of washed myofibrils and stimulated emission depletion (STED) super-resolution microscopy of myocardial sarcomeres labeled with sequence-specific anti-titin antibodies revealed that truncated titin was structurally integrated into the sarcomere. Sarcomere length-dependent anti-titin epitope position, shape, and intensity analyses pointed at possible structural defects in the I/A junction and the M-band of TTNtv+ sarcomeres, which probably contribute, possibly via faulty mechanosensor function, to the development of manifest DCM.
Collapse
Affiliation(s)
- Dalma Kellermayer
- Heart and Vascular Center
- Department of Biophysics and Radiation Biology, and
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | - Balázs Kiss
- Department of Biophysics and Radiation Biology, and
| | - György Török
- Department of Biophysics and Radiation Biology, and
| | | | | | | | | | | | - Siegfried Labeit
- DZHK Partnersite Mannheim-Heidelberg, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ambrus Gángó
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gábor Bedics
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Csaba Bödör
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | | | | |
Collapse
|
7
|
Koslow M, Mondaca-Ruff D, Xu X. Transcriptome studies of inherited dilated cardiomyopathies. Mamm Genome 2023; 34:312-322. [PMID: 36749382 PMCID: PMC10426000 DOI: 10.1007/s00335-023-09978-z] [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: 07/25/2022] [Accepted: 01/16/2023] [Indexed: 02/08/2023]
Abstract
Dilated cardiomyopathy (DCM) is a group of heart muscle diseases that often lead to heart failure, with more than 50 causative genes have being linked to DCM. The heterogenous nature of the inherited DCMs suggest the need of precision medicine. Consistent with this emerging concept, transcriptome studies in human patients with DCM indicated distinct molecular signature for DCMs of different genetic etiology. To facilitate this line of research, we reviewed the status of transcriptome studies of inherited DCMs by focusing on three predominant DCM causative genes, TTN, LMNA, and BAG3. Besides studies in human patients, we summarized transcriptomic analysis of these inherited DCMs in a variety of model systems ranging from iPSCs to rodents and zebrafish. We concluded that the RNA-seq technology is a powerful genomic tool that has already led to the discovery of new modifying genes, signaling pathways, and related therapeutic avenues. We also pointed out that both temporal (different pathological stages) and spatial (different cell types) information need to be considered for future transcriptome studies. While an important bottle neck is the low throughput in experimentally testing differentially expressed genes, new technologies in efficient animal models such as zebrafish starts to be developed. It is anticipated that the RNA-seq technology will continue to uncover both unique and common pathological events, aiding the development of precision medicine for inherited DCMs.
Collapse
Affiliation(s)
- Matthew Koslow
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - David Mondaca-Ruff
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
8
|
Zheng K, Lou MN. [Recent studies on dilated cardiomyopathy caused by TTN mutations in children]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:217-222. [PMID: 36854701 PMCID: PMC9979384 DOI: 10.7499/j.issn.1008-8830.2208163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/15/2022] [Indexed: 03/02/2023]
Abstract
The mutations of TTN gene that encodes titin are the most common mutation type among the genetic causes of dilated cardiomyopathy (DCM). This article reviews the worldwide studies on potential molecular pathogenesis (transcription, post-translational modification, etc.), clinical phenotypes, and gene therapies of pediatric DCM caused by TTN mutations, with the hope of providing a reference for the precision treatment of pediatric DCM caused by TTN mutations.
Collapse
Affiliation(s)
- Kui Zheng
- Department of Cardiology, Hebei Children's Hospital/Hebei Provincial Key Laboratory of Pediatric Cardiovascular Disease, Shijiazhuang 050031, China
| | - Mei-Na Lou
- Department of Cardiology, Hebei Children's Hospital/Hebei Provincial Key Laboratory of Pediatric Cardiovascular Disease, Shijiazhuang 050031, China
| |
Collapse
|
9
|
Qi Y, Ji X, Ding H, Wang Y, Liu X, Zhang Y, Yin A. A spectrum of clinical severity of recessive titinopathies in prenatal. Front Genet 2023; 13:1064474. [PMID: 36761691 PMCID: PMC9907677 DOI: 10.3389/fgene.2022.1064474] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/23/2022] [Indexed: 01/26/2023] Open
Abstract
Variants in TTN are associated with a broad range of clinical phenotypes, from dominant adult-onset dilated cardiomyopathy to recessive infantile-onset myopathy. However, few foetal cases have been reported for multiple reasons. Next-generation sequencing has facilitated the prenatal identification of a growing number of suspected titinopathy variants. We investigated six affected foetuses from three families, completed the intrauterine course of the serial phenotypic spectrum of TTN, and discussed the genotype-phenotype correlations from a broader perspective. The recognizable prenatal feature onset at the second trimester was started with reduced movement, then contracture 3-6 weeks later, followed with/without hydrops, finally at late pregnancy was accompanied with polyhydramnio (major) or oligohydramnios. Two cases with typical arthrogryposis-hydrops sequences identified a meta-only transcript variant c.36203-1G>T. Deleterious transcriptional consequences of the substitution were verified by minigene splicing analysis. Case 3 identified a homozygous splicing variant in the constitutively expressed Z-disc. It presented a milder phenotype than expected, which was presumably saved by the isoform of corons. A summary of the foetal-onset titinopathy cases implied that variants in TTN present with a series of signs and a spectrum of clinical severity, which followed the dosage/positional effect; the meta-only transcript allele involvement may be a prerequisite for the development of fatal hydrops.
Collapse
Affiliation(s)
- Yiming Qi
- Prenatal Diagnosis Centre, Guangdong Women and Children Hospital, Guangzhou, China,Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, China
| | - Xueqi Ji
- Prenatal Diagnosis Centre, Guangdong Women and Children Hospital, Guangzhou, China,Guangzhou Medical University, Guangzhou, China
| | - Hongke Ding
- Prenatal Diagnosis Centre, Guangdong Women and Children Hospital, Guangzhou, China,Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, China
| | - Yunan Wang
- Prenatal Diagnosis Centre, Guangdong Women and Children Hospital, Guangzhou, China,Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, China
| | | | - Yan Zhang
- Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, China
| | - Aihua Yin
- Prenatal Diagnosis Centre, Guangdong Women and Children Hospital, Guangzhou, China,Maternal and Children Metabolic-Genetic Key Laboratory, Guangdong Women and Children Hospital, Guangzhou, China,*Correspondence: Aihua Yin,
| |
Collapse
|
10
|
Kim YG, Ha C, Shin S, Park JH, Jang JH, Kim JW. Enrichment of titin-truncating variants in exon 327 in dilated cardiomyopathy and its relevance to reduced nonsense-mediated mRNA decay efficiency. Front Genet 2023; 13:1087359. [PMID: 36685919 PMCID: PMC9845391 DOI: 10.3389/fgene.2022.1087359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/07/2022] [Indexed: 01/05/2023] Open
Abstract
Titin truncating variants (TTNtvs) are the most common genetic cause of dilated cardiomyopathy (DCM). Among four regions of titin, A-band enrichment of DCM-causing TTNtvs is widely accepted but the underlying mechanism is still unknown. Meanwhile, few reports have identified exon 327 as a highly mutated A-band exon but the degree of exon 327 enrichment has not been quantitatively investigated. To find the real hotspot of DCM-causing TTNtvs, we aimed to reassess the degree of TTNtv enrichment in known titin regions and in exon 327, separately. In addition, we tried to explain exon 327 clustering in terms of nonsense-mediated mRNA decay (NMD) efficiency and a dominant negative mechanism recently proposed. Research papers focusing on TTNtvs found in patients with DCM were collected. A total of 612 patients with TTNtv-realated DCM were obtained from 10 studies. In the four regions of TTN and exon 327, the degree of TTNtvs enrichment was calculated in a way that the effect of distribution of highly expressed exons was normalized. As a result, exon 327 was the only region that showed significant enrichment for DCM-related TTNtv (p < .001). On the other hand, other A-band exons had almost the same number of TTNtv of random distribution. A review of RNAseq data revealed that the median allelic imbalance deviation of exon 327 TTNtvs was .04, indicating almost zero NMD. From these findings, we propose that the widely accepted A-band enrichment of DCM-related TTNtv is mostly attributable to exon 327 enrichment. In addition, based on the recently demonstrated dominant negative mechanism, the extremely low NMD efficiency seems to contribute to exon 327 enrichment.
Collapse
Affiliation(s)
- Young-gon Kim
- Samsung Medical Center, Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Changhee Ha
- Samsung Medical Center, Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sunghwan Shin
- Samsung Medical Center, Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jong-ho Park
- Clinical Genomics Center, Samsung Medical Center, Seoul, South Korea
| | - Ja-Hyun Jang
- Samsung Medical Center, Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jong-Won Kim
- Samsung Medical Center, Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, South Korea,Clinical Genomics Center, Samsung Medical Center, Seoul, South Korea,*Correspondence: Jong-Won Kim,
| |
Collapse
|
11
|
Ghasemi S, Mahdavi M, Maleki M, Salahshourifar I, Kalayinia S. A novel likely pathogenic variant in the FBXO32 gene associated with dilated cardiomyopathy according to whole‑exome sequencing. BMC Med Genomics 2022; 15:234. [PMID: 36344977 PMCID: PMC9641816 DOI: 10.1186/s12920-022-01388-5] [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: 07/06/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Background Familial dilated cardiomyopathy (DCM) is a genetic heart disorder characterized by progressive heart failure and sudden cardiac death. Over 250 genes have been reported in association with DCM; nonetheless, the genetic cause of most DCM patients has been unknown. The goal of the present study was to determine the genetic etiology of familial DCM in an Iranian family. Methods Whole-exome sequencing was performed to identify the underlying variants in an Iranian consanguineous family with DCM. The presence of the candidate variant was confirmed and screened in available relatives by PCR and Sanger sequencing. The pathogenic effect of the candidate variant was assessed by bioinformatics analysis, homology modeling, and docking. Results One novel likely pathogenic deletion, c.884_886del: p.Lys295del, in F-box only protein 32 (muscle-specific ubiquitin-E3 ligase protein; FBXO32) was identified. Based on bioinformatics and modeling analysis, c.884_886del was the most probable cause of DCM in the studied family. Conclusions Our findings indicate that variants in FBXO32 play a role in recessive DCM. Variants in FBXO32 may disturb the degradation of target proteins in the ubiquitin–proteasome system and lead to severe DCM. We suggest considering this gene variants in patients with recessively inherited DCM.
Collapse
|
12
|
Marcello M, Cetrangolo V, Savarese M, Udd B. Use of animal models to understand titin physiology and pathology. J Cell Mol Med 2022; 26:5103-5112. [PMID: 36065969 PMCID: PMC9575118 DOI: 10.1111/jcmm.17533] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 12/01/2022] Open
Abstract
In recent years, increasing attention has been paid to titin (TTN) and its mutations. Heterozygous TTN truncating variants (TTNtv) increase the risk of a cardiomyopathy. At the same time, TTNtv and few missense variants have been identified in patients with mainly recessive skeletal muscle diseases. The pathogenic mechanisms underlying titin‐related diseases are still partly unknown. Similarly, the titin mechanical and functional role in the muscle contraction are far from being exhaustively clarified. In the last few years, several animal models carrying variants in the titin gene have been developed and characterized to study the structural and mechanical properties of specific titin domains or to mimic patients' mutations. This review describes the main animal models so far characterized, including eight mice models and three fish models (Medaka and Zebrafish) and discusses the useful insights provided by a thorough characterization of the cell‐, tissue‐ and organism‐phenotypes in these models.
Collapse
Affiliation(s)
| | | | - Marco Savarese
- Folkhälsan Research Center, Helsinki, Finland.,Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Bjarne Udd
- Folkhälsan Research Center, Helsinki, Finland.,Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Department of Neurology, Vaasa Central Hospital, Vaasa, Finland
| |
Collapse
|
13
|
Katraki-Pavlou S, Kastana P, Bousis D, Ntenekou D, Varela A, Davos CH, Nikou S, Papadaki E, Tsigkas G, Athanasiadis E, Herradon G, Mikelis CM, Beis D, Papadimitriou E. Protein tyrosine phosphatase receptor zeta 1 deletion triggers defective heart morphogenesis in mice and zebrafish. Am J Physiol Heart Circ Physiol 2021; 322:H8-H24. [PMID: 34767486 PMCID: PMC8754060 DOI: 10.1152/ajpheart.00400.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein tyrosine phosphatase receptor-ζ1 (PTPRZ1) is a transmembrane
tyrosine phosphatase receptor highly expressed in embryonic stem cells. In the
present work, gene expression analyses of Ptprz1−/− and Ptprz1+/+ mice endothelial cells and hearts pointed to
an unidentified role of PTPRZ1 in heart development through the regulation of
heart-specific transcription factor genes. Echocardiography analysis in mice
identified that both systolic and diastolic functions are affected in Ptprz1−/− compared with Ptprz1+/+ hearts, based on a dilated left
ventricular (LV) cavity, decreased ejection fraction and fraction shortening,
and increased angiogenesis in Ptprz1−/−
hearts, with no signs of cardiac hypertrophy. A zebrafish ptprz1−/− knockout was also generated and exhibited
misregulated expression of developmental cardiac markers, bradycardia, and
defective heart morphogenesis characterized by enlarged ventricles and defected
contractility. A selective PTPRZ1 tyrosine phosphatase inhibitor affected
zebrafish heart development and function in a way like what is observed in the
ptprz1−/− zebrafish. The same
inhibitor had no effect in the function of the adult zebrafish heart, suggesting
that PTPRZ1 is not important for the adult heart function, in line with data
from the human cell atlas showing very low to negligible PTPRZ1 expression in
the adult human heart. However, in line with the animal models, Ptprz1 was expressed in many different cell types in
the human fetal heart, such as valvar, fibroblast-like, cardiomyocytes, and
endothelial cells. Collectively, these data suggest that PTPRZ1 regulates
cardiac morphogenesis in a way that subsequently affects heart function and
warrant further studies for the involvement of PTPRZ1 in idiopathic congenital
cardiac pathologies. NEW & NOTEWORTHY Protein tyrosine phosphatase receptor
ζ1 (PTPRZ1) is expressed in fetal but not adult heart and seems
to affect heart development. In both mouse and zebrafish animal models, loss of
PTPRZ1 results in dilated left ventricle cavity, decreased ejection fraction,
and fraction shortening, with no signs of cardiac hypertrophy. PTPRZ1 also seems
to be involved in atrioventricular canal specification, outflow tract
morphogenesis, and heart angiogenesis. These results suggest that PTPRZ1 plays a
role in heart development and support the hypothesis that it may be involved in
congenital cardiac pathologies.
Collapse
Affiliation(s)
- Stamatiki Katraki-Pavlou
- Zebrafish Disease Models Lab, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Greece.,Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Greece
| | - Pinelopi Kastana
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Greece
| | - Dimitris Bousis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Greece
| | - Despoina Ntenekou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Greece
| | - Aimilia Varela
- Cardiovascular Research Laboratory, Biomedical Research Foundation, Academy of Athens, Greece
| | - Constantinos H Davos
- Cardiovascular Research Laboratory, Biomedical Research Foundation, Academy of Athens, Greece
| | - Sophia Nikou
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, Greece
| | - Eleni Papadaki
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, Greece
| | - Grigorios Tsigkas
- Department of Cardiology, Patras University Hospital, Rio, Patras, Greece
| | | | - Gonzalo Herradon
- Department of Pharmaceutical and Health Sciences, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Dimitris Beis
- Zebrafish Disease Models Lab, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Greece
| | - Evangelia Papadimitriou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Greece
| |
Collapse
|