1
|
Granzier HL, Labeit S. Discovery of Titin and Its Role in Heart Function and Disease. Circ Res 2025; 136:135-157. [PMID: 39745989 DOI: 10.1161/circresaha.124.323051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 01/04/2025]
Abstract
This review examines the giant elastic protein titin and its critical roles in heart function, both in health and disease, as discovered since its identification nearly 50 years ago. Encoded by the TTN (titin gene), titin has emerged as a major disease locus for cardiac disorders. Functionally, titin acts as a third myofilament type, connecting sarcomeric Z-disks and M-bands, and regulating myocardial passive stiffness and stretch sensing. Its I-band segment, which includes the N2B element and the PEVK (proline, glutamate, valine, and lysine-rich regions), serves as a viscoelastic spring, adjusting sarcomere length and force in response to cardiac stretch. The review details how alternative splicing of titin pre-mRNA produces different isoforms that greatly impact passive tension and cardiac function, under physiological and pathological conditions. Key posttranslational modifications, especially phosphorylation, play crucial roles in adjusting titin's stiffness, allowing for rapid adaptation to changing hemodynamic demands. Abnormal titin modifications and dysregulation of isoforms are linked to cardiac diseases such as heart failure with preserved ejection fraction, where increased stiffness impairs diastolic function. In addition, the review discusses the importance of the A-band region of titin in setting thick filament length and enhancing Ca²+ sensitivity, contributing to the Frank-Starling Mechanism of the heart. TTN truncating variants are frequently associated with dilated cardiomyopathy, and the review outlines potential disease mechanisms, including haploinsufficiency, sarcomere disarray, and altered thick filament regulation. Variants in TTN have also been linked to conditions such as peripartum cardiomyopathy and chemotherapy-induced cardiomyopathy. Therapeutic avenues are explored, including targeting splicing factors such as RBM20 (RNA binding motif protein 20) to adjust isoform ratios or using engineered heart tissues to study disease mechanisms. Advances in genetic engineering, including CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats), offer promise for modifying TTN to treat titin-related cardiomyopathies. This comprehensive review highlights titin's structural, mechanical, and signaling roles in heart function and the impact of TTN mutations on cardiac diseases.
Collapse
Affiliation(s)
- Henk L Granzier
- Department of Cellular and Molecular Medicine, Molecular Cardiovascular Research Program, The University of Arizona, Tucson (H.L.G.)
| | - Siegfried Labeit
- Department of Integrative Pathophysiology, Medical Faculty Mannheim, DZHK Partnersite Mannheim-Heidelberg, University of Heidelberg, Germany (S.L.)
| |
Collapse
|
2
|
Mir R, Altemani FH, Algehainy NA, Alanazi MA, Elfaki I, Alsayed BA, Mir MM, Mustafa SK, Moawadh MS, Tayeb FJ, Alfaifi J, Alatawi SM, Alhiwety MS, Ullah MF. Identification of Novel Genomic Variants in COVID-19 Patients Using Whole-Exome Sequencing: Exploring the Plausible Targets of Functional Genomics. Biochem Genet 2024:10.1007/s10528-024-10970-8. [PMID: 39557769 DOI: 10.1007/s10528-024-10970-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 11/02/2024] [Indexed: 11/20/2024]
Abstract
Covid-19 caused by SARS-CoV-2 virus has emerged as an immense burden and an unparalleled global health challenge in recorded human history. The clinical characteristics and risk factors of COVID-19 exhibit considerable variability, leading to a spectrum of clinical severity. Moreover, the likelihood of exposure to the virus may differ based on comorbidity status as comorbid illnesses have mechanisms that can considerably increase mortality by reducing the body's ability to withstand injury. The mammalian target of rapamycin (mTOR) pathway is essential for orchestrating innate immune cell defense, including cytokine production and is dysregulated in severe Coronavirus Disease 2019 (COVID-19) individuals. Through genome-wide, association studies, numerous genetic variants in the human host have been identified that have a significant impact on the immune response to SARS-CoV-2. To identify potentially significant genetic variants in Covid-19 patients that could affect the risk, severity, and clinical outcome of the infection, this study has used whole-exome sequencing (WES) on the 16 COVID-19 patients with varying comorbidities and severity of the disease including fatal outcomes. Among them, 8 patients made a full recovery and were discharged, while 8 patients unfortunately did not survive due to the severity of the illness and majority of them were males. The study identified 10,204 variants in the patients. From 1120 variants, which were chosen for novel variant analysis using mutation, function prediction tools to identify deleterious variants that could affect normal gene function, 116 variants of 57 genes were found to be deleterious. These variants were further classified as likely pathogenic and variants of uncertain significance. The data showed that among the likely pathogenic variants five genes were identified in connection to immune response whereas two were related to respiratory system. The common variants associated with the covid-19 phenotype showed the top 10 significant genes identified in this study such as ERCC2, FBXO5, HTR3D, FAIM, DNAH17, MTOR, IGHMBP2, ZNF530, QSER1, and FOXRED2 with variant rs1057079 of the MTOR gene representing the highest odds ratio (1.7, p = 8.7e-04). The mammalian target of rapamycin (mTOR) pathway variant rs1057079 was reported with high odds ratio, may orchestrate innate immune cell defense, including cytokine production, and is dysregulated. This study concluded that the mTOR signaling gene variant (rs1057079) is associated with different degrees of covid-19 severity and is essential for orchestrating innate immune cell defense including cytokine production. Inhibiting mTOR and its corresponding deleterious immune responses with medicinal approaches may provide a novel avenue for treating severe COVID-19 illness. Besides the PPI network exhibited a significantly high local clustering coefficient of 0.424 (p = 0.000536), suggesting the presence of tightly knit functional modules. These findings enhance our comprehension of the intricate interactions between genetic factors and COVID-19 disease.
Collapse
Affiliation(s)
- Rashid Mir
- Department of Medical Laboratory Technology Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, 71491, Tabuk, Saudi Arabia.
| | - Faisal H Altemani
- Department of Medical Laboratory Technology Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Naseh A Algehainy
- Department of Medical Laboratory Technology Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Mohammad A Alanazi
- Department of Medical Laboratory Technology Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Badr A Alsayed
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Mohammad Muzaffar Mir
- Department of Clinical Biochemistry, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Syed Khalid Mustafa
- Department of Chemistry, Faculty of Science, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Mamdoh S Moawadh
- Department of Medical Laboratory Technology Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Faris J Tayeb
- Department of Medical Laboratory Technology Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Jaber Alfaifi
- Department of Child Health, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Sael M Alatawi
- Department of Medical Laboratory Technology Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | | | - Mohammad Fahad Ullah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, King Faisel Road, 7149, Tabuk, Saudi Arabia.
| |
Collapse
|
3
|
Nasonova SN, Meshkov AN, Zhirov IV, Osmolovskaya YF, Shoshina AA, Gagloev AV, Dzhumaniiazova IH, Zelenova EA, Erema VV, Gusakova MS, Ivanov MV, Terekhov MV, Kashtanova DA, Nekrasova AI, Mitrofanov SI, Shingaliev AS, Yudin VS, Keskinov AA, Gomyranova NV, Chubykina UV, Ezhov MV, Tereshchenko SN, Yudin SM, Boytsov SA. [A clinical case of reverse left ventricular remodeling in patient with pathogenic TTN mutation. Case report]. TERAPEVT ARKH 2024; 96:901-908. [PMID: 39467245 DOI: 10.26442/00403660.2024.09.202852] [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: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 10/30/2024]
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure, sudden cardiac death, and heart transplantation in young patients. The causes of DCM are varied and include genetic factors and metabolic, infectious, toxic and others factors. Today it is known that germline mutations in more than 98 genes can be associated with the occurrence of DCM. However, the penetrance of these genes often depends on a combination of factors, including modifiable ones, i.e. those that change under the influence of the environment. About 20-25% of genetically determined forms of DCM are due to mutations in the titin gene (TTN). Titin is the largest protein in the body, which is an important component of the sarcomer. Although titin is the largest protein in the human body, its role in the physiology of heart and disease is not yet fully understood. However, a mutation in the TTN gene may later represent a potential therapeutic target for genetic and acquired cardiomyopathy. Thus, the analysis of clinical cases of cardiomyopathy in patients with identified mutations in the TTN gene is of great scientific interest. The article presents a clinical case of manifestation of DCM in patient with a revealed pathogenic variant of mutation in the gene TTN and reverse left ventricular remodeling of the against the background of optimal therapy of heart failure in a subsequent outpatient observation.
Collapse
Affiliation(s)
- S N Nasonova
- Chazov National Medical Research Center of Cardiology
| | - A N Meshkov
- Chazov National Medical Research Center of Cardiology
- National Research Center for Therapy and Preventive Medicine
| | - I V Zhirov
- Chazov National Medical Research Center of Cardiology
| | | | - A A Shoshina
- Chazov National Medical Research Center of Cardiology
| | - A V Gagloev
- Chazov National Medical Research Center of Cardiology
| | | | - E A Zelenova
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - V V Erema
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - M S Gusakova
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - M V Ivanov
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - M V Terekhov
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - D A Kashtanova
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - A I Nekrasova
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - S I Mitrofanov
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - A S Shingaliev
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - V S Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - A A Keskinov
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | | | - U V Chubykina
- Chazov National Medical Research Center of Cardiology
| | - M V Ezhov
- Chazov National Medical Research Center of Cardiology
| | | | - S M Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks
| | - S A Boytsov
- Chazov National Medical Research Center of Cardiology
| |
Collapse
|
4
|
Leońska-Duniec A, Maculewicz E, Massidda M, Buryta M, Mastalerz A, Cięszczyk P. Impact of the TTN C > T Polymorphism on Selected Variables of Aerobic and Anaerobic Capacity after a 12-Week Training Program. J Hum Kinet 2024; 94:117-125. [PMID: 39563773 PMCID: PMC11571456 DOI: 10.5114/jhk/191847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/31/2024] [Indexed: 11/21/2024] Open
Abstract
The TTN gene encodes a large muscle protein called titin, which provides structure, stability, and flexibility to skeletal and cardiac sarcomeres. The aim of this study was to determine whether the TTN C > T polymorphism (rs10497520) influenced training-induced changes in selected variables of aerobic and anaerobic capacity. We studied genotypes distribution in a group of 156 Caucasian females examined for aerobic capacity evaluated by maximal oxygen uptake (VO2max), and anaerobic capacity measured with the Wingate anaerobic test, before and after a 12-week training program. The most important finding was a genotype by training interaction for anaerobic capacity (AnC) during the Wingate test (p = 0.003). In response to training, carriers of the CT and TT genotypes demonstrated a significant increase in the total amount of work accomplished. We also showed that the applied training program improved all the Wingate test variables in the CT + TT genotype group by 10%. The obtained results suggest that the CT and TT genotypes may enhance anaerobic power and anaerobic capacity changes induced by regular training. We also suggest that T allele carriers may possess a metabolic adaptive advantage towards the anaerobic metabolism. Thus, the TTN gene may be considered a promising marker used in sports science, underlying variability in achieving sporting goals in events where the anaerobic energy system predominates.
Collapse
Affiliation(s)
- Agata Leońska-Duniec
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Ewelina Maculewicz
- Faculty of Physical Education, Jozef Pilsudski University of Physical Education in Warsaw, Warsaw, Poland
| | - Myosotis Massidda
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Maciej Buryta
- Institute of Physical Culture Sciences, University of Szczecin, Szczecin, Poland
| | - Andrzej Mastalerz
- Faculty of Physical Education, Jozef Pilsudski University of Physical Education in Warsaw, Warsaw, Poland
| | - Paweł Cięszczyk
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, Gdansk, Poland
| |
Collapse
|
5
|
Zhang X, Wang Y, Li H, Wang DW, Chen C. Insights into the post-translational modifications in heart failure. Ageing Res Rev 2024; 100:102467. [PMID: 39187021 DOI: 10.1016/j.arr.2024.102467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Heart failure (HF), as the terminal manifestation of multiple cardiovascular diseases, causes a huge socioeconomic burden worldwide. Despite the advances in drugs and medical-assisted devices, the prognosis of HF remains poor. HF is well-accepted as a myriad of subcellular dys-synchrony related to detrimental structural and functional remodelling of cardiac components, including cardiomyocytes, fibroblasts, endothelial cells and macrophages. Through the covalent chemical process, post-translational modifications (PTMs) can coordinate protein functions, such as re-localizing cellular proteins, marking proteins for degradation, inducing interactions with other proteins and tuning enzyme activities, to participate in the progress of HF. Phosphorylation, acetylation, and ubiquitination predominate in the currently reported PTMs. In addition, advanced HF is commonly accompanied by metabolic remodelling including enhanced glycolysis. Thus, glycosylation induced by disturbed energy supply is also important. In this review, firstly, we addressed the main types of HF. Then, considering that PTMs are associated with subcellular locations, we summarized the leading regulation mechanisms in organelles of distinctive cell types of different types of HF, respectively. Subsequently, we outlined the aforementioned four PTMs of key proteins and signaling sites in HF. Finally, we discussed the perspectives of PTMs for potential therapeutic targets in HF.
Collapse
Affiliation(s)
- Xudong Zhang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Yan Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| |
Collapse
|
6
|
Shetty NS, Gaonkar M, Pampana A, Patel N, Li P, Arora G, Arora P. Titin truncating variants, cardiovascular risk factors and the risk of atrial fibrillation and heart failure. NATURE CARDIOVASCULAR RESEARCH 2024; 3:899-906. [PMID: 39196037 DOI: 10.1038/s44161-024-00511-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/20/2024] [Indexed: 08/29/2024]
Abstract
High-proportion spliced-in (hiPSI) titin truncating variant (TTNtv) carriers have a higher risk of atrial fibrillation and heart failure1. However, the role of cardiovascular risk factors in modifying the risk of atrial fibrillation and heart failure attributed to hiPSI TTNtv carriers is unknown. Here, we investigate the role of cardiovascular risk, quantified using the pooled cohort equations (PCEs), in influencing the hazard of outcomes attributed to hiPSI TTNtvs among UK Biobank participants without baseline cardiovascular disease. The cohort was stratified based on hiPSI TTNtv carrier status and cardiovascular risk (low: <5%, intermediate: 5.0-7.5% and high: >7.5%). The primary outcome was a composite of atrial fibrillation, heart failure or death. TTNtv noncarriers with low cardiovascular risk were used as the reference group for all analyses. Among 179,752 participants (median age: 56 (49, 62) years; 57.5% female), the risk of the primary outcome was lower in hiPSI TTNtv carriers with low cardiovascular risk (adjusted hazard ratio: 2.23 (95% confidence interval: 1.62-3.07)) than those with high cardiovascular risk (adjusted hazard ratio: 8.21 (95% confidence interval: 6.63-10.18)). A favorable cardiovascular risk factor profile may partially offset the risk of clinical outcomes among hiPSI TTNtv carriers.
Collapse
Affiliation(s)
- Naman S Shetty
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mokshad Gaonkar
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Akhil Pampana
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nirav Patel
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Peng Li
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Garima Arora
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Pankaj Arora
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA.
- Section of Cardiology, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.
| |
Collapse
|
7
|
Han YS, Pakkam M, Fogarty MJ, Sieck GC, Brozovich FV. Alterations in cardiac contractile and regulatory proteins contribute to age-related cardiac dysfunction in male rats. Physiol Rep 2024; 12:e70012. [PMID: 39169429 PMCID: PMC11338742 DOI: 10.14814/phy2.70012] [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: 03/26/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
Aging is associated with cardiac contractile abnormalities, but the etiology of these contractile deficits is unclear. We hypothesized that cardiac contractile and regulatory protein expression is altered during aging. To investigate this possibility, left ventricular (LV) lysates were prepared from young (6 months) and old (24 months) Fischer344 rats. There are no age-related changes in SERCA2 expression or phospholamban phosphorylation. Additionally, neither titin isoform expression nor phosphorylation differed. However, there is a significant increase in β-isoform of the myosin heavy chain (MyHC) expression and phosphorylation of TnI and MyBP-C during aging. In permeabilized strips of papillary muscle, force and Ca2+ sensitivity are reduced during aging, consistent with the increase in β-MyHC expression and TnI phosphorylation. However, the increase in MyBP-C phosphorylation during aging may represent a mechanism to compensate for age-related contractile deficits. In isolated cardiomyocytes loaded with Fura-2, the peak of the Ca2+ transient is reduced, but the kinetics of the Ca2+ transient are not altered. Furthermore, the extent of shortening and the rates of both sarcomere shortening and re-lengthening are reduced. These results demonstrate that aging is associated with changes in contractile and regulatory protein expression and phosphorylation, which affect the mechanical properties of cardiac muscle.
Collapse
Affiliation(s)
- Young Soo Han
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Madona Pakkam
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Matthew J. Fogarty
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Gary C. Sieck
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Frank V. Brozovich
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Department of Cardiovascular DiseasesMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
8
|
Arnautu DA, Cozma D, Lala IR, Arnautu SF, Tomescu MC, Andor M. Risk Assessment and Personalized Treatment Options in Inherited Dilated Cardiomyopathies: A Narrative Review. Biomedicines 2024; 12:1643. [PMID: 39200108 PMCID: PMC11351202 DOI: 10.3390/biomedicines12081643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 09/01/2024] Open
Abstract
Considering the worldwide impact of heart failure, it is crucial to develop approaches that can help us comprehend its root cause and make accurate predictions about its outcome. This is essential for lowering the suffering and death rates connected with this widespread illness. Cardiomyopathies frequently result from genetic factors, and the study of heart failure genetics is advancing quickly. Dilated cardiomyopathy (DCM) is the most prevalent kind of cardiomyopathy, encompassing both genetic and nongenetic abnormalities. It is distinguished by the enlargement of the left ventricle or both ventricles, accompanied by reduced contractility. The discovery of the molecular origins and subsequent awareness of the molecular mechanism is broadening our knowledge of DCM development. Additionally, it emphasizes the complicated nature of DCM and the necessity to formulate several different strategies to address the diverse underlying factors contributing to this disease. Genetic variants that can be transmitted from one generation to another can be a significant contributor to causing family or sporadic hereditary DCM. Genetic variants also play a significant role in determining susceptibility for acquired triggers for DCM. The genetic causes of DCM can have a large range of phenotypic expressions. It is crucial to select patients who are most probable to gain advantages from genetic testing. The purpose of this research is to emphasize the significance of identifying genetic DCM, the relationships between genotype and phenotype, risk assessment, and personalized therapy for both those affected and their relatives. This approach is expected to gain importance once treatment is guided by genotype-specific advice and disease-modifying medications.
Collapse
Affiliation(s)
- Diana-Aurora Arnautu
- Multidisciplinary Heart Research Center, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (D.-A.A.); (M.-C.T.)
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Dragos Cozma
- Department of Cardiology, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Ioan-Radu Lala
- Department of Cardiology, Western University Vasile Goldis, 310025 Arad, Romania
| | - Sergiu-Florin Arnautu
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Mirela-Cleopatra Tomescu
- Multidisciplinary Heart Research Center, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (D.-A.A.); (M.-C.T.)
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Minodora Andor
- Multidisciplinary Heart Research Center, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (D.-A.A.); (M.-C.T.)
- Department of Internal Medicine I, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
9
|
Varghese TP. Genetic Markers of Cardiovascular Disease. Curr Probl Cardiol 2024; 49:102588. [PMID: 38657720 DOI: 10.1016/j.cpcardiol.2024.102588] [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: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Cardiovascular Disorders (CVDs) are the leading cause mortality in developed as well as developing nations, and has now emerged as one of the leading causes of disability and mortality around the globe. According to the World Health Organization, four out of every five patients with cardiovascular disease die from a myocardial infarction each year. Numerous genes have been linked to coronary artery disease, influencing mechanisms such as blood pressure regulation, lipid metabolism, inflammation, and cardiac activity. Genetic variations or mutations in these genes can affect lipid metabolism, blood pressure management, and heart function, increasing the risk of obesity, metabolic disorders, and resulting in the development of cardiovascular disease. Understanding the role of genes and related complications are essential for the identification, management, and prevention of cardiovascular conditions. Performing a genetic test for variations in the gene may help identify people as well as their families who are at a greater risk of heart disease, which enables risk identification and timely intervention. . This article investigates the applications of genetic biomarkers in cardiac disorders such as coronary artery disease, hypertension, arrhythmias, cardiomyopathy, and heart failure, with an emphasis on individual genes and their effects on mutation.
Collapse
Affiliation(s)
- Treesa P Varghese
- Department of Pharmacy Practice, Yenepoya Pharmacy College & Research centre, Yenepoya (Deemed to be University), Ayush campus, Naringana, Mangalore, Karnataka, India.
| |
Collapse
|
10
|
Solaro RJ, Goldspink PH, Wolska BM. Emerging Concepts of Mechanisms Controlling Cardiac Tension: Focus on Familial Dilated Cardiomyopathy (DCM) and Sarcomere-Directed Therapies. Biomedicines 2024; 12:999. [PMID: 38790961 PMCID: PMC11117855 DOI: 10.3390/biomedicines12050999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
Novel therapies for the treatment of familial dilated cardiomyopathy (DCM) are lacking. Shaping research directions to clinical needs is critical. Triggers for the progression of the disorder commonly occur due to specific gene variants that affect the production of sarcomeric/cytoskeletal proteins. Generally, these variants cause a decrease in tension by the myofilaments, resulting in signaling abnormalities within the micro-environment, which over time result in structural and functional maladaptations, leading to heart failure (HF). Current concepts support the hypothesis that the mutant sarcomere proteins induce a causal depression in the tension-time integral (TTI) of linear preparations of cardiac muscle. However, molecular mechanisms underlying tension generation particularly concerning mutant proteins and their impact on sarcomere molecular signaling are currently controversial. Thus, there is a need for clarification as to how mutant proteins affect sarcomere molecular signaling in the etiology and progression of DCM. A main topic in this controversy is the control of the number of tension-generating myosin heads reacting with the thin filament. One line of investigation proposes that this number is determined by changes in the ratio of myosin heads in a sequestered super-relaxed state (SRX) or in a disordered relaxed state (DRX) poised for force generation upon the Ca2+ activation of the thin filament. Contrasting evidence from nanometer-micrometer-scale X-ray diffraction in intact trabeculae indicates that the SRX/DRX states may have a lesser role. Instead, the proposal is that myosin heads are in a basal OFF state in relaxation then transfer to an ON state through a mechano-sensing mechanism induced during early thin filament activation and increasing thick filament strain. Recent evidence about the modulation of these mechanisms by protein phosphorylation has also introduced a need for reconsidering the control of tension. We discuss these mechanisms that lead to different ideas related to how tension is disturbed by levels of mutant sarcomere proteins linked to the expression of gene variants in the complex landscape of DCM. Resolving the various mechanisms and incorporating them into a unified concept is crucial for gaining a comprehensive understanding of DCM. This deeper understanding is not only important for diagnosis and treatment strategies with small molecules, but also for understanding the reciprocal signaling processes that occur between cardiac myocytes and their micro-environment. By unraveling these complexities, we can pave the way for improved therapeutic interventions for managing DCM.
Collapse
Affiliation(s)
- R. John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
| | - Paul H. Goldspink
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
| | - Beata M. Wolska
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
- Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
11
|
Weston TGR, Rees M, Gautel M, Fraternali F. Walking with giants: The challenges of variant impact assessment in the giant sarcomeric protein titin. WIREs Mech Dis 2024; 16:e1638. [PMID: 38155593 DOI: 10.1002/wsbm.1638] [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: 06/15/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
Titin, the so-called "third filament" of the sarcomere, represents a difficult challenge for the determination of damaging genetic variants. A single titin molecule extends across half the length of a sarcomere in striated muscle, fulfilling a variety of vital structural and signaling roles, and has been linked to an equally varied range of myopathies, resulting in a significant burden on individuals and healthcare systems alike. While the consequences of truncating variants of titin are well-documented, the ramifications of the missense variants prevalent in the general population are less so. We here present a compendium of titin missense variants-those that result in a single amino-acid substitution in coding regions-reported to be pathogenic and discuss these in light of the nature of titin and the variant position within the sarcomere and their domain, the structural, pathological, and biophysical characteristics that define them, and the methods used for characterization. Finally, we discuss the current knowledge and integration of the multiple fields that have contributed to our understanding of titin-related pathology and offer suggestions as to how these concurrent methodologies may aid the further development in our understanding of titin and hopefully extend to other, less well-studied giant proteins. This article is categorized under: Cardiovascular Diseases > Genetics/Genomics/Epigenetics Congenital Diseases > Genetics/Genomics/Epigenetics Congenital Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Timir G R Weston
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Martin Rees
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Mathias Gautel
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Franca Fraternali
- Institute of Structural and Molecular Biology, University College London, London, UK
| |
Collapse
|
12
|
Gan P, Wang Z, Bezprozvannaya S, McAnally JR, Tan W, Li H, Bassel-Duby R, Liu N, Olson EN. RBPMS regulates cardiomyocyte contraction and cardiac function through RNA alternative splicing. Cardiovasc Res 2024; 120:56-68. [PMID: 37890031 PMCID: PMC10898938 DOI: 10.1093/cvr/cvad166] [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: 06/15/2023] [Revised: 07/20/2023] [Accepted: 09/23/2023] [Indexed: 10/29/2023] Open
Abstract
AIMS RNA binding proteins play essential roles in mediating RNA splicing and are key post-transcriptional regulators in the heart. Our recent study demonstrated that RBPMS (RNA binding protein with multiple splicing) is crucial for cardiac development through modulating mRNA splicing, but little is known about its functions in the adult heart. In this study, we aim to characterize the post-natal cardiac function of Rbpms and its mechanism of action. METHODS AND RESULTS We generated a cardiac-specific knockout mouse line and found that cardiac-specific loss of Rbpms caused severe cardiomyocyte contractile defects, leading to dilated cardiomyopathy and early lethality in adult mice. We showed by proximity-dependent biotin identification assay and mass spectrometry that RBPMS associates with spliceosome factors and other RNA binding proteins, such as RBM20, that are important in cardiac function. We performed paired-end RNA sequencing and RT-PCR and found that RBPMS regulates mRNA alternative splicing of genes associated with sarcomere structure and function, such as Ttn, Pdlim5, and Nexn, generating new protein isoforms. Using a minigene splicing reporter assay, we determined that RBPMS regulates target gene splicing through recognizing tandem intronic CAC motifs. We also showed that RBPMS knockdown in human induced pluripotent stem cell-derived cardiomyocytes impaired cardiomyocyte contraction. CONCLUSION This study identifies RBPMS as an important regulator of cardiomyocyte contraction and cardiac function by modulating sarcomeric gene alternative splicing.
Collapse
Affiliation(s)
- Peiheng Gan
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Zhaoning Wang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Svetlana Bezprozvannaya
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| | - John R McAnally
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Wei Tan
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Hui Li
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Ning Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| |
Collapse
|
13
|
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
|
14
|
ElSaygh J, Zaher A, Peterson SJ, Parikh MA, Frishman WH. Titin: The Missing Link in Cardiac Physiology. Cardiol Rev 2024:00045415-990000000-00209. [PMID: 38334419 DOI: 10.1097/crd.0000000000000656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Titin, an extraordinary protein known for its colossal size and multifaceted roles, is a cornerstone in the structural and functional dynamics of striated muscle tissues, including the heart and skeletal muscles. Its sheer enormity, with a molecular weight exceeding 3000 kDa, is paralleled only by the immense influence it exerts on muscle physiology. This review will delve into the remarkable structural organization of Titin and the genetics of this molecule, including the common mutations resulting in various cardiomyopathies. We will delve deeper into its role in dilated cardiomyopathy, familial restrictive cardiomyopathy, hypertrophic cardiomyopathy, and left ventricular noncompaction cardiomyopathy. This review culminates by discussing the prospects of therapeutic strategies targeting Titin. While these interventions remain primarily theoretical, the possibilities are intriguing. Patients with Titin truncation mutations present unique challenges, but innovative approaches like gene therapy or preemptive treatments with drugs such as angiotensin-converting enzyme inhibitors or beta-blockers offer hope. This multi-pronged approach highlights the significance of understanding Titin's multifaceted role and its potential as a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Jude ElSaygh
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
| | - Anas Zaher
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
| | - Stephen J Peterson
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
- Weill Department of Medicine, Weill Cornell Medicine, NY
| | - Manish A Parikh
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
- Weill Department of Medicine, Weill Cornell Medicine, NY
| | | |
Collapse
|
15
|
Wang S, Zhang Z, He J, Liu J, Guo X, Chu H, Xu H, Wang Y. Comprehensive review on gene mutations contributing to dilated cardiomyopathy. Front Cardiovasc Med 2023; 10:1296389. [PMID: 38107262 PMCID: PMC10722203 DOI: 10.3389/fcvm.2023.1296389] [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: 09/18/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the most common primary myocardial diseases. However, to this day, it remains an enigmatic cardiovascular disease (CVD) characterized by ventricular dilatation, which leads to myocardial contractile dysfunction. It is the most common cause of chronic congestive heart failure and the most frequent indication for heart transplantation in young individuals. Genetics and various other factors play significant roles in the progression of dilated cardiomyopathy, and variants in more than 50 genes have been associated with the disease. However, the etiology of a large number of cases remains elusive. Numerous studies have been conducted on the genetic causes of dilated cardiomyopathy. These genetic studies suggest that mutations in genes for fibronectin, cytoskeletal proteins, and myosin in cardiomyocytes play a key role in the development of DCM. In this review, we provide a comprehensive description of the genetic basis, mechanisms, and research advances in genes that have been strongly associated with DCM based on evidence-based medicine. We also emphasize the important role of gene sequencing in therapy for potential early diagnosis and improved clinical management of DCM.
Collapse
Affiliation(s)
- Shipeng Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zhiyu Zhang
- Department of Cardiovascular Medicine, The Second People's Hospital of Yibin, Yibin, China
| | - Jiahuan He
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Junqian Liu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xia Guo
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Haoxuan Chu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Hanchi Xu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yushi Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Madè A, Bibi A, Garcia-Manteiga JM, Tascini AS, Piella SN, Tikhomirov R, Voellenkle C, Gaetano C, Leszek P, Castelvecchio S, Menicanti L, Martelli F, Greco S. circRNA-miRNA-mRNA Deregulated Network in Ischemic Heart Failure Patients. Cells 2023; 12:2578. [PMID: 37947656 PMCID: PMC10648415 DOI: 10.3390/cells12212578] [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: 09/15/2023] [Revised: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
Noncoding RNAs (ncRNAs), which include circular RNAs (circRNAs) and microRNAs (miRNAs), regulate the development of cardiovascular diseases (CVD). Notably, circRNAs can interact with miRNAs, influencing their specific mRNA targets' levels and shaping a competing endogenous RNAs (ceRNA) network. However, these interactions and their respective functions remain largely unexplored in ischemic heart failure (IHF). This study is aimed at identifying circRNA-centered ceRNA networks in non-end-stage IHF. Approximately 662 circRNA-miRNA-mRNA interactions were identified in the heart by combining state-of-the-art bioinformatics tools with experimental data. Importantly, KEGG terms of the enriched mRNA indicated CVD-related signaling pathways. A specific network centered on circBPTF was validated experimentally. The levels of let-7a-5p, miR-18a-3p, miR-146b-5p, and miR-196b-5p were enriched in circBPTF pull-down experiments, and circBPTF silencing inhibited the expression of HDAC9 and LRRC17, which are targets of miR-196b-5p. Furthermore, as suggested by the enriched pathway terms of the circBPTF ceRNA network, circBPTF inhibition elicited endothelial cell cycle arrest. circBPTF expression increased in endothelial cells exposed to hypoxia, and its upregulation was confirmed in cardiac samples of 36 end-stage IHF patients compared to healthy controls. In conclusion, circRNAs act as miRNA sponges, regulating the functions of multiple mRNA targets, thus providing a novel vision of HF pathogenesis and laying the theoretical foundation for further experimental studies.
Collapse
Affiliation(s)
- Alisia Madè
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Alessia Bibi
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
- Department of Biosciences, University of Milan, 20122 Milan, Italy
| | - Jose Manuel Garcia-Manteiga
- Center for Omics Sciences COSR, BioInformatics Laboratory, San Raffaele Scientific Institute, 20132 Milan, Italy; (J.M.G.-M.); (A.S.T.)
| | - Anna Sofia Tascini
- Center for Omics Sciences COSR, BioInformatics Laboratory, San Raffaele Scientific Institute, 20132 Milan, Italy; (J.M.G.-M.); (A.S.T.)
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Santiago Nicolas Piella
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Roman Tikhomirov
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, National Institute of Cardiology, 04-628 Warsaw, Poland;
| | - Serenella Castelvecchio
- Department of Adult Cardiac Surgery, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (S.C.); (L.M.)
| | - Lorenzo Menicanti
- Department of Adult Cardiac Surgery, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (S.C.); (L.M.)
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| |
Collapse
|
17
|
A Novel Nonsense Pathogenic TTN Variant Identified in a Patient with Severe Dilated Cardiomyopathy. Curr Issues Mol Biol 2023; 45:2422-2430. [PMID: 36975527 PMCID: PMC10047881 DOI: 10.3390/cimb45030157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Both genetic and environmental factors contribute to the development of dilated cardiomyopathy. Among the genes involved, TTN mutations, including truncated variants, explain 25% of DCM cases. We performed genetic counseling and analysis on a 57-year-old woman diagnosed with severe DCM and presenting relevant acquired risk factors for DCM (hypertension, diabetes, smoking habit, and/or previous alcohol and cocaine abuse) and with a family history of both DCM and sudden cardiac death. The left ventricular systolic function, as assessed by standard echocardiography, was 20%. The genetic analysis performed using TruSight Cardio panel, including 174 genes related to cardiac genetic diseases, revealed a novel nonsense TTN variant (TTN:c.103591A > T, p.Lys34531*), falling within the M-band region of the titin protein. This region is known for its important role in maintaining the structure of the sarcomere and in promoting sarcomerogenesis. The identified variant was classified as likely pathogenic based on ACMG criteria. The current results support the need of genetic analysis in the presence of a family history, even when relevant acquired risk factors for DCM may have contributed to the severity of the disease.
Collapse
|
18
|
Titin activates myosin filaments in skeletal muscle by switching from an extensible spring to a mechanical rectifier. Proc Natl Acad Sci U S A 2023; 120:e2219346120. [PMID: 36812205 PMCID: PMC9992839 DOI: 10.1073/pnas.2219346120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
Titin is a molecular spring in parallel with myosin motors in each muscle half-sarcomere, responsible for passive force development at sarcomere length (SL) above the physiological range (>2.7 μm). The role of titin at physiological SL is unclear and is investigated here in single intact muscle cells of the frog (Rana esculenta), by combining half-sarcomere mechanics and synchrotron X-ray diffraction in the presence of 20 μM para-nitro-blebbistatin, which abolishes the activity of myosin motors and maintains them in the resting state even during activation of the cell by electrical stimulation. We show that, during cell activation at physiological SL, titin in the I-band switches from an SL-dependent extensible spring (OFF-state) to an SL-independent rectifier (ON-state) that allows free shortening while resisting stretch with an effective stiffness of ~3 pN nm-1 per half-thick filament. In this way, I-band titin efficiently transmits any load increase to the myosin filament in the A-band. Small-angle X-ray diffraction signals reveal that, with I-band titin ON, the periodic interactions of A-band titin with myosin motors alter their resting disposition in a load-dependent manner, biasing the azimuthal orientation of the motors toward actin. This work sets the stage for future investigations on scaffold and mechanosensing-based signaling functions of titin in health and disease.
Collapse
|
19
|
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
|
20
|
Usui Y, Kimoto M, Hanashima A, Hashimoto K, Mohri S. Cardiac hemodynamics and ventricular stiffness of sea-run cherry salmon (Oncorhynchus masou masou) differ critically from those of landlocked masu salmon. PLoS One 2022; 17:e0267264. [PMID: 36331913 PMCID: PMC9635730 DOI: 10.1371/journal.pone.0267264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Ventricular diastolic mechanical properties are important determinants of cardiac function and are optimized by changes in cardiac structure and physical properties. Oncorhynchus masou masou is an anadromous migratory fish of the Salmonidae family, and several ecological studies on it have been conducted; however, the cardiac functions of the fish are not well known. Therefore, we investigated ventricular diastolic function in landlocked (masu salmon) and sea-run (cherry salmon) types at 29–30 months post fertilization. Pulsed-wave Doppler echocardiography showed that the atrioventricular inflow waveforms of cherry salmon were biphasic with early diastolic filling and atrial contraction, whereas those of masu salmon were monophasic with atrial contraction. In addition, end-diastolic pressure–volume relationship analysis revealed that the dilatability per unit myocardial mass of the ventricle in cherry salmon was significantly suppressed compared to that in masu salmon, suggesting that the ventricle of the cherry salmon was relatively stiffer (relative ventricular stiffness index; p = 0.0263). Contrastingly, the extensibility of cardiomyocytes, characterized by the expression pattern of Connectin isoforms in their ventricles, was similar in both types. Histological analysis showed that the percentage of the collagen accumulation area in the compact layer of cherry salmon increased compared with that of the masu salmon, which may contribute to ventricle stiffness. Although the heart mass of cherry salmon was about 11-fold greater than that of masu salmon, there was no difference in the morphology of the isolated cardiomyocytes, suggesting that the heart of the cherry salmon grows by cardiomyocyte proliferation, but not cell hypertrophy. The cardiac physiological function of the teleosts varies with differences in their developmental processes and life history. Our multidimensional analysis of the O. masou heart may provide a clue to the process by which the heart acquires a biphasic blood-filling pattern, i.e., a ventricular diastolic suction.
Collapse
Affiliation(s)
- Yuu Usui
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Japan
- * E-mail:
| | - Misaki Kimoto
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Japan
| | - Akira Hanashima
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Japan
| | - Ken Hashimoto
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Japan
| | - Satoshi Mohri
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Japan
| |
Collapse
|
21
|
Kawana M, Spudich JA, Ruppel KM. Hypertrophic cardiomyopathy: Mutations to mechanisms to therapies. Front Physiol 2022; 13:975076. [PMID: 36225299 PMCID: PMC9548533 DOI: 10.3389/fphys.2022.975076] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/22/2022] [Indexed: 01/10/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) affects more than 1 in 500 people in the general population with an extensive burden of morbidity in the form of arrhythmia, heart failure, and sudden death. More than 25 years since the discovery of the genetic underpinnings of HCM, the field has unveiled significant insights into the primary effects of these genetic mutations, especially for the myosin heavy chain gene, which is one of the most commonly mutated genes. Our group has studied the molecular effects of HCM mutations on human β-cardiac myosin heavy chain using state-of-the-art biochemical and biophysical tools for the past 10 years, combining insights from clinical genetics and structural analyses of cardiac myosin. The overarching hypothesis is that HCM-causing mutations in sarcomere proteins cause hypercontractility at the sarcomere level, and we have shown that an increase in the number of myosin molecules available for interaction with actin is a primary driver. Recently, two pharmaceutical companies have developed small molecule inhibitors of human cardiac myosin to counteract the molecular consequences of HCM pathogenesis. One of these inhibitors (mavacamten) has recently been approved by the FDA after completing a successful phase III trial in HCM patients, and the other (aficamten) is currently being evaluated in a phase III trial. Myosin inhibitors will be the first class of medication used to treat HCM that has both robust clinical trial evidence of efficacy and that targets the fundamental mechanism of HCM pathogenesis. The success of myosin inhibitors in HCM opens the door to finding other new drugs that target the sarcomere directly, as we learn more about the genetics and fundamental mechanisms of this disease.
Collapse
Affiliation(s)
- Masataka Kawana
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, United States,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, United States
| | - Kathleen M. Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, United States,*Correspondence: Kathleen M. Ruppel,
| |
Collapse
|
22
|
Leońska-Duniec A, Borczyk M, Piechota M, Korostyński M, Brodkiewicz A, Cięszczyk P. TTN Variants Are Associated with Physical Performance and Provide Potential Markers for Sport-Related Phenotypes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10173. [PMID: 36011809 PMCID: PMC9408402 DOI: 10.3390/ijerph191610173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 06/15/2023]
Abstract
TTN encodes the third myofilament, titin, which plays structural, mechanical, regulatory, and developmental roles in sarcomeres. The aim of this research was to determine the interaction between novel and previously described TTN variants and athletic performance, as well as competition level, in Caucasians. Firstly, 100 athletes and 47 controls were recruited, and whole-genome sequencing was performed. Secondly, 348 athletes (108 endurance, 100 sprint/power, 140 mixed-sport athletes) and 403 volunteers were included, and real-time PCR was performed. We found a significant overrepresentation of the rs10497520 CT and TT genotypes in the sprint/power athlete group (95% CI, 1.41-3.66, p = 0.0013). The rs10497520 T carriers were 2.17 times more likely to become sprint/power athletes (95% CI 1.35-3.49, p = 0.0021). We also found that the likelihood of having the TT genotype was higher for the highly elite and sub-elite sprint/power athletes. Possessing at least one TAA (rs10497520, rs55837610, rs72648256) haplotype resulted in an increase in the log-odds ratio by 0.80 (p = 0.0015), 1.42 (p = 0.003), and 0.77 (p = 0.044) for all, highly elite, and sub-elite sprint/power athletes, respectively. We demonstrated that harbouring the rs10497520 T allele, individually and in a haplotype combination, increased the chance of being an elite sprint/power athlete, indicating that this allele may be favourable for sprint/power performance.
Collapse
Affiliation(s)
- Agata Leońska-Duniec
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland
| | - Małgorzata Borczyk
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
| | - Marcin Piechota
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
| | - Michał Korostyński
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
| | - Andrzej Brodkiewicz
- Department of Pediatrics, Child Nephrology, Dialisotherapy and Management of Acute Poisoning, Pomeranian Medical University, 70-204 Szczecin, Poland
| | - Paweł Cięszczyk
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland
| |
Collapse
|
23
|
Exploring the Potential of Symmetric Exon Deletion to Treat Non-Ischemic Dilated Cardiomyopathy by Removing Frameshift Mutations in TTN. Genes (Basel) 2022; 13:genes13061093. [PMID: 35741855 PMCID: PMC9222585 DOI: 10.3390/genes13061093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/30/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Non-ischemic dilated cardiomyopathy (DCM) is one of the most frequent pathologies requiring cardiac transplants. Even though the etiology of this disease is complex, frameshift mutations in the giant sarcomeric protein Titin could explain up to 25% of the familial and 18% of the sporadic cases of DCM. Many studies have shown the potential of genome editing using CRISPR/Cas9 to correct truncating mutations in sarcomeric proteins and have established the grounds for myoediting. However, these therapies are still in an immature state, with only few studies showing an efficient treatment of cardiac diseases. This publication hypothesizes that the Titin (TTN)-specific gene structure allows the application of myoediting approaches in a broad range of locations to reframe TTNtvvariants and to treat DCM patients. Additionally, to pave the way for the generation of efficient myoediting approaches for DCM, we screened and selected promising target locations in TTN. We conceptually explored the deletion of symmetric exons as a therapeutic approach to restore TTN’s reading frame in cases of frameshift mutations. We identified a set of 94 potential candidate exons of TTN that we consider particularly suitable for this therapeutic deletion. With this study, we aim to contribute to the development of new therapies to efficiently treat titinopathies and other diseases caused by mutations in genes encoding proteins with modular structures, e.g., Obscurin.
Collapse
|
24
|
El Kadiri Y, Ratbi I, Sefiani A, Lyahyai J. Clinical and molecular genetic analysis of early-onset myopathy with fatal cardiomyopathy: Novel biallelic M-line TTN mutation and review of the literature. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
25
|
Wang C, Zhang Y, Methawasin M, Braz CU, Gao-Hu J, Yang B, Strom J, Gohlke J, Hacker T, Khatib H, Granzier H, Guo W. RBM20 S639G mutation is a high genetic risk factor for premature death through RNA-protein condensates. J Mol Cell Cardiol 2022; 165:115-129. [PMID: 35041844 PMCID: PMC8940686 DOI: 10.1016/j.yjmcc.2022.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/03/2022] [Accepted: 01/09/2022] [Indexed: 12/13/2022]
Abstract
Dilated cardiomyopathy (DCM) is a heritable and genetically heterogenous disease often idiopathic and a leading cause of heart failure with high morbidity and mortality. DCM caused by RNA binding motif protein 20 (RBM20) mutations is diverse and needs a more complete mechanistic understanding. RBM20 mutation S637G (S639G in mice) is linked to severe DCM and early death in human patients. In this study, we generated a RBM20 S639G mutation knock-in (KI) mouse model to validate the function of S639G mutation and examine the underlying mechanisms. KI mice exhibited severe DCM and premature death with a ~ 50% mortality in two months old homozygous (HM) mice. KI mice had enlarged atria and increased ANP and BNP biomarkers. The S639G mutation promoted RBM20 trafficking and ribonucleoprotein (RNP) granules in the sarcoplasm. RNA Seq data revealed differentially expressed and spliced genes were associated with arrhythmia, cardiomyopathy, and sudden death. KI mice also showed a reduction of diastolic stiffness and impaired contractility at both the left ventricular (LV) chamber and cardiomyocyte levels. Our results indicate that the RBM20 S639G mutation leads to RNP granules causing severe heart failure and early death and this finding strengthens the novel concept that RBM20 cardiomyopathy is a RNP granule disease.
Collapse
Affiliation(s)
- Chunyan Wang
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Yanghai Zhang
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Mei Methawasin
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Camila Urbano Braz
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Jeffrey Gao-Hu
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Betty Yang
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Joshua Strom
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Jochen Gohlke
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Timothy Hacker
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | - Hasan Khatib
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Wei Guo
- Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI 53706, USA.
| |
Collapse
|
26
|
Mesquita T, Lin Y, Ibrahim A. Chronic low-grade inflammation in heart failure with preserved ejection fraction. Aging Cell 2021; 20:e13453. [PMID: 34382743 PMCID: PMC8441359 DOI: 10.1111/acel.13453] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/21/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is currently the predominant form of HF with a dramatic increase in risk with age. Low-grade inflammation, as occurs with aging (termed "inflammaging"), is a common feature of HFpEF pathology. Suppression of proinflammatory pathways has been associated with attenuated HFpEF disease severity and better outcomes. From this perspective, inflammasome signaling plays a central role in mediating chronic inflammation and cardiovascular disease progression. However, the causal link between the inflammasome-immune signaling axis on the age-dependent progression of HFpEF remains conjectural. In this review, we summarize the current understanding of the role of inflammatory pathways in age-dependent cardiac function decline. We will also evaluate recent advances and evidence regarding the inflammatory pathway in the pathophysiology of HFpEF, with special attention to inflammasome signaling.
Collapse
Affiliation(s)
- Thassio Mesquita
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCAUSA
| | - Yen‐Nien Lin
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCAUSA
- Division of Cardiovascular MedicineDepartment of MedicineChina Medical University and HospitalTaichungTaiwan
| | - Ahmed Ibrahim
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCAUSA
| |
Collapse
|
27
|
The Sarcomeric Spring Protein Titin: Biophysical Properties, Molecular Mechanisms, and Genetic Mutations Associated with Heart Failure and Cardiomyopathy. Curr Cardiol Rep 2021; 23:121. [PMID: 34269900 DOI: 10.1007/s11886-021-01550-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/07/2021] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW The giant protein titin forms the "elastic" filament of the sarcomere, essential for the mechanical compliance of the heart muscle. Titin serves a biological spring, and therefore structural modifications of titin affect function of the myocardium and are associated with heart failure and cardiomyopathy. RECENT FINDINGS In this review, we discuss the current understanding of titin's biophysical properties and how modifications contribute to cardiac function and heart failure. In addition, we review the most recent data on the clinical impact and phenotype heterogeneity of TTN truncating variants, including diseases involving striated muscles, and prospects for future therapies. Because of the giant structure of the titin protein and the complexity of its function, titin's role in health and disease is not yet completely understood. Future research efforts need to focus on novel therapeutic approaches able to modulate titin transcriptional and post-translational modification.
Collapse
|
28
|
de Boer RA, Aboumsallem JP, Bracun V, Leedy D, Cheng R, Patel S, Rayan D, Zaharova S, Rymer J, Kwan JM, Levenson J, Ronco C, Thavendiranathan P, Brown SA. A new classification of cardio-oncology syndromes. CARDIO-ONCOLOGY 2021; 7:24. [PMID: 34154667 PMCID: PMC8218489 DOI: 10.1186/s40959-021-00110-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/30/2021] [Indexed: 12/21/2022]
Abstract
Increasing evidence suggests a multifaceted relationship exists between cancer and cardiovascular disease (CVD). Here, we introduce a 5-tier classification system to categorize cardio-oncology syndromes (COS) that represent the aspects of the relationship between cancer and CVD. COS Type I is characterized by mechanisms whereby the abrupt onset or progression of cancer can lead to cardiovascular dysfunction. COS Type II includes the mechanisms by which cancer therapies can result in acute or chronic CVD. COS Type III is characterized by the pro-oncogenic environment created by the release of cardiokines and high oxidative stress in patients with cardiovascular dysfunction. COS Type IV is comprised of CVD therapies and diagnostic procedures which have been associated with promoting or unmasking cancer. COS Type V is characterized by factors causing systemic and genetic predisposition to both CVD and cancer. The development of this framework may allow for an increased facilitation of cancer care while optimizing cardiovascular health through focused treatment targeting the COS type.
Collapse
Affiliation(s)
- Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Valentina Bracun
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Douglas Leedy
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Richard Cheng
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Sahishnu Patel
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - David Rayan
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Svetlana Zaharova
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | | | - Jennifer M Kwan
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Joshua Levenson
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Claudio Ronco
- Department of Medicine, University of Padova, Padova, Italy.,International Renal Research Institute of Vicenza, Vicenza, Italy.,Department of Nephrology, San Bortolo Hospital, Vicenza, Italy
| | | | - Sherry-Ann Brown
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
29
|
Paulus WJ, Zile MR. From Systemic Inflammation to Myocardial Fibrosis: The Heart Failure With Preserved Ejection Fraction Paradigm Revisited. Circ Res 2021; 128:1451-1467. [PMID: 33983831 PMCID: PMC8351796 DOI: 10.1161/circresaha.121.318159] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In accordance with the comorbidity-inflammation paradigm, comorbidities and especially metabolic comorbidities are presumed to drive development and severity of heart failure with preserved ejection fraction through a cascade of events ranging from systemic inflammation to myocardial fibrosis. Recently, novel experimental and clinical evidence emerged, which strengthens the validity of the inflammatory/profibrotic paradigm. This evidence consists among others of (1) myocardial infiltration by immunocompetent cells not only because of an obesity-induced metabolic load but also because of an arterial hypertension-induced hemodynamic load. The latter is sensed by components of the extracellular matrix like basal laminin, which also interact with cardiomyocyte titin; (2) expression in cardiomyocytes of inducible nitric oxide synthase because of circulating proinflammatory cytokines. This results in myocardial accumulation of degraded proteins because of a failing unfolded protein response; (3) definition by machine learning algorithms of phenogroups of patients with heart failure with preserved ejection fraction with a distinct inflammatory/profibrotic signature; (4) direct coupling in mediation analysis between comorbidities, inflammatory biomarkers, and deranged myocardial structure/function with endothelial expression of adhesion molecules already apparent in early preclinical heart failure with preserved ejection fraction (HF stage A, B). This new evidence paves the road for future heart failure with preserved ejection fraction treatments such as biologicals directed against inflammatory cytokines, stimulation of protein ubiquitylation with phosphodiesterase 1 inhibitors, correction of titin stiffness through natriuretic peptide-particulate guanylyl cyclase-PDE9 (phosphodiesterase 9) signaling and molecular/cellular regulatory mechanisms that control myocardial fibrosis.
Collapse
Affiliation(s)
- Walter J Paulus
- Amsterdam University Medical Centers, The Netherlands (W.J.P.)
| | - Michael R Zile
- RHJ Department of Veterans Affairs Medical Center, Medical University of South Carolina, Charleston (M.R.Z.)
| |
Collapse
|
30
|
Xiao L, Li C, Sun Y, Chen Y, Wei H, Hu D, Yu T, Li X, Jin L, Shi L, Marian AJ, Wang DW. Clinical Significance of Variants in the TTN Gene in a Large Cohort of Patients With Sporadic Dilated Cardiomyopathy. Front Cardiovasc Med 2021; 8:657689. [PMID: 33996946 PMCID: PMC8120103 DOI: 10.3389/fcvm.2021.657689] [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: 01/23/2021] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Mutations in the TTN gene are the most common causes of dilated cardiomyopathy (DCM). The clinical significance of TTN gene variants remains inadequately understood. Methods: Whole-exome sequencing and phenotypic characterisation were performed, and patients were followed up for a median of 44 months. Results: We analyzed the association of the TTN variants with the clinical outcomes in a prospective study of 1,041 patients with sporadic DCM. TTN truncating variants (tTTN) were detected in 120 (11.5%) patients as compared with 2.4/10,000 East Asian populations in the Genome Aggregation Database (GnomAD; p < 0.0001). Pathogenic TTN missense variants were also enriched in DCM as compared with the GnomAD populations (27.6 vs. 5.9%, p < 0.0001). DCM patients with tTTN had a lower left ventricular ejection fraction (28.89 ± 8.72 vs. 31.81 ± 9.97, p = 0.002) and a lower frequency of the left bundle branch block (3.3 vs. 11.3%, p = 0.011) than those without or with mutations in other known causal genes (OCG). However, tTTN were not associated with the composite primary endpoint of cardiac death and heart transplantation during the follow-up period [adjusted hazard ratio (HR): 0.912; 95% confidence interval: 0.464–1.793; p = 0.790]. There was also no sex-dependent effect. Concomitant tTTN and pathogenic variants in OCG were present in only eight DCM patients and did not affect the outcome. Conclusion: The phenotype of DCM caused by tTTN, major causes of sporadic DCM, is not distinctly different from those caused by other causal genes for DCM.
Collapse
Affiliation(s)
- Lei Xiao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chenze Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Sun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yanghui Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Haoran Wei
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Hu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Yu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Xianqing Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Li Jin
- Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Leming Shi
- Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Ali J Marian
- Center for Cardiovascular Genetics, Houston, TX, United States
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Connell PS, Berkman AM, Souder BM, Pirozzi EJ, Lovin JJ, Rosenfeld JA, Liu P, Tunuguntla H, Allen HD, Denfield SW, Kim JJ, Landstrom AP. Amino Acid-Level Signal-to-Noise Analysis Aids in Pathogenicity Prediction of Incidentally Identified TTN-Encoded Titin Truncating Variants. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2021; 14:e003131. [PMID: 33226272 PMCID: PMC7887062 DOI: 10.1161/circgen.120.003131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/14/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND TTN, the largest gene in the human body, encodes TTN (titin), a protein that plays key structural, developmental, and regulatory roles in skeletal and cardiac muscle. Variants in TTN, particularly truncating variants (TTNtvs), have been implicated in the pathogenicity of cardiomyopathy. Despite this link, there is also a high burden of TTNtvs in the ostensibly healthy general population. This complicates the diagnostic interpretation of incidentally identified TTNtvs, which are of increasing abundance given expanding clinical exome sequencing. METHODS Incidentally identified TTNtvs were obtained from a large referral database of clinical exome sequencing (Baylor Genetics) and compared with rare population variants from genome aggregation database and cardiomyopathy-associated variants from cohort studies in the literature. A subset of TTNtv-positive children evaluated for cardiomyopathy at Texas Children's Hospital was retrospectively reviewed for clinical features of cardiomyopathy. Amino acid-level signal-to-noise analysis was performed. RESULTS Pathological hotspots were identified within the A-band and N-terminal I-band that closely correlated with regions of high percent-spliced in of exons. Incidental TTNtvs and population TTNtvs did not localize to these regions. Variants were reclassified based on current American College of Medical Genetics and Genomics criteria with incorporation of signal-to-noise analysis among Texas Children's Hospital cases. Those reclassified as likely pathogenic or pathogenic were more likely to have evidence of cardiomyopathy on echocardiography than those reclassified as variants of unknown significance. CONCLUSIONS Incidentally found TTNtvs are common among clinical exome sequencing referrals. Pathological hotspots within the A-band of TTN may be informative in determining variant pathogenicity when incorporated into current American College of Medical Genetics and Genomics guidelines.
Collapse
Affiliation(s)
- Patrick S. Connell
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine, Houston, TX
| | - Amy M. Berkman
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC
| | - BriAnna M. Souder
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine, Houston, TX
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC
| | - Elisa J. Pirozzi
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC
| | - Julia J. Lovin
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine, Houston, TX
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
- Baylor Genetics Laboratories, Houston, TX
| | - Pengfei Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
- Baylor Genetics Laboratories, Houston, TX
| | - Hari Tunuguntla
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine, Houston, TX
| | - Hugh D. Allen
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine, Houston, TX
| | - Susan W. Denfield
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine, Houston, TX
| | - Jeffrey J. Kim
- Department of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine, Houston, TX
| | - Andrew P. Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC
- Department of Cell Biology, Duke University School of Medicine, Durham, NC
| |
Collapse
|
32
|
Zhao W, Chen Y, Yang W, Han Y, Wang Z, Huang F, Qiu Z, Yang K, Jin W. Effects of Cardiomyocyte-Specific Deletion of STAT3-A Murine Model of Heart Failure With Preserved Ejection Fraction. Front Cardiovasc Med 2020; 7:613123. [PMID: 33365331 PMCID: PMC7750364 DOI: 10.3389/fcvm.2020.613123] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/17/2020] [Indexed: 11/13/2022] Open
Abstract
Aims: There is a high incidence of heart failure with preserved ejection fraction (HFpEF), but the options of treatment are limited. A new animal model of HFpEF is urgently needed for in-depth research on HFpEF. Signal transducer and activator of transcription 3 (STAT3) may affect the passive stiffness of myocardium, which determines cardiac diastolic function. We hypothesized that cardiomyocyte-specific deletion of STAT3 increases cardiac passive stiffness, which results the murine features of HFpEF. Methods and Results: Cardiomyocyte-specific deletion of STAT3 (STAT3cKO) mice was generated by the Cre/FLOXp method. The STAT3cKO mice showed heavier cardiac fibrosis and cardiac hypertrophy comparing with wild-type (WT) mice. Furthermore, STAT3cKO mice showed increased serum brain natriuretic peptide (BNP) level, and growth stimulation expressed gene 2 (ST2) level. Other indicators reflecting cardiac passive stiffness and diastolic function, including end diastolic pressure volume relation, MV A value, MV E value, E/A and E/E' had different fold changes. All these changes were accompanied by decreasing levels of protein kinase G (PKG). Bioinformatic analysis of STAT3cKO mice hearts suggested cGMP-PKG signaling pathway might participate in the pathogenesis of HFpEF by means of adjusting different biological functions. Conclusions: Cardiomyocyte-specific deletion of STAT3 results in a murine HFpEF model which imitates the clinical characteristics partly by affecting cardiac PKG levels. Better understanding of the factors influencing HFpEF may finally provided innovative therapies.
Collapse
Affiliation(s)
- Weilin Zhao
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanjia Chen
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenbo Yang
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanxin Han
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyan Wang
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fanyi Huang
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeping Qiu
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ke Yang
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Jin
- Department of Vascular & Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Michel K, Herwig M, Werner F, Špiranec Spes K, Abeßer M, Schuh K, Dabral S, Mügge A, Baba HA, Skryabin BV, Hamdani N, Kuhn M. C-type natriuretic peptide moderates titin-based cardiomyocyte stiffness. JCI Insight 2020; 5:139910. [PMID: 33055420 PMCID: PMC7710274 DOI: 10.1172/jci.insight.139910] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Heart failure is often accompanied by titin-dependent myocardial stiffness. Phosphorylation of titin by cGMP-dependent protein kinase I (PKGI) increases cardiomyocyte distensibility. The upstream pathways stimulating PKGI-mediated titin phosphorylation are unclear. We studied whether C-type natriuretic peptide (CNP), via its guanylyl cyclase-B (GC-B) receptor and cGMP/PKGI signaling, modulates titin-based ventricular compliance. To dissect GC-B–mediated effects of endogenous CNP in cardiomyocytes, we generated mice with cardiomyocyte-restricted GC-B deletion (CM GC-B–KO mice). The impact on heart morphology and function, myocyte passive tension, and titin isoform expression and phosphorylation was studied at baseline and after increased afterload induced by transverse aortic constriction (TAC). Pressure overload increased left ventricular endothelial CNP expression, with an early peak after 3 days. Concomitantly, titin phosphorylation at Ser4080, the site phosphorylated by PKGI, was augmented. Notably, in CM GC-B–KO mice this titin response was abolished. TAC-induced hypertrophy and fibrosis were not different between genotypes. However, the KO mice presented mild systolic and diastolic dysfunction together with myocyte stiffness, which were not observed in control littermates. In vitro, recombinant PKGI rescued reduced titin-Ser4080 phosphorylation and reverted passive stiffness of GC-B–deficient cardiomyocytes. CNP-induced activation of GC-B/cGMP/PKGI signaling in cardiomyocytes provides a protecting regulatory circuit preventing titin-based myocyte stiffening during early phases of pressure overload. C-type natriuretic peptide via GC-B/cGMP/PKGI signalling in cardiomyocytes attenuates titin-based cardiomyocyte stiffening during early phases of pressure-overload.
Collapse
Affiliation(s)
- Konstanze Michel
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Melissa Herwig
- Institute of Physiology and.,Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Franziska Werner
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | | | - Marco Abeßer
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Kai Schuh
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Swati Dabral
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Andreas Mügge
- Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Hideo A Baba
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Boris V Skryabin
- Medical Faculty, Core Facility TRAnsgenic animal and genetic engineering Models (TRAM), University of Münster, Münster, Germany
| | - Nazha Hamdani
- Institute of Physiology and.,Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
34
|
Lim TB, Lavenniah A, Foo RSY. Circles in the heart and cardiovascular system. Cardiovasc Res 2020; 116:269-278. [PMID: 31552406 DOI: 10.1093/cvr/cvz227] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022] Open
Abstract
The combination of next-generation sequencing, advanced bioinformatics analysis, and molecular research has now established circular RNAs (circRNAs) as a heterogeneous group of non-coding RNA that is widely and abundantly expressed. CircRNAs are single-stranded RNA, covalently backspliced to form closed circular loops. Different models of back-splicing have been proposed, and mechanisms for circRNA function include sequestering microRNAs, direct interaction with proteins, regulation of transcription, and translation. Exploring the role of circRNAs in different disease settings, and understanding how they contribute to disease progression promises to provide valuable insight into potential novel therapeutic approaches. Here, we review the growing number of published research on circRNAs in the heart and cardiovascular system and summarize the circRNAs that have been implicated in disease.
Collapse
Affiliation(s)
- Tingsen Benson Lim
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore 117599, Singapore.,Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore 138672, Singapore
| | - Annadoray Lavenniah
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore 117599, Singapore.,Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore 138672, Singapore
| | - Roger Sik-Yin Foo
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore 117599, Singapore.,Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore 138672, Singapore
| |
Collapse
|
35
|
Rich KA, Moscarello T, Siskind C, Brock G, Tan CA, Vatta M, Winder TL, Elsheikh B, Vicini L, Tucker B, Palettas M, Hershberger RE, Kissel JT, Morales A, Roggenbuck J. Novel heterozygous truncating titin variants affecting the A-band are associated with cardiomyopathy and myopathy/muscular dystrophy. Mol Genet Genomic Med 2020; 8:e1460. [PMID: 32815318 PMCID: PMC7549586 DOI: 10.1002/mgg3.1460] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/10/2020] [Accepted: 07/31/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Variants in TTN are frequently identified in the genetic evaluation of skeletal myopathy or cardiomyopathy. However, due to the high frequency of TTN variants in the general population, incomplete penetrance, and limited understanding of the spectrum of disease, interpretation of TTN variants is often difficult for laboratories and clinicians. Currently, cardiomyopathy is associated with heterozygous A-band TTN variants, whereas skeletal myopathy is largely associated with homozygous or compound heterozygous TTN variants. Recent reports show pathogenic variants in TTN may result in a broader phenotypic spectrum than previously recognized. METHODS Here we report the results of a multisite study that characterized the phenotypes of probands with variants in TTN. We investigated TTN genotype-phenotype correlations in probands with skeletal myopathy and/or cardiomyopathy. Probands with TTN truncating variants (TTNtv) or pathogenic missense variants were ascertained from two academic medical centers. Variants were identified via clinical genetic testing and reviewed according to the American College of Medical Genetics criteria. Clinical and family history data were documented via retrospective chart review. Family studies were performed for probands with atypical phenotypes. RESULTS Forty-nine probands were identified with TTNtv or pathogenic missense variants. Probands were classified by clinical presentation: cardiac (n = 30), skeletal muscle (n = 12), or both (cardioskeletal, n = 7). Within the cardioskeletal group, 5/7 probands had heterozygous TTNtv predicted to affect the distal (3') end of the A-band. All cardioskeletal probands had onset of proximal-predominant muscle weakness before diagnosis of cardiovascular disease, five pedigrees support dominant transmission. CONCLUSION Although heterozygous TTNtv in the A-band is known to cause dilated cardiomyopathy, we present evidence that these variants may in some cases cause a novel, dominant skeletal myopathy with a limb-girdle pattern of weakness. These findings emphasize the importance of multidisciplinary care for patients with A-band TTNtv who may be at risk for multisystem disease.
Collapse
Affiliation(s)
- Kelly A Rich
- The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Tia Moscarello
- Stanford Center for Inherited Cardiovascular Disease, Stanford University, Stanford, CA, USA
| | - Carly Siskind
- Stanford Health Care, Stanford University, Stanford, CA, USA
| | - Guy Brock
- The Ohio State University Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | | | | | | | - Bakri Elsheikh
- The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Leah Vicini
- The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Brianna Tucker
- Stanford Center for Inherited Cardiovascular Disease, Stanford University, Stanford, CA, USA
| | - Marilly Palettas
- The Ohio State University Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Ray E Hershberger
- The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - John T Kissel
- The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Ana Morales
- The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, USA.,Invitae Corporation, San Francisco, CA, USA
| | - Jennifer Roggenbuck
- The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
36
|
Brayson D, Holohan S, Bardswell SC, Arno M, Lu H, Jensen HK, Tran PK, Barallobre‐Barreiro J, Mayr M, dos Remedios CG, Tsang VT, Frigiola A, Kentish JC. Right Ventricle Has Normal Myofilament Function But Shows Perturbations in the Expression of Extracellular Matrix Genes in Patients With Tetralogy of Fallot Undergoing Pulmonary Valve Replacement. J Am Heart Assoc 2020; 9:e015342. [PMID: 32805183 PMCID: PMC7660801 DOI: 10.1161/jaha.119.015342] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/28/2020] [Indexed: 02/06/2023]
Abstract
Background Patients with repair of tetralogy of Fallot (rToF) who are approaching adulthood often exhibit pulmonary valve regurgitation, leading to right ventricle (RV) dilatation and dysfunction. The regurgitation can be corrected by pulmonary valve replacement (PVR), but the optimal surgical timing remains under debate, mainly because of the poorly understood nature of RV remodeling in patients with rToF. The goal of this study was to probe for pathologic molecular, cellular, and tissue changes in the myocardium of patients with rToF at the time of PVR. Methods and Results We measured contractile function of permeabilized myocytes, collagen content of tissue samples, and the expression of mRNA and selected proteins in RV tissue samples from patients with rToF undergoing PVR for severe pulmonary valve regurgitation. The data were compared with nondiseased RV tissue from unused donor hearts. Contractile performance and passive stiffness of the myofilaments in permeabilized myocytes were similar in rToF-PVR and RV donor samples, as was collagen content and cross-linking. The patients with rToF undergoing PVR had enhanced mRNA expression of genes associated with connective tissue diseases and tissue remodeling, including the small leucine-rich proteoglycans ASPN (asporin), LUM (lumican), and OGN (osteoglycin), although their protein levels were not significantly increased. Conclusions RV myofilaments from patients with rToF undergoing PVR showed no functional impairment, but the changes in extracellular matrix gene expression may indicate the early stages of remodeling. Our study found no evidence of major damage at the cellular and tissue levels in the RV of patients with rToF who underwent PVR according to current clinical criteria.
Collapse
Affiliation(s)
- Daniel Brayson
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| | - So‐Jin Holohan
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| | - Sonya C. Bardswell
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| | - Matthew Arno
- Genomics CentreFaculty of Life Sciences and MedicineKing’s College LondonLondonUnited Kingdom
| | - Han Lu
- Genomics CentreFaculty of Life Sciences and MedicineKing’s College LondonLondonUnited Kingdom
| | | | | | - Javier Barallobre‐Barreiro
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| | - Manuel Mayr
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| | | | | | - Alessandra Frigiola
- Great Ormond Street HospitalLondonUnited Kingdom
- Guys and St Thomas’ NHS Foundation TrustSt Thomas’ HospitalLondonUnited Kingdom
- School of Biomedical Engineering and Imaging SciencesKings CollegeLondonUnited Kingdom
| | - Jonathan C. Kentish
- School of Cardiovascular Medicine and SciencesKing's College London BHF Centre for Research ExcellenceLondonUnited Kingdom
| |
Collapse
|
37
|
Nagueh SF. Heart failure with preserved ejection fraction: insights into diagnosis and pathophysiology. Cardiovasc Res 2020; 117:999-1014. [PMID: 32717061 DOI: 10.1093/cvr/cvaa228] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/15/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for at least half the cases of heart failure, currently diagnosed. There are several cardiac and non-cardiac manifestations of the syndrome. Structure and function abnormalities can include all four cardiac chambers. The left ventricle has abnormal systolic and diastolic functions which can be examined by invasive and non-invasive measurements. In addition, the left atrium enlarges with abnormal left atrial function, pulmonary hypertension occurs, and the right ventricle can develop hypertrophy, enlargement, and systolic dysfunction. There are a paucity of data on calcium handling in HFpEF patients. Growing literature supports the presence of abnormalities in titin and its phosphorylation, and increased interstitial fibrosis contributing to increased chamber stiffness. A systemic inflammatory state causing reduced myocardial cyclic guanosine monophosphate along with defects in the unfolded protein response have been recently reported. Diagnosis relies on signs and symptoms of heart failure, preserved ejection fraction, and detection of diastolic function abnormalities based on echocardiographic findings and abnormally elevated natriuretic peptide levels or invasive measurements of wedge pressure at rest or with exercise. There are currently two diagnostic algorithms: H2FPEF, and HFA-PEFF with limited data comparing their performance head to head in the same patient population. Despite the growing understanding of the syndrome's pathophysiology, there have been little success in developing specific treatment for patients with HFpEF.
Collapse
Affiliation(s)
- Sherif F Nagueh
- Methodist DeBakey Heart and Vascular Center, 6550 Fannin, SM-1801, Houston, TX 77030, USA
| |
Collapse
|
38
|
POPDC2 a novel susceptibility gene for conduction disorders. J Mol Cell Cardiol 2020; 145:74-83. [PMID: 32535041 DOI: 10.1016/j.yjmcc.2020.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/22/2020] [Accepted: 06/09/2020] [Indexed: 01/25/2023]
Abstract
Despite recent progress in the understanding of cardiac ion channel function and its role in inherited forms of ventricular arrhythmias, the molecular basis of cardiac conduction disorders often remains unresolved. We aimed to elucidate the genetic background of familial atrioventricular block (AVB) using a whole exome sequencing (WES) approach. In monozygotic twins with a third-degree AVB and in another, unrelated family with first-degree AVB, we identified a heterozygous nonsense mutation in the POPDC2 gene causing a premature stop at position 188 (POPDC2W188⁎), deleting parts of its cAMP binding-domain. Popeye-domain containing (POPDC) proteins are predominantly expressed in the skeletal muscle and the heart, with particularly high expression of POPDC2 in the sinoatrial node of the mouse. We now show by quantitative PCR experiments that in the human heart the POPDC-modulated two-pore domain potassium (K2P) channel TREK-1 is preferentially expressed in the atrioventricular node. Co-expression studies in Xenopus oocytes revealed that POPDC2W188⁎ causes a loss-of-function with impaired TREK-1 modulation. Consistent with the high expression level of POPDC2 in the murine sinoatrial node, POPDC2W188⁎ knock-in mice displayed stress-induced sinus bradycardia and pauses, a phenotype that was previously also reported for POPDC2 and TREK-1 knock-out mice. We propose that the POPDC2W188⁎ loss-of-function mutation contributes to AVB pathogenesis by an aberrant modulation of TREK-1, highlighting that POPDC2 represents a novel arrhythmia gene for cardiac conduction disorders.
Collapse
|
39
|
Fochi S, Lorenzi P, Galasso M, Stefani C, Trabetti E, Zipeto D, Romanelli MG. The Emerging Role of the RBM20 and PTBP1 Ribonucleoproteins in Heart Development and Cardiovascular Diseases. Genes (Basel) 2020; 11:genes11040402. [PMID: 32276354 PMCID: PMC7230170 DOI: 10.3390/genes11040402] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022] Open
Abstract
Alternative splicing is a regulatory mechanism essential for cell differentiation and tissue organization. More than 90% of human genes are regulated by alternative splicing events, which participate in cell fate determination. The general mechanisms of splicing events are well known, whereas only recently have deep-sequencing, high throughput analyses and animal models provided novel information on the network of functionally coordinated, tissue-specific, alternatively spliced exons. Heart development and cardiac tissue differentiation require thoroughly regulated splicing events. The ribonucleoprotein RBM20 is a key regulator of the alternative splicing events required for functional and structural heart properties, such as the expression of TTN isoforms. Recently, the polypyrimidine tract-binding protein PTBP1 has been demonstrated to participate with RBM20 in regulating splicing events. In this review, we summarize the updated knowledge relative to RBM20 and PTBP1 structure and molecular function; their role in alternative splicing mechanisms involved in the heart development and function; RBM20 mutations associated with idiopathic dilated cardiovascular disease (DCM); and the consequences of RBM20-altered expression or dysfunction. Furthermore, we discuss the possible application of targeting RBM20 in new approaches in heart therapies.
Collapse
|
40
|
Ahmed Z, Zeeshan S, Mendhe D, Dong X. Human gene and disease associations for clinical-genomics and precision medicine research. Clin Transl Med 2020; 10:297-318. [PMID: 32508008 PMCID: PMC7240856 DOI: 10.1002/ctm2.28] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/15/2022] Open
Abstract
We are entering the era of personalized medicine in which an individual's genetic makeup will eventually determine how a doctor can tailor his or her therapy. Therefore, it is becoming critical to understand the genetic basis of common diseases, for example, which genes predispose and rare genetic variants contribute to diseases, and so on. Our study focuses on helping researchers, medical practitioners, and pharmacists in having a broad view of genetic variants that may be implicated in the likelihood of developing certain diseases. Our focus here is to create a comprehensive database with mobile access to all available, authentic and actionable genes, SNPs, and classified diseases and drugs collected from different clinical and genomics databases worldwide, including Ensembl, GenCode, ClinVar, GeneCards, DISEASES, HGMD, OMIM, GTR, CNVD, Novoseek, Swiss-Prot, LncRNADisease, Orphanet, GWAS Catalog, SwissVar, COSMIC, WHO, and FDA. We present a new cutting-edge gene-SNP-disease-drug mobile database with a smart phone application, integrating information about classified diseases and related genes, germline and somatic mutations, and drugs. Its database includes over 59 000 protein-coding and noncoding genes; over 67 000 germline SNPs and over a million somatic mutations reported for over 19 000 protein-coding genes located in over 1000 regions, published with over 3000 articles in over 415 journals available at the PUBMED; over 80 000 ICDs; over 123 000 NDCs; and over 100 000 classified gene-SNP-disease associations. We present an application that can provide new insights into the information about genetic basis of human complex diseases and contribute to assimilating genomic with phenotypic data for the availability of gene-based designer drugs, precise targeting of molecular fingerprints for tumor, appropriate drug therapy, predicting individual susceptibility to disease, diagnosis, and treatment of rare illnesses are all a few of the many transformations expected in the decade to come.
Collapse
Affiliation(s)
- Zeeshan Ahmed
- Institute for Health, Health Care Policy and Aging Research, RutgersThe State University of New JerseyNew BrunswickNew JerseyUSA
- Department of Medicine, Rutgers Robert Wood Johnson Medical SchoolRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Saman Zeeshan
- Rutgers Cancer Institute of New Jersey, RutgersThe State University of New JerseyNew BrunswickNew JerseyUSA
| | - Dinesh Mendhe
- Institute for Health, Health Care Policy and Aging Research, RutgersThe State University of New JerseyNew BrunswickNew JerseyUSA
| | - XinQi Dong
- Institute for Health, Health Care Policy and Aging Research, RutgersThe State University of New JerseyNew BrunswickNew JerseyUSA
- Department of Medicine, Rutgers Robert Wood Johnson Medical SchoolRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| |
Collapse
|
41
|
Tharp CA, Haywood ME, Sbaizero O, Taylor MRG, Mestroni L. The Giant Protein Titin's Role in Cardiomyopathy: Genetic, Transcriptional, and Post-translational Modifications of TTN and Their Contribution to Cardiac Disease. Front Physiol 2019; 10:1436. [PMID: 31849696 PMCID: PMC6892752 DOI: 10.3389/fphys.2019.01436] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure, sudden cardiac death and heart transplant. DCM is inherited in approximately 50% of cases, in which the most frequent genetic defects are truncation variants of the titin gene (TTNtv). TTN encodes titin, which is the largest protein in the body and is an essential component of the sarcomere. Titin serves as a biological spring, spanning half of the sarcomere and connecting the Z-disk to the M-line, with scaffold and signaling functions. Truncations of titin are believed to lead to either haploinsufficiency and loss-of-function, or to a “poison peptide” effect. However, other titin mechanisms are postulated to influence cardiac function including post-translational modifications, in particular changes in titin phosphorylation that alters the stiffness of the protein, and diversity of alternative splicing that generates different titin isoforms. In this article, we review the role of TTN mutations in development of DCM, how differential expression of titin isoforms relate to DCM pathophysiology, and discuss how post-translational modifications of titin can affect cardiomyocyte function. Current research efforts aim to elucidate the contribution of titin to myofibril assembly, stability, and signal transduction, and how mutant titin leads to cardiac dysfunction and human disease. Future research will need to translate this knowledge toward novel therapeutic approaches that can modulate titin transcriptional and post-translational defects to treat DCM and heart failure.
Collapse
Affiliation(s)
- Charles A Tharp
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mary E Haywood
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Matthew R G Taylor
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Luisa Mestroni
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
42
|
Ahmed Z, Zeeshan S, Xiong R, Liang BT. Debutant iOS app and gene-disease complexities in clinical genomics and precision medicine. Clin Transl Med 2019; 8:26. [PMID: 31586224 PMCID: PMC6778157 DOI: 10.1186/s40169-019-0243-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023] Open
Abstract
Background The last decade has seen a dramatic increase in the availability of scientific data, where human-related biological databases have grown not only in count but also in volume, posing unprecedented challenges in data storage, processing, analysis, exchange, and curation. Next generation sequencing (NGS) advancements have facilitated and accelerated the process of identifying genetic variations. Adopting NGS with Whole-Genome and RNA sequencing in a diagnostic context has the potential to improve disease-risk detection in support of precision medicine and drug discovery. Several bioinformatics pipelines have been developed to strengthen variant interpretation by efficiently processing and analyzing sequence data, whereas many published results show how genomics data can be proactively incorporated into medical practices and improve utilization of clinical information. To utilize the wealth of genomics and health, there is a crucial need to generate appropriate gene-disease annotation repositories accessed through modern technology. Results Our focus here is to create a comprehensive database with mobile access to actionable genes and classified diseases, considered the foundation for clinical genomics and precision medicine. We present a publicly available iOS app, PAS-Gen, which invites global users to freely download it on iPhone and iPad devices, quickly adopt its easy to use interface, and search for genes and related diseases. PAS-Gen was developed using Swift, XCODE, and PHP scripting that uses Web and MySQL database servers, which includes over 59,000 protein-coding and non-coding genes, and over 90,000 classified gene-disease associations. PAS-Gen is founded on the clinical and scientific premise that easier healthcare and genomics data sharing will accelerate future medical discoveries. Conclusions We present a cutting-edge gene-disease database with a smart phone application, integrating information on classified diseases and related genes. The PAS-Gen app will assist researchers, medical practitioners, and pharmacists by providing a broad and view of genes that may be implicated in the likelihood of developing certain diseases. This tool with accelerate users’ abilities to understand the genetic basis of human complex diseases and by assimilating genomic and phenotypic data will support future work to identify gene-specific designer drugs, target precise molecular fingerprints for tumors, suggest appropriate drug therapies, predict individual susceptibility to disease, and diagnose and treat rare illnesses.
Collapse
Affiliation(s)
- Zeeshan Ahmed
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center (UConn Health), 263 Farmington Ave, Farmington, CT, 06032, USA. .,Institute for Systems Genomics, University of Connecticut, 263 Farmington Ave, Farmington, CT, 06032, USA.
| | - Saman Zeeshan
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Ruoyun Xiong
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center (UConn Health), 263 Farmington Ave, Farmington, CT, 06032, USA.,The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Bruce T Liang
- Pat and Jim Calhoun Cardiology Center, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT, 06032, USA
| |
Collapse
|
43
|
Junttila MJ, Holmström L, Pylkäs K, Mantere T, Kaikkonen K, Porvari K, Kortelainen ML, Pakanen L, Kerkelä R, Myerburg RJ, Huikuri HV. Primary Myocardial Fibrosis as an Alternative Phenotype Pathway of Inherited Cardiac Structural Disorders. Circulation 2019; 137:2716-2726. [PMID: 29915098 DOI: 10.1161/circulationaha.117.032175] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/15/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Myocardial fibrosis is a common postmortem finding among young individuals with sudden cardiac death. Because there is no known single cause, we tested the hypothesis that some cases of myocardial fibrosis in the absence of identifiable causes (primary myocardial fibrosis [PMF]) are associated with genetic variants. METHODS Tissue was obtained at autopsy from 4031 consecutive individuals with sudden cardiac death in Northern Finland, among whom PMF was the only structural finding in 145 subjects with sudden cardiac death. We performed targeted next-generation sequencing using a panel of 174 genes associated with myocardial structure and ion channel function when autopsies did not identify a secondary basis for myocardial fibrosis. All variants with an effect on protein and with a minor allele frequency <0.01 were classified as pathogenic or variants of uncertain significance on the basis of American College of Medical Genetics consensus guidelines. RESULTS Among the 96 specimens with DNA passing quality control (66%), postmortem genetic tests identified 24 variants of known or uncertain significance in 26 subjects (27%). Ten were pathogenic/likely pathogenic variants in 10 subjects (10%), and 14 were variants of uncertain significance in 11 genes among 16 subjects (17%). Five variants were in genes associated with arrhythmogenic right ventricular cardiomyopathy, 6 in hypertrophic cardiomyopathy-associated genes, and 11 in dilated cardiomyopathy-associated genes; 2 were not associated with these disorders. Four unique variants of uncertain significance cosegregated among multiple unrelated subjects with PMF. No pathogenic/likely pathogenic variants were detected in ion channel-encoding genes. CONCLUSIONS A large proportion of subjects with PMF at autopsy had variants in genes associated with arrhythmogenic right ventricular cardiomyopathy, dilated cardiomyopathy, and hypertrophic cardiomyopathy without autopsy findings of those diseases, suggesting that PMF can be an alternative phenotypic expression of structural disease-associated genetic variants or that risk-associated fibrosis was expressing before the primary disease. These findings have clinical implications for postmortem genetic testing and family risk profiling.
Collapse
Affiliation(s)
- M Juhani Junttila
- Research Unit of Internal Medicine, University of Oulu and University Hospital of Oulu, Finland (M.J.J., L.H., K.K., H.V.H.)
| | - Lauri Holmström
- Research Unit of Internal Medicine, University of Oulu and University Hospital of Oulu, Finland (M.J.J., L.H., K.K., H.V.H.)
| | - Katri Pylkäs
- Laboratory of Cancer Genetics and Tumor Biology, Cancer and Translational Medicine Research Unit and Biocenter Oulu (K. Pylkäs, T.M.)
| | - Tuomo Mantere
- Laboratory of Cancer Genetics and Tumor Biology, Cancer and Translational Medicine Research Unit and Biocenter Oulu (K. Pylkäs, T.M.)
| | - Kari Kaikkonen
- Research Unit of Internal Medicine, University of Oulu and University Hospital of Oulu, Finland (M.J.J., L.H., K.K., H.V.H.)
| | - Katja Porvari
- Department of Forensic Medicine, Research Unit of Internal Medicine, Medical Research Center Oulu (K, Porvari, M.-L.K., L.P.)
| | - Marja-Leena Kortelainen
- Department of Forensic Medicine, Research Unit of Internal Medicine, Medical Research Center Oulu (K, Porvari, M.-L.K., L.P.)
| | - Lasse Pakanen
- Department of Forensic Medicine, Research Unit of Internal Medicine, Medical Research Center Oulu (K, Porvari, M.-L.K., L.P.).,National Institute for Health and Welfare, Forensic Medicine Unit, Oulu, Finland (L.P.)
| | - Risto Kerkelä
- Research Unit of Biomedicine (R.K.), University of Oulu, Finland
| | - Robert J Myerburg
- Division of Cardiology, University of Miami Miller School of Medicine, FL (R.J.M.)
| | - Heikki V Huikuri
- Research Unit of Internal Medicine, University of Oulu and University Hospital of Oulu, Finland (M.J.J., L.H., K.K., H.V.H.)
| |
Collapse
|
44
|
Najafi A, van de Locht M, Schuldt M, Schönleitner P, van Willigenburg M, Bollen I, Goebel M, Ottenheijm CAC, van der Velden J, Helmes M, Kuster DWD. End-diastolic force pre-activates cardiomyocytes and determines contractile force: role of titin and calcium. J Physiol 2019; 597:4521-4531. [PMID: 31314138 PMCID: PMC6852589 DOI: 10.1113/jp277985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/10/2019] [Indexed: 12/22/2022] Open
Abstract
Titin functions as a molecular spring, and cardiomyocytes are able, through splicing, to control the length of titin. We hypothesized that together with diastolic [Ca2+], titin‐based stretch pre‐activates cardiomyocytes during diastole and is a major determinant of force production in the subsequent contraction. Through this mechanism titin would play an important role in active force development and length‐dependent activation. Mutations in the splicing factor RNA binding motif protein 20 (RBM20) result in expression of large, highly compliant titin isoforms. We measured single cardiomyocyte work loops that mimic the cardiac cycle in wild‐type (WT) and heterozygous (HET) RBM20‐deficient rats. In addition, we studied the role of diastolic [Ca2+] in membrane‐permeabilized WT and HET cardiomyocytes. Intact cardiomyocytes isolated from HET left ventricles were unable to produce normal levels of work (55% of WT) at low pacing frequencies, but this difference disappeared at high pacing frequencies. Length‐dependent activation (force–sarcomere length relationship) was blunted in HET cardiomyocytes, but the force–end‐diastolic force relationship was not different between HET and WT cardiomyocytes. To delineate the effects of diastolic [Ca2+] and titin pre‐activation on force generation, measurements were performed in detergent‐permeabilized cardiomyocytes. Cardiac twitches were simulated by transiently exposing permeabilized cardiomyocytes to 2 µm Ca2+. Increasing diastolic [Ca2+] from 1 to 80 nm increased force development twofold in WT. Higher diastolic [Ca2+] was needed in HET. These findings are consistent with our hypothesis that pre‐activation increases active force development. Highly compliant titin allows cells to function at higher diastolic [Ca2+].
Collapse
Affiliation(s)
- Aref Najafi
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands.,Netherlands Heart Institute, PO box 19258, 3501 DG, Utrecht, the Netherlands
| | - Martijn van de Locht
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Maike Schuldt
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | | | | | - Ilse Bollen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Max Goebel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Coen A C Ottenheijm
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands.,Netherlands Heart Institute, PO box 19258, 3501 DG, Utrecht, the Netherlands
| | - Michiel Helmes
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands.,Ionoptix, de Boelelaan 1108, 1081 HV, Amsterdam, the Netherlands.,CytoCypher, de Boelelaan 1108, 1081 HV, Amsterdam, the Netherlands
| | - Diederik W D Kuster
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| |
Collapse
|
45
|
Tonino P, Kiss B, Gohlke J, Smith JE, Granzier H. Fine mapping titin's C-zone: Matching cardiac myosin-binding protein C stripes with titin's super-repeats. J Mol Cell Cardiol 2019; 133:47-56. [PMID: 31158359 DOI: 10.1016/j.yjmcc.2019.05.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 01/04/2023]
Abstract
Titin is largely comprised of serially-linked immunoglobulin (Ig) and fibronectin type-III (Fn3) domains. Many of these domains are arranged in an 11 domain super-repeat pattern that is repeated 11 times, forming the so-named titin C-zone in the A-band region of the sarcomere. Each super-repeat is thought to provide binding sites for thick filament proteins, such as cMyBP-C (cardiac myosin-binding protein C). However, it remains to be established which of titin's 11 C-zone super-repeats anchor cMyBP-C as titin contains 11 super-repeats and cMyBP-C is found in 9 stripes only. To study the layout of titin's C-zone in relation to MyBP-C, immunolabeling studies were performed on mouse skinned myocardium with antibodies to titin and cMyBP-C, using both immuno-electron microscopy and super-resolution optical microscopy. Results indicate that cMyBP-C locates near the interface between titin's C-zone super-repeats. Studies on a mouse model in which two of titin's C-zone repeats have been genetically deleted support that the first Ig domain of a super-repeat is important for anchoring cMyBP-C but also Fn3 domains located at the end of the preceding repeat. Furthermore, not all super-repeat interfaces are equal as the interface between super-repeat 1 and 2 (close to titin's D-zone) does not contain cMyBP-C. Finally, titin's C-zone does not extend all the way to the bare zone but instead terminates at the level of the second myosin crown. This study enhances insights in the molecular layout of the C-zone of titin, its relation to cMyBP-C, and its possible roles in cardiomyopathies.
Collapse
Affiliation(s)
- Paola Tonino
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - Balazs Kiss
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - Jochen Gohlke
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - John E Smith
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
46
|
van der Pijl RJ, Granzier HL, Ottenheijm CAC. Diaphragm contractile weakness due to reduced mechanical loading: role of titin. Am J Physiol Cell Physiol 2019; 317:C167-C176. [PMID: 31042425 DOI: 10.1152/ajpcell.00509.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The diaphragm, the main muscle of inspiration, is constantly subjected to mechanical loading. Only during controlled mechanical ventilation, as occurs during thoracic surgery and in the intensive care unit, is mechanical loading of the diaphragm arrested. Animal studies indicate that the diaphragm is highly sensitive to unloading, causing rapid muscle fiber atrophy and contractile weakness; unloading-induced diaphragm atrophy and contractile weakness have been suggested to contribute to the difficulties in weaning patients from ventilator support. The molecular triggers that initiate the rapid unloading atrophy of the diaphragm are not well understood, although proteolytic pathways and oxidative signaling have been shown to be involved. Mechanical stress is known to play an important role in the maintenance of muscle mass. Within the muscle's sarcomere, titin is considered to play an important role in the stress-response machinery. Titin is a giant protein that acts as a mechanosensor regulating muscle protein expression in a sarcomere strain-dependent fashion. Thus titin is an attractive candidate for sensing the sudden mechanical arrest of the diaphragm when patients are mechanically ventilated, leading to changes in muscle protein expression. Here, we provide a novel perspective on how titin and its biomechanical sensing and signaling might be involved in the development of mechanical unloading-induced diaphragm weakness.
Collapse
Affiliation(s)
- Robbert J van der Pijl
- Department of Cellular and Molecular Medicine, University of Arizona , Tucson, Arizona.,Department of Physiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Henk L Granzier
- Department of Cellular and Molecular Medicine, University of Arizona , Tucson, Arizona
| | - Coen A C Ottenheijm
- Department of Cellular and Molecular Medicine, University of Arizona , Tucson, Arizona.,Department of Physiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Zeeshan S, Xiong R, Liang BT, Ahmed Z. 100 Years of evolving gene-disease complexities and scientific debutants. Brief Bioinform 2019; 21:885-905. [PMID: 30972412 DOI: 10.1093/bib/bbz038] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/06/2019] [Accepted: 03/08/2019] [Indexed: 12/22/2022] Open
Abstract
It's been over 100 years since the word `gene' is around and progressively evolving in several scientific directions. Time-to-time technological advancements have heavily revolutionized the field of genomics, especially when it's about, e.g. triple code development, gene number proposition, genetic mapping, data banks, gene-disease maps, catalogs of human genes and genetic disorders, CRISPR/Cas9, big data and next generation sequencing, etc. In this manuscript, we present the progress of genomics from pea plant genetics to the human genome project and highlight the molecular, technical and computational developments. Studying genome and epigenome led to the fundamentals of development and progression of human diseases, which includes chromosomal, monogenic, multifactorial and mitochondrial diseases. World Health Organization has classified, standardized and maintained all human diseases, when many academic and commercial online systems are sharing information about genes and linking to associated diseases. To efficiently fathom the wealth of this biological data, there is a crucial need to generate appropriate gene annotation repositories and resources. Our focus has been how many gene-disease databases are available worldwide and which sources are authentic, timely updated and recommended for research and clinical purposes. In this manuscript, we have discussed and compared 43 such databases and bioinformatics applications, which enable users to connect, explore and, if possible, download gene-disease data.
Collapse
Affiliation(s)
- Saman Zeeshan
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, USA
| | - Ruoyun Xiong
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington Ave, Farmington, CT, USA
| | - Bruce T Liang
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington Ave, Farmington, CT, USA.,Pat and Jim Calhoun Cardiology Center, School of Medicine, University of Connecticut Health Center, Farmington Ave, Farmington, CT, USA
| | - Zeeshan Ahmed
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington Ave, Farmington, CT, USA
| |
Collapse
|
48
|
Misaka T, Yoshihisa A, Takeishi Y. Titin in muscular dystrophy and cardiomyopathy: Urinary titin as a novel marker. Clin Chim Acta 2019; 495:123-128. [PMID: 30959043 DOI: 10.1016/j.cca.2019.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 01/12/2023]
Abstract
Titin, encoded by the gene TTN, is the largest human protein, and plays central roles in sarcomeric structures and functions in skeletal and cardiac muscles. Mutations of TTN are causally related to specific types of muscular dystrophies and cardiomyopathies. A developed methodology of next generation sequencing has recently led to the identification of novel TTN mutations in such diseases. The clinical significance of titin is now emerging as a target for genetic strategies. Titin-related muscular dystrophies include tibial muscular dystrophy, limb-girdle muscular dystrophy, Emery-Dreifuss muscular dystrophy, hereditary myopathy with early respiratory failure, central core myopathy, centronuclear myopathies, and Salih myopathy. Truncation mutations of TTN have been identified as the most frequent genetic cause of dilated cardiomyopathy. In this review article, we highlight the role of titin and impact of TTN mutations in the pathogenesis of muscular dystrophies and cardiomyopathies. Recently, a novel sensitive sandwich enzyme-linked immunosorbent assay (ELISA) for the detection of the urinary titin N-terminal fragments (U-TN) has been established. We discuss the clinical significance of U-TN in the diagnosis of muscular dystrophies and differential diagnosis of cardiomyopathies, as well as risk stratification in dilated cardiomyopathy.
Collapse
Affiliation(s)
- Tomofumi Misaka
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan.; Department of Advanced Cardiac Therapeutics, Fukushima Medical University, Fukushima, Japan
| | - Akiomi Yoshihisa
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan.; Department of Advanced Cardiac Therapeutics, Fukushima Medical University, Fukushima, Japan..
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
49
|
Mijailovich SM, Stojanovic B, Nedic D, Svicevic M, Geeves MA, Irving TC, Granzier HL. Nebulin and titin modulate cross-bridge cycling and length-dependent calcium sensitivity. J Gen Physiol 2019; 151:680-704. [PMID: 30948421 PMCID: PMC6504291 DOI: 10.1085/jgp.201812165] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 01/15/2019] [Accepted: 03/03/2019] [Indexed: 12/13/2022] Open
Abstract
Various mutations in the structural proteins nebulin and titin that are present in human disease are known to affect the contractility of striated muscle. Loss of nebulin is associated with reduced actin filament length and impairment of myosin binding to actin, whereas titin is thought to regulate muscle passive elasticity and is likely involved in length-dependent activation. Here, we sought to assess the modulation of muscle function by these sarcomeric proteins by using the computational platform muscle simulation code (MUSICO) to quantitatively separate the effects of structural changes, kinetics of cross-bridge cycling, and calcium sensitivity of the thin filaments. The simulations show that variation in thin filament length cannot by itself account for experimental observations of the contractility in nebulin-deficient muscle, but instead must be accompanied by a decreased myosin binding rate. Additionally, to match the observed calcium sensitivity, the rate of TnI detachment from actin needed to be increased. Simulations for cardiac muscle provided quantitative estimates of the effects of different titin-based passive elasticities on muscle force and activation in response to changes in sarcomere length and interfilament lattice spacing. Predicted force-pCa relations showed a decrease in both active tension and sensitivity to calcium with a decrease in passive tension and sarcomere length. We conclude that this behavior is caused by partial redistribution of the muscle load between active muscle force and titin-dependent passive force, and also by redistribution of stretch along the thin filament, which together modulate the release of TnI from actin. These data help advance understanding of how nebulin and titin mutations affect muscle function.
Collapse
Affiliation(s)
- Srboljub M Mijailovich
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA .,Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL
| | - Boban Stojanovic
- University of Kragujevac, Faculty of Science, Kragujevac, Serbia
| | - Djordje Nedic
- University of Kragujevac, Faculty of Science, Kragujevac, Serbia
| | - Marina Svicevic
- University of Kragujevac, Faculty of Science, Kragujevac, Serbia
| | - Michael A Geeves
- Department of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Thomas C Irving
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL
| | | |
Collapse
|
50
|
Kellermayer D, Smith JE, Granzier H. Titin mutations and muscle disease. Pflugers Arch 2019; 471:673-682. [PMID: 30919088 DOI: 10.1007/s00424-019-02272-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/11/2019] [Indexed: 12/12/2022]
Abstract
The introduction of next-generation sequencing technology has revealed that mutations in the gene that encodes titin (TTN) are linked to multiple skeletal and cardiac myopathies. The most prominent of these myopathies is dilated cardiomyopathy (DCM). Over 60 genes are linked to the etiology of DCM, but by far, the leading cause of DCM is mutations in TTN with truncating variants in TTN (TTNtvs) associated with familial DCM in ∼ 20% of the cases. Titin is a large (3-4 MDa) and abundant protein that forms the third myofilament type of striated muscle where it spans half the sarcomere, from the Z-disk to the M-line. The underlying mechanisms by which titin mutations induce disease are poorly understood and targeted therapies are not available. Here, we review what is known about TTN mutations in muscle disease, with a major focus on DCM. We highlight that exon skipping might provide a possible therapeutic avenue to address diseases that arise from TTNtvs.
Collapse
Affiliation(s)
- Dalma Kellermayer
- Department of Cellular and Molecular Medicine, University of Arizona, MRB 325. 1656 E Mabel Street, Tucson, AZ, 85724-5217, USA.,Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85721, USA
| | - John E Smith
- Department of Cellular and Molecular Medicine, University of Arizona, MRB 325. 1656 E Mabel Street, Tucson, AZ, 85724-5217, USA.,Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85721, USA
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, MRB 325. 1656 E Mabel Street, Tucson, AZ, 85724-5217, USA. .,Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|