211 results on '"Dos Remedios, CG"'
Search Results
2. Tryptophan metabolism to kynurenine is a potential novel contributor to hypotension in human sepsis.
- Author
-
Changsirivathanathamrong D, Wang Y, Rajbhandari D, Maghzal GJ, Mak WM, Woolfe C, Duflou J, Gebski V, Dos Remedios CG, Celermajer DS, and Stocker R
- Published
- 2011
- Full Text
- View/download PDF
3. Differential effects of myosin activators on myocardial contractile function in nonfailing and failing human hearts.
- Author
-
Choi J, Wood PT, Holmes JB, Dominic KL, Dos Remedios CG, Campbell KS, and Stelzer JE
- Subjects
- Humans, Male, Middle Aged, Female, Adult, Myocardium metabolism, Uracil analogs & derivatives, Uracil pharmacology, Calcium metabolism, Myosins metabolism, Aged, Heart Failure physiopathology, Heart Failure drug therapy, Heart Failure metabolism, Myocardial Contraction drug effects, Urea analogs & derivatives, Urea pharmacology
- Abstract
The second-generation myosin activator danicamtiv (DN) has shown improved function compared with the first-generation myosin activator omecamtiv mecarbil (OM) in nonfailing myocardium by enhancing cardiac force generation but attenuating slowed relaxation. However, whether the functional improvement with DN compared with OM persists in remodeled failing myocardium remains unknown. Therefore, this study aimed to investigate the differential contractile responses to myosin activators in nonfailing and failing myocardium. Mechanical measurements were performed in detergent-skinned myocardium isolated from donor and failing human hearts. Steady-state force, stretch activation responses and loaded shortening velocity were analyzed at submaximal [Ca
2+ ] in the absence or presence of 0.5 µmol/L OM or 2 µmol/L DN. The effects of DN and OM on Ca2+ sensitivity of force generation were determined by incubating myocardial preparations at various [Ca2+ ]. The inherent impairment in force generation and cross-bridge behavior sensitized the failing myocardium to the effects of myosin activators. Specifically, increased Ca2+ sensitivity of force generation, slowed rates of cross-bridge recruitment and detachment following acute stretch, slowed loaded shortening velocity, and diminished power output were more prominent following treatment with OM or DN in failing myocardium compared with donor myocardium. Although these effects were less pronounced with DN compared with OM in failing myocardium, DN impaired contractile properties in failing myocardium that were not affected in donor myocardium. Our results indicate that similar to first-generation myosin activators, the DN-induced slowing of cross-bridge kinetics may result in a prolongation of systolic ejection and delayed diastolic relaxation in the heart failure setting. NEW & NOTEWORTHY This is the first study to provide a detailed mechanistic comparison of omecamtiv mecarbil (OM) and danicamtiv (DN) in failing and nonfailing human myocardium. These findings have clinical implications and the potential to inform the clinical utility of myosin activators in the heart failure setting.- Published
- 2025
- Full Text
- View/download PDF
4. Myocardial Posttranscriptional Landscape in Peripartum Cardiomyopathy.
- Author
-
Li A, Fang B, Li M, Koay YC, Malecki C, Hunter B, Harney D, Dos Remedios CG, Larance M, O'Sullivan JF, and Lal S
- Subjects
- Humans, Female, Pregnancy, Adult, Myocardium metabolism, Myocardium pathology, Cardiomyopathy, Dilated genetics, Cardiomyopathy, Dilated metabolism, Cardiomyopathy, Dilated physiopathology, Pregnancy Complications, Cardiovascular genetics, Pregnancy Complications, Cardiovascular metabolism, Pregnancy Complications, Cardiovascular physiopathology, Cardiomyopathies genetics, Cardiomyopathies metabolism, Cardiomyopathies physiopathology, Case-Control Studies, Peripartum Period, Proteomics
- Abstract
Background: Pregnancy imposes significant cardiovascular adaptations, including progressive increases in plasma volume and cardiac output. For most women, this physiological adaptation resolves at the end of pregnancy, but some women develop pathological dilatation and ultimately heart failure late in pregnancy or in the postpartum period, manifesting as peripartum cardiomyopathy (PPCM). Despite the mortality risk of this form of heart failure, the molecular mechanisms underlying PPCM have not been extensively examined in human hearts., Methods: Protein and metabolite profiles from left ventricular tissue of end-stage PPCM patients (N=6-7) were compared with dilated cardiomyopathy (DCM; N=5-6) and nonfailing donors (N=7-18) using unbiased quantitative mass spectrometry. All samples were derived from the Sydney Heart Bank. Data are available via ProteomeXchange with identifier PXD055986. Differential protein expression and metabolite abundance and Kyoto Encyclopedia of Genes and Genomes pathway analyses were performed., Results: Proteomic analysis identified 2 proteins, SBSPON (somatomedin B and thrombospondin type 1 domain-containing protein precursor) and TNS3 (tensin 3), that were uniquely downregulated in PPCM. SBSPON, an extracellular matrix protein, and TNS3, involved in actin remodeling and cell signaling, may contribute to impaired tissue remodeling and fibrosis in PPCM. Metabolomic analysis revealed elevated levels of homogentisate and deoxycholate and reduced levels of lactate and alanine in PPCM, indicating disrupted metabolic pathways and glucose utilization. Both PPCM and DCM shared pathways related to inflammation, immune responses, and signal transduction. However, thyroid hormone signaling was notably reduced in PPCM, affecting contractility and calcium handling through altered expression of PLN (phospholamban) and Sarcoendoplasmic Reticulum Calcium ATPase (SERCA). Enhanced endoplasmic reticulum stress and altered endocytosis pathways in PPCM suggested additional mechanisms of energy metabolism disruption., Conclusions: The present study reveals unique posttranslational molecular features of the PPCM myocardium, which mediates cellular and metabolic remodeling, and holds promise as potential targets for therapeutic intervention., Competing Interests: None.
- Published
- 2024
- Full Text
- View/download PDF
5. Integrating Clinical Phenotype With Multiomics Analyses of Human Cardiac Tissue Unveils Divergent Metabolic Remodeling in Genotype-Positive and Genotype-Negative Patients With Hypertrophic Cardiomyopathy.
- Author
-
Nollet EE, Schuldt M, Sequeira V, Binek A, Pham TV, Schoonvelde SAC, Jansen M, Schomakers BV, van Weeghel M, Vaz FM, Houtkooper RH, Van Eyk JE, Jimenez CR, Michels M, Bedi KC Jr, Margulies KB, Dos Remedios CG, Kuster DWD, and van der Velden J
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Myocardium metabolism, Myocardium pathology, Metabolomics, Proteomics, Lipidomics, Lipid Metabolism genetics, Sarcomeres metabolism, Sarcomeres genetics, Energy Metabolism genetics, Aged, Multiomics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic pathology, Genotype, Phenotype
- Abstract
Background: Hypertrophic cardiomyopathy (HCM) is caused by sarcomere gene mutations (genotype-positive HCM) in ≈50% of patients and occurs in the absence of mutations (genotype-negative HCM) in the other half of patients. We explored how alterations in the metabolomic and lipidomic landscape are involved in cardiac remodeling in both patient groups., Methods: We performed proteomics, metabolomics, and lipidomics on myectomy samples (genotype-positive N=19; genotype-negative N=22; and genotype unknown N=6) from clinically well-phenotyped patients with HCM and on cardiac tissue samples from sex- and age-matched and body mass index-matched nonfailing donors (N=20). These data sets were integrated to comprehensively map changes in lipid-handling and energy metabolism pathways. By linking metabolomic and lipidomic data to variability in clinical data, we explored patient group-specific associations between cardiac and metabolic remodeling., Results: HCM myectomy samples exhibited (1) increased glucose and glycogen metabolism, (2) downregulation of fatty acid oxidation, and (3) reduced ceramide formation and lipid storage. In genotype-negative patients, septal hypertrophy and diastolic dysfunction correlated with lowering of acylcarnitines, redox metabolites, amino acids, pentose phosphate pathway intermediates, purines, and pyrimidines. In contrast, redox metabolites, amino acids, pentose phosphate pathway intermediates, purines, and pyrimidines were positively associated with septal hypertrophy and diastolic impairment in genotype-positive patients., Conclusions: We provide novel insights into both general and genotype-specific metabolic changes in HCM. Distinct metabolic alterations underlie cardiac disease progression in genotype-negative and genotype-positive patients with HCM., Competing Interests: Disclosures Dr Margulies is a consultant for Bristol Myers Squibb and receives research support from Amgen.
- Published
- 2024
- Full Text
- View/download PDF
6. Biosensing using antibody-modulated motility of actin filaments on myosin-coated surfaces.
- Author
-
Kekic M, Hanson KL, Perumal AS, Solana G, Rajendran K, Dash S, Nicolau DV Jr, Dobroiu S, Dos Remedios CG, and Nicolau DV
- Subjects
- Actin Cytoskeleton metabolism, Myosins metabolism, Cytoskeleton metabolism, Antibodies metabolism, Kinesins metabolism, Actins, Biosensing Techniques
- Abstract
Motor proteins, such as myosin and kinesin, are biological molecular motors involved in force generation and intracellular transport within living cells. The characteristics of molecular motors, i.e., their motility over long distances, their capacity of transporting cargoes, and their very efficient energy consumption, recommend them as potential operational elements of a new class of dynamic nano-devices, with potential applications in biosensing, analyte concentrators, and biocomputation. A possible design of a biosensor based on protein molecular motor comprises a surface with immobilized motors propelling cytoskeletal filaments, which are decorated with antibodies, presented as side-branches. Upon biomolecular recognition of these branches by secondary antibodies, the 'extensions' on the cytoskeletal filaments can achieve considerable lengths (longer than several diameters of the cytoskeletal filament carrier), thus geometrically impairing or halting motility. Because the filaments are several micrometers long, this sensing mechanism converts an event in the nanometer range, i.e., antibody-antigen sizes, into an event in the micrometer range: the visualization of the halting of motility of microns-long cytoskeletal filaments. Here we demonstrate the proof of concept of a sensing system comprising heavy-mero-myosin immobilized on surfaces propelling actin filaments decorated with actin antibodies, whose movement is halted upon the recognition with secondary anti-actin antibodies. Because antibodies to the actin-myosin system are involved in several rare diseases, the first possible application for such a device may be their prognosis and diagnosis. The results also provide insights into guidelines for designing highly sensitive and very fast biosensors powered by motor proteins., Competing Interests: Declaration of competing interest The authors declare that they have no competing interests., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. The microtubule signature in cardiac disease: etiology, disease stage, and age dependency.
- Author
-
Algül S, Dorsch LM, Sorop O, Vink A, Michels M, Dos Remedios CG, Dalinghaus M, Merkus D, Duncker DJ, Kuster DWD, and van der Velden J
- Abstract
Employing animal models to study heart failure (HF) has become indispensable to discover and test novel therapies, but their translatability remains challenging. Although cytoskeletal alterations are linked to HF, the tubulin signature of common experimental models has been incompletely defined. Here, we assessed the tubulin signature in a large set of human cardiac samples and myocardium of animal models with cardiac remodeling caused by pressure overload, myocardial infarction or a gene defect. We studied levels of total, acetylated, and detyrosinated α-tubulin and desmin in cardiac tissue from hypertrophic (HCM) and dilated cardiomyopathy (DCM) patients with an idiopathic (n = 7), ischemic (n = 7) or genetic origin (n = 59), and in a pressure-overload concentric hypertrophic pig model (n = 32), pigs with a myocardial infarction (n = 28), mature pigs (n = 6), and mice (n = 15) carrying the HCM-associated MYBPC3
2373insG mutation. In the human samples, detyrosinated α-tubulin was increased 4-fold in end-stage HCM and 14-fold in pediatric DCM patients. Acetylated α-tubulin was increased twofold in ischemic patients. Across different animal models, the tubulin signature remained mostly unaltered. Only mature pigs were characterized by a 0.5-fold decrease in levels of total, acetylated, and detyrosinated α-tubulin. Moreover, we showed increased desmin levels in biopsies from NYHA class II HCM patients (2.5-fold) and the pressure-overload pig model (0.2-0.3-fold). Together, our data suggest that desmin levels increase early on in concentric hypertrophy and that animal models only partially recapitulate the proliferated and modified tubulin signature observed clinically. Our data warrant careful consideration when studying maladaptive responses to changes in the tubulin content in animal models., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
8. Transcriptomic Comparison of Human Peripartum and Dilated Cardiomyopathy Identifies Differences in Key Disease Pathways.
- Author
-
Taylor J, Yeung ACY, Ashton A, Faiz A, Guryev V, Fang B, Lal S, Grosser M, Dos Remedios CG, Braet F, McLachlan CS, and Li A
- Abstract
Peripartum cardiomyopathy (PPCM) is a rare form of acute onset heart failure that presents in otherwise healthy pregnant women around the time of delivery. While most of these women respond to early intervention, about 20% progress to end-stage heart failure that symptomatically resembles dilated cardiomyopathy (DCM). In this study, we examined two independent RNAseq datasets from the left ventricle of end-stage PPCM patients and compared gene expression profiles to female DCM and non-failing donors. Differential gene expression, enrichment analysis and cellular deconvolution were performed to identify key processes in disease pathology. PPCM and DCM display similar enrichment in metabolic pathways and extracellular matrix remodeling suggesting these are similar processes across end-stage systolic heart failure. Genes involved in golgi vesicles biogenesis and budding were enriched in PPCM left ventricles compared to healthy donors but were not found in DCM. Furthermore, changes in immune cell populations are evident in PPCM but to a lesser extent compared to DCM, where the latter is associated with pronounced pro-inflammatory and cytotoxic T cell activity. This study reveals several pathways that are common to end-stage heart failure but also identifies potential targets of disease that may be unique to PPCM and DCM.
- Published
- 2023
- Full Text
- View/download PDF
9. Multi-Omic Architecture of Obstructive Hypertrophic Cardiomyopathy.
- Author
-
Garmany R, Bos JM, Tester DJ, Giudicessi JR, Dos Remedios CG, Dasari S, Nagaraj NK, Nair AA, Johnson KL, Ryan ZC, Maleszewski JJ, Ommen SR, Dearani JA, and Ackerman MJ
- Subjects
- Humans, Proteomics, Multiomics, Proto-Oncogene Proteins p21(ras) metabolism, Hypertrophy, Left Ventricular, Mitogen-Activated Protein Kinases metabolism, Proteome genetics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism
- Abstract
Background: Hypertrophic cardiomyopathy (HCM) is characterized by asymmetric left ventricular hypertrophy. Currently, hypertrophy pathways responsible for HCM have not been fully elucidated. Their identification could serve as a nidus for the generation of novel therapeutics aimed at halting disease development or progression. Herein, we performed a comprehensive multi-omic characterization of hypertrophy pathways in HCM., Methods: Flash-frozen cardiac tissues were collected from genotyped HCM patients (n=97) undergoing surgical myectomy and tissue from 23 controls. RNA sequencing and mass spectrometry-enabled deep proteome and phosphoproteomic assessment were performed. Rigorous differential expression, gene set enrichment, and pathway analyses were performed to characterize HCM-mediated alterations with emphasis on hypertrophy pathways., Results: We identified transcriptional dysregulation with 1246 (8%) differentially expressed genes and elucidated downregulation of 10 hypertrophy pathways. Deep proteomic analysis identified 411 proteins (9%) that differed between HCM and controls with strong dysregulation of metabolic pathways. Seven hypertrophy pathways were upregulated with antagonistic upregulation of 5 of 10 hypertrophy pathways shown to be downregulated in the transcriptome. Most upregulated hypertrophy pathways encompassed the rat sarcoma-mitogen-activated protein kinase signaling cascade. Phosphoproteomic analysis demonstrated hyperphosphorylation of the rat sarcoma-mitogen-activated protein kinase system suggesting activation of this signaling cascade. There was a common transcriptomic and proteomic profile regardless of genotype., Conclusions: At time of surgical myectomy, the ventricular proteome, independent of genotype, reveals widespread upregulation and activation of hypertrophy pathways, mainly involving the rat sarcoma-mitogen-activated protein kinase signaling cascade. In addition, there is a counterregulatory transcriptional downregulation of the same pathways. Rat sarcoma-mitogen-activated protein kinase activation may serve a crucial role in hypertrophy observed in HCM.
- Published
- 2023
- Full Text
- View/download PDF
10. Transcriptional bursts and heterogeneity among cardiomyocytes in hypertrophic cardiomyopathy.
- Author
-
Burkart V, Kowalski K, Aldag-Niebling D, Beck J, Frick DA, Holler T, Radocaj A, Piep B, Zeug A, Hilfiker-Kleiner D, Dos Remedios CG, van der Velden J, Montag J, and Kraft T
- Abstract
Transcriptional bursting is a common expression mode for most genes where independent transcription of alleles leads to different ratios of allelic mRNA from cell to cell. Here we investigated burst-like transcription and its consequences in cardiac tissue from Hypertrophic Cardiomyopathy (HCM) patients with heterozygous mutations in the sarcomeric proteins cardiac myosin binding protein C (cMyBP-C, MYBPC3 ) and cardiac troponin I (cTnI, TNNI3 ). Using fluorescence in situ hybridization (RNA-FISH) we found that both, MYBPC3 and TNNI3 are transcribed burst-like. Along with that, we show unequal allelic ratios of TNNI3 -mRNA among single cardiomyocytes and unequally distributed wildtype cMyBP-C protein across tissue sections from heterozygous HCM-patients. The mutations led to opposing functional alterations, namely increasing (cMyBP-C
c.927-2A>G ) or decreasing (cTnIR145W ) calcium sensitivity. Regardless, all patients revealed highly variable calcium-dependent force generation between individual cardiomyocytes, indicating contractile imbalance, which appears widespread in HCM-patients. Altogether, we provide strong evidence that burst-like transcription of sarcomeric genes can lead to an allelic mosaic among neighboring cardiomyocytes at mRNA and protein level. In HCM-patients, this presumably induces the observed contractile imbalance among individual cardiomyocytes and promotes HCM-development., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Burkart, Kowalski, Aldag-Niebling, Beck, Frick, Holler, Radocaj, Piep, Zeug, Hilfiker-Kleiner, dos Remedios, van der Velden, Montag and Kraft.)- Published
- 2022
- Full Text
- View/download PDF
11. A bibliometric review of peripartum cardiomyopathy compared to other cardiomyopathies using artificial intelligence and machine learning.
- Author
-
Grosser M, Lin H, Wu M, Zhang Y, Tipper S, Venter D, Lu J, and Dos Remedios CG
- Abstract
As developments in artificial intelligence and machine learning become more widespread in healthcare, their potential to transform clinical outcomes also increases. Peripartum cardiomyopathy is a rare and poorly-characterised condition that presents as heart failure in the last trimester prior to delivery or within 5-6 months postpartum. The lack of a definitive understanding of the molecular causes and clinical progress of this condition suggests that bibliometrics will be well-suited to creating new insights into this serious clinical problem. We examine similarities and differences between peripartum and its closely related familial dilated cardiomyopathy and idiopathic dilated cardiomyopathy. Using PubMed as the source of bibliometric data, we apply artificial intelligence-supported natural language processing to compare extracted data and genes association with these cardiomyopathies. Gene data were enhanced with additional metadata from third-party datasets and then analysed for their impact and specificity for peripartum cardiomyopathy. Artificial intelligence identified 14 genes that distinguished peripartum from both dilated and familial dilated cardiomyopathy. They are as follows: CTSD , RLN2, MMP23B*, SLC17A5, ST2*, PTHLH, CFH*, CFI, GPT , MR1, Rln1, SRI , STAT5A* and THBD. We then used the Human Protein Atlas website that uses affinity-purified rabbit polyclonal antibodies to identify genes that are expressed at the protein level (bold), or as RNA transcripts (*) in healthy human left ventricles. Additional analysis focussed on the full set of peripartum genes on linkage and specificity to cardiomyopathy yielded a different set of thirteen genes (bold font indicates those expressed in cardiomyocytes: PRL, RLN2, PLN , ST2, CTSD , F2, ACE, STAT3, TTN , SPP1, LGALS3, miR-146a, GNB3 , SRI). This type of analysis can highlight new avenues for research, aimed at improving genomics-driven peripartum cardiomyopathy diagnosis as well as potential pathological and clinical sub-classification. We expect that this will allow for future improvements in identification, treatment and management of this condition. The first step in the application of these bibliometric-based artificial intelligence methods is to understand the current knowledge, and it is the aim of this paper to show how this might be achieved., Competing Interests: Conflict of interestMG, HL, DV, and ST are employees of 23 Strands Pty Ltd (Australia), a privately held company, but their employment does not alter the authors’ adherence to the publication policies., (© International Union for Pure and Applied Biophysics (IUPAB) and Springer-Verlag GmbH Germany, part of Springer Nature 2022.)
- Published
- 2022
- Full Text
- View/download PDF
12. Transcriptional co-activators YAP1-TAZ of Hippo signalling in doxorubicin-induced cardiomyopathy.
- Author
-
Berecz T, Yiu A, Vittay O, Orsolits B, Mioulane M, Dos Remedios CG, Ketteler R, Merkely B, Apáti Á, Harding SE, Hellen N, and Foldes G
- Subjects
- Cardiotoxicity etiology, Doxorubicin adverse effects, Doxorubicin metabolism, Humans, Myocytes, Cardiac metabolism, YAP-Signaling Proteins, Cardiomyopathies chemically induced, Cardiomyopathies metabolism, Transcription Factors genetics, Transcription Factors metabolism, Transcription Factors pharmacology
- Abstract
Aims: Hippo signalling is an evolutionarily conserved pathway that controls organ size by regulating apoptosis, cell proliferation, and stem cell self-renewal. Recently, the pathway has been shown to exert powerful growth regulatory activity in cardiomyocytes. However, the functional role of this stress-related and cell death-related pathway in the human heart and cardiomyocytes is not known. In this study, we investigated the role of the transcriptional co-activators of Hippo signalling, YAP and TAZ, in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in response to cardiotoxic agents and investigated the effects of modulating the pathway on cardiomyocyte function and survival., Methods and Results: RNA-sequencing analysis of human heart samples with doxorubicin-induced end-stage heart failure and healthy controls showed that YAP and ERBB2 (HER2) as upstream regulators of differentially expressed genes correlated with doxorubicin treatment. Thus, we tested the effects of doxorubicin on hiPSC-CMs in vitro. Using an automated high-content screen of 96 clinically relevant antineoplastic and cardiotherapeutic drugs, we showed that doxorubicin induced the highest activation of YAP/TAZ nuclear translocation in both hiPSC-CMs and control MCF7 breast cancer cells. The overexpression of YAP rescued doxorubicin-induced cell loss in hiPSC-CMs by inhibiting apoptosis and inducing proliferation. In contrast, silencing of YAP and TAZ by siRNAs resulted in elevated mitochondrial membrane potential loss in response to doxorubicin. hiPSC-CM calcium transients did not change in response to YAP/TAZ silencing., Conclusions: Our results suggest that Hippo signalling is involved in clinical anthracycline-induced cardiomyopathy. Modelling with hiPSC-CMs in vitro showed similar responses to doxorubicin as adult cardiomyocytes and revealed a potential cardioprotective effect of YAP in doxorubicin-induced cardiotoxicity., (© 2021 The Authors. ESC Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.)
- Published
- 2022
- Full Text
- View/download PDF
13. Titin-truncating mutations associated with dilated cardiomyopathy alter length-dependent activation and its modulation via phosphorylation.
- Author
-
Vikhorev PG, Vikhoreva NN, Yeung W, Li A, Lal S, Dos Remedios CG, Blair CA, Guglin M, Campbell KS, Yacoub MH, de Tombe P, and Marston SB
- Subjects
- Adult, Cardiomyopathy, Dilated genetics, Cardiomyopathy, Dilated pathology, Cardiomyopathy, Dilated physiopathology, Connectin genetics, Cyclic AMP-Dependent Protein Kinases metabolism, Female, Genetic Predisposition to Disease, Humans, Kinetics, Male, Middle Aged, Mutation, Myofibrils pathology, Phenotype, Phosphoprotein Phosphatases metabolism, Phosphorylation, Viral Proteins metabolism, Young Adult, Cardiomyopathy, Dilated metabolism, Carrier Proteins metabolism, Connectin metabolism, Myocardial Contraction, Myocytes, Cardiac metabolism, Myofibrils metabolism, Troponin I metabolism
- Abstract
Aims: Dilated cardiomyopathy (DCM) is associated with mutations in many genes encoding sarcomere proteins. Truncating mutations in the titin gene TTN are the most frequent. Proteomic and functional characterizations are required to elucidate the origin of the disease and the pathogenic mechanisms of TTN-truncating variants., Methods and Results: We isolated myofibrils from DCM hearts carrying truncating TTN mutations and measured the Ca2+ sensitivity of force and its length dependence. Simultaneous measurement of force and adenosine triphosphate (ATP) consumption in skinned cardiomyocytes was also performed. Phosphorylation levels of troponin I (TnI) and myosin binding protein-C (MyBP-C) were manipulated using protein kinase A and λ phosphatase. mRNA sequencing was employed to overview gene expression profiles. We found that Ca2+ sensitivity of myofibrils carrying TTN mutations was significantly higher than in myofibrils from donor hearts. The length dependence of the Ca2+ sensitivity was absent in DCM myofibrils with TTN-truncating variants. No significant difference was found in the expression level of TTN mRNA between the DCM and donor groups. TTN exon usage and splicing were also similar. However, we identified down-regulation of genes encoding Z-disk proteins, while the atrial-specific regulatory myosin light chain gene, MYL7, was up-regulated in DCM patients with TTN-truncating variants., Conclusion: Titin-truncating mutations lead to decreased length-dependent activation and increased elasticity of myofibrils. Phosphorylation levels of TnI and MyBP-C seen in the left ventricles are essential for the length-dependent changes in Ca2+ sensitivity in healthy donors, but they are reduced in DCM patients with TTN-truncating variants. A decrease in expression of Z-disk proteins may explain the observed decrease in myofibril passive stiffness and length-dependent activation., (© The Author(s) 2020. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2022
- Full Text
- View/download PDF
14. Truncated titin proteins and titin haploinsufficiency are targets for functional recovery in human cardiomyopathy due to TTN mutations.
- Author
-
Fomin A, Gärtner A, Cyganek L, Tiburcy M, Tuleta I, Wellers L, Folsche L, Hobbach AJ, von Frieling-Salewsky M, Unger A, Hucke A, Koser F, Kassner A, Sielemann K, Streckfuß-Bömeke K, Hasenfuss G, Goedel A, Laugwitz KL, Moretti A, Gummert JF, Dos Remedios CG, Reinecke H, Knöll R, van Heesch S, Hubner N, Zimmermann WH, Milting H, and Linke WA
- Subjects
- Haploinsufficiency, Humans, Mutation, Myocytes, Cardiac metabolism, Tissue Donors, Cardiomyopathies genetics, Connectin genetics, Connectin metabolism, Heart Transplantation, Induced Pluripotent Stem Cells metabolism
- Abstract
Heterozygous truncating variants in TTN (TTNtv), the gene coding for titin, cause dilated cardiomyopathy (DCM), but the underlying pathomechanisms are unclear and disease management remains uncertain. Truncated titin proteins have not yet been considered as a contributor to disease development. Here, we studied myocardial tissues from nonfailing donor hearts and 113 patients with end-stage DCM for titin expression and identified a TTNtv in 22 patients with DCM (19.5%). We directly demonstrate titin haploinsufficiency in TTNtv-DCM hearts and the absence of compensatory changes in the alternative titin isoform Cronos. Twenty-one TTNtv-DCM hearts in our cohort showed stable expression of truncated titin proteins. Expression was variable, up to half of the total titin protein pool, and negatively correlated with patient age at heart transplantation. Truncated titin proteins were not detected in sarcomeres but were present in intracellular aggregates, with deregulated ubiquitin-dependent protein quality control. We produced human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs), comparing wild-type controls to cells with a patient-derived, prototypical A-band-TTNtv or a CRISPR-Cas9–generated M-band-TTNtv. TTNtv-hiPSC-CMs showed reduced wild-type titin expression and contained truncated titin proteins whose proportion increased upon inhibition of proteasomal activity. In engineered heart muscle generated from hiPSC-CMs, depressed contractility caused by TTNtv could be reversed by correction of the mutation using CRISPR-Cas9, eliminating truncated titin proteins and raising wild-type titin content. Functional improvement also occurred when wild-type titin protein content was increased by proteasome inhibition. Our findings reveal the major pathomechanisms of TTNtv-DCM and can be exploited for new therapies to treat TTNtv-related cardiomyopathies.
- Published
- 2021
- Full Text
- View/download PDF
15. Nanoscale Organisation of Ryanodine Receptors and Junctophilin-2 in the Failing Human Heart.
- Author
-
Hou Y, Bai J, Shen X, de Langen O, Li A, Lal S, Dos Remedios CG, Baddeley D, Ruygrok PN, Soeller C, and Crossman DJ
- Abstract
The disrupted organisation of the ryanodine receptors (RyR) and junctophilin (JPH) is thought to underpin the transverse tubule (t-tubule) remodelling in a failing heart. Here, we assessed the nanoscale organisation of these two key proteins in the failing human heart. Recently, an advanced feature of the t-tubule remodelling identified large flattened t-tubules called t-sheets, that were several microns wide. Previously, we reported that in the failing heart, the dilated t-tubules up to ~1 μm wide had increased collagen, and we hypothesised that the t-sheets would also be associated with collagen deposits. Direct stochastic optical reconstruction microscopy (dSTORM), confocal microscopy, and western blotting were used to evaluate the cellular distribution of excitation-contraction structures in the cardiac myocytes from patients with idiopathic dilated cardiomyopathy (IDCM) compared to myocytes from the non-failing (NF) human heart. The dSTORM imaging of RyR and JPH found no difference in the colocalisation between IDCM and NF myocytes, but there was a higher colocalisation at the t-tubule and sarcolemma compared to the corbular regions. Western blots revealed no change in the JPH expression but did identify a ~50% downregulation of RyR ( p = 0.02). The dSTORM imaging revealed a trend for the smaller t-tubular RyR clusters (~24%) and reduced the t-tubular RyR cluster density (~35%) that resulted in a 50% reduction of t-tubular RyR tetramers in the IDCM myocytes ( p < 0.01). Confocal microscopy identified the t-sheets in all the IDCM hearts examined and found that they are associated with the reticular collagen fibres within the lumen. However, the size and density of the RyR clusters were similar in the myocyte regions associated with t-sheets and t-tubules. T-tubule remodelling is associated with a reduced RyR expression that may contribute to the reduced excitation-contraction coupling in the failing human heart., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Hou, Bai, Shen, de Langen, Li, Lal, dos Remedios, Baddeley, Ruygrok, Soeller and Crossman.)
- Published
- 2021
- Full Text
- View/download PDF
16. Involvement of GPR37L1 in murine blood pressure regulation and human cardiac disease pathophysiology.
- Author
-
Mouat MA, Coleman JLJ, Wu J, Dos Remedios CG, Feneley MP, Graham RM, and Smith NJ
- Subjects
- Adult, Animals, Case-Control Studies, Female, Genotype, Heart Failure genetics, Heart Failure physiopathology, Humans, Hypertension genetics, Hypertension physiopathology, Male, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Phenotype, Receptors, G-Protein-Coupled genetics, Species Specificity, Mice, Blood Pressure, Heart Failure metabolism, Hypertension metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Multiple mouse lines lacking the orphan G protein-coupled receptor, GPR37L1, have elicited disparate cardiovascular phenotypes. The first Gpr37l1 knockout mice study to be published reported a marked elevation in systolic blood pressure (SBP; ∼60 mmHg), revealing a potential therapeutic opportunity. The phenotype differed from our own independently generated knockout line, where male mice exhibited equivalent baseline blood pressure to wild type. Here, we attempted to reproduce the first study by characterizing the cardiovascular phenotype of both the original knockout and transgenic lines alongside a C57BL/6J control line, using the same method of blood pressure measurement. The present study supports the findings from our independently developed Gpr37l1 knockout line, finding that SBP and diastolic blood pressure (DBP) are not different in the original Gpr37l1 knockout male mice (SBP: 130.9 ± 5.3 mmHg; DBP: 90.7 ± 3.0 mmHg) compared with C57BL/6J mice (SBP: 123.1 ± 4.1 mmHg; DBP: 87.0 ± 2.7 mmHg). Instead, we attribute the apparent hypertension of the knockout line originally described to comparison with a seemingly hypotensive transgenic line (SBP 103.7 ± 5.0 mmHg; DBP 71.9 ± 3.7 mmHg). Additionally, we quantified myocardial GPR37L1 transcript in humans, which was suggested to be downregulated in cardiovascular disease. We found that GPR37L1 has very low native transcript levels in human myocardium and that expression is not different in tissue samples from patients with heart failure compared with sex-matched healthy control tissue. These findings indicate that cardiac GPR37L1 expression is unlikely to contribute to the pathophysiology of human heart failure. NEW & NOTEWORTHY This study characterizes systolic blood pressure (SBP) in a Gpr37l1 knockout mouse line, which was previously reported to have ∼60 mmHg higher SBP compared with a transgenic line. We observed only a ∼27 mmHg SBP difference between the lines. However, when compared with C57BL/6J mice, knockout mice showed no difference in SBP. We also investigated GPR37L1 mRNA abundance in human hearts and observed no difference between healthy and failing heart samples.
- Published
- 2021
- Full Text
- View/download PDF
17. Sex-Specific Control of Human Heart Maturation by the Progesterone Receptor.
- Author
-
Sim CB, Phipson B, Ziemann M, Rafehi H, Mills RJ, Watt KI, Abu-Bonsrah KD, Kalathur RKR, Voges HK, Dinh DT, Ter Huurne M, Vivien CJ, Kaspi A, Kaipananickal H, Hidalgo A, Delbridge LMD, Robker RL, Gregorevic P, Dos Remedios CG, Lal S, Piers AT, Konstantinov IE, Elliott DA, El-Osta A, Oshlack A, Hudson JE, and Porrello ER
- Subjects
- Female, Humans, Male, Sex Factors, Heart physiopathology, Receptors, Progesterone metabolism
- Abstract
Background: Despite in-depth knowledge of the molecular mechanisms controlling embryonic heart development, little is known about the signals governing postnatal maturation of the human heart., Methods: Single-nucleus RNA sequencing of 54 140 nuclei from 9 human donors was used to profile transcriptional changes in diverse cardiac cell types during maturation from fetal stages to adulthood. Bulk RNA sequencing and the Assay for Transposase-Accessible Chromatin using sequencing were used to further validate transcriptional changes and to profile alterations in the chromatin accessibility landscape in purified cardiomyocyte nuclei from 21 human donors. Functional validation studies of sex steroids implicated in cardiac maturation were performed in human pluripotent stem cell-derived cardiac organoids and mice., Results: Our data identify the progesterone receptor as a key mediator of sex-dependent transcriptional programs during cardiomyocyte maturation. Functional validation studies in human cardiac organoids and mice demonstrate that the progesterone receptor drives sex-specific metabolic programs and maturation of cardiac contractile properties., Conclusions: These data provide a blueprint for understanding human heart maturation in both sexes and reveal an important role for the progesterone receptor in human heart development.
- Published
- 2021
- Full Text
- View/download PDF
18. 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.
- Author
-
Brayson D, Holohan SJ, Bardswell SC, Arno M, Lu H, Jensen HK, Tran PK, Barallobre-Barreiro J, Mayr M, Dos Remedios CG, Tsang VT, Frigiola A, and Kentish JC
- Subjects
- Adolescent, Adult, Child, Collagen analysis, Down-Regulation, Extracellular Matrix Proteins isolation & purification, Female, Gene Expression Profiling methods, Heart Valve Prosthesis Implantation, Humans, Male, Middle Aged, Muscle Contraction physiology, Polymerase Chain Reaction, Pulmonary Valve surgery, Pulmonary Valve Insufficiency surgery, RNA, Messenger metabolism, Small Leucine-Rich Proteoglycans metabolism, Tetralogy of Fallot surgery, Up-Regulation, Young Adult, Extracellular Matrix genetics, Gene Expression, Myocytes, Cardiac physiology, Myofibrils physiology, Tetralogy of Fallot genetics, Ventricular Function, Right genetics
- 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.
- Published
- 2020
- Full Text
- View/download PDF
19. Nanomolar ATP binding to single myosin cross-bridges in rigor: a molecular approach to studying myosin ATP kinetics using single human cardiomyocytes.
- Author
-
Pandzic E, Morkel CA, Li A, Cooke R, Whan RM, and Dos Remedios CG
- Abstract
Our knowledge in the field of cardiac muscle and associated cardiomyopathies has been evolving incrementally over the past 60 years and all was possible due to the parallel progress in techniques and methods allowing to take a fresh glimpse at an old problem. Here, we describe an exciting tool used to examine the various states of the human cardiac myosin at the single molecule level. By imaging single Alexa
647 -ATP binding to permeabilised cardiomyocytes using total internal reflection fluorescence (TIRF) microscopy, we are able to acquire large populations of events in a short timeframe (~ 5000 sites in ~ 10 min) and measure each binding event with high spatio-temporal resolution. The applied algorithm decomposes the point pattern of single molecule binding events into individually distinct binding sites that enables us to recover kinetic parameters, such as bound or free time per site. This single molecule binding approach is a useful tool used to examine muscle contractility. Of particular importance is its application to probing the dynamic lifetimes and proportion of myosins in the super-relaxed state in human cardiomyopathies.- Published
- 2020
- Full Text
- View/download PDF
20. Marked Up-Regulation of ACE2 in Hearts of Patients With Obstructive Hypertrophic Cardiomyopathy: Implications for SARS-CoV-2-Mediated COVID-19.
- Author
-
Bos JM, Hebl VB, Oberg AL, Sun Z, Herman DS, Teekakirikul P, Seidman JG, Seidman CE, Dos Remedios CG, Maleszewski JJ, Schaff HV, Dearani JA, Noseworthy PA, Friedman PA, Ommen SR, Brozovich FV, and Ackerman MJ
- Subjects
- Adolescent, Adult, Aged, Angiotensin-Converting Enzyme 2, COVID-19, Cardiomyopathy, Hypertrophic metabolism, Case-Control Studies, Child, Genotype, Humans, Middle Aged, Myocardium metabolism, Pandemics, RNA, Messenger metabolism, Real-Time Polymerase Chain Reaction, SARS-CoV-2, Young Adult, Betacoronavirus, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic virology, Coronavirus Infections complications, Peptidyl-Dipeptidase A genetics, Peptidyl-Dipeptidase A metabolism, Pneumonia, Viral complications
- Abstract
Objective: To explore the transcriptomic differences between patients with hypertrophic cardiomyopathy (HCM) and controls., Patients and Methods: RNA was extracted from cardiac tissue flash frozen at therapeutic surgical septal myectomy for 106 patients with HCM and 39 healthy donor hearts. Expression profiling of 37,846 genes was performed using the Illumina Human HT-12v3 Expression BeadChip. All patients with HCM were genotyped for pathogenic variants causing HCM. Technical validation was performed using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. This study was started on January 1, 1999, and final analysis was completed on April 20, 2020., Results: Overall, 22% of the transcriptome (8443 of 37,846 genes) was expressed differentially between HCM and control tissues. Analysis by genotype revealed that gene expression changes were similar among genotypic subgroups of HCM, with only 4% (1502 of 37,846) to 6% (2336 of 37,846) of the transcriptome exhibiting differential expression between genotypic subgroups. The qRT-PCR confirmed differential expression in 92% (11 of 12 genes) of tested transcripts. Notably, in the context of coronavirus disease 2019 (COVID-19), the transcript for angiotensin I converting enzyme 2 (ACE2), a negative regulator of the angiotensin system, was the single most up-regulated gene in HCM (fold-change, 3.53; q-value =1.30×10
-23 ), which was confirmed by qRT-PCR in triplicate (fold change, 3.78; P=5.22×10-4 ), and Western blot confirmed greater than 5-fold overexpression of ACE2 protein (fold change, 5.34; P=1.66×10-6 )., Conclusion: More than 20% of the transcriptome is expressed differentially between HCM and control tissues. Importantly, ACE2 was the most up-regulated gene in HCM, indicating perhaps the heart's compensatory effort to mount an antihypertrophic, antifibrotic response. However, given that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses ACE2 for viral entry, this 5-fold increase in ACE2 protein may confer increased risk for COVID-19 manifestations and outcomes in patients with increased ACE2 transcript expression and protein levels in the heart., (Copyright © 2020 Mayo Foundation for Medical Education and Research. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
21. Sex-specific cardiac remodeling in early and advanced stages of hypertrophic cardiomyopathy.
- Author
-
Nijenkamp LLAM, Bollen IAE, Niessen HWM, Dos Remedios CG, Michels M, Poggesi C, Ho CY, Kuster DWD, and van der Velden J
- Subjects
- Adolescent, Adult, Aged, Capillaries pathology, Cardiac Myosins genetics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic physiopathology, Carrier Proteins genetics, Case-Control Studies, Child, Disease Progression, Female, Humans, Male, Middle Aged, Mutation, Myocytes, Cardiac pathology, Myocytes, Cardiac physiology, Myosin Heavy Chains genetics, Risk Factors, Sex Characteristics, Troponin T genetics, Ventricular Remodeling genetics, Young Adult, Cardiomyopathy, Hypertrophic pathology, Ventricular Remodeling physiology
- Abstract
Hypertrophic cardiomyopathy (HCM) is the most frequent genetic cardiac disease with a prevalence of 1:500 to 1:200. While most patients show obstructive HCM and a relatively stable clinical phenotype (stage II), a small group of patients progresses to end-stage HCM (stage IV) within a relatively brief period. Previous research has shown sex-differences in stage II HCM with more diastolic dysfunction in female than in male patients. Moreover, female patients more often show progression to heart failure. Here we investigated if differences in functional and structural properties of the heart may underlie sex-differences in disease progression from stage II to stage IV HCM. Cardiac tissue from stage II and IV patients was obtained during myectomy (n = 54) and heart transplantation (n = 10), respectively. Isometric force was measured in membrane-permeabilized cardiomyocytes to define active and passive myofilament force development. Titin isoform composition was assessed using gel electrophoresis, and the amount of fibrosis and capillary density were determined with histology. In accordance with disease stage-dependent adverse cardiac remodeling end-stage patients showed a thinner interventricular septal wall and larger left ventricular and atrial diameters compared to stage II patients. Cardiomyocyte contractile properties and fibrosis were comparable between stage II and IV, while capillary density was significantly lower in stage IV compared to stage II. Women showed more adverse cellular remodeling compared to men at stage II, evident from more compliant titin, more fibrosis and lower capillary density. However, the disease stage-dependent reduction in capillary density was largest in men. In conclusion, the more severe cellular remodeling in female compared to male stage II patients suggests a more advanced disease stage at the time of myectomy in women. Changes in cardiomyocyte contractile properties do not explain the progression of stage II to stage IV, while reduced capillary density may underlie disease progression to end-stage heart failure., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
22. Collaborative Regulation of LRG1 by TGF-β1 and PPAR-β/δ Modulates Chronic Pressure Overload-Induced Cardiac Fibrosis.
- Author
-
Liu C, Lim ST, Teo MHY, Tan MSY, Kulkarni MD, Qiu B, Li A, Lal S, Dos Remedios CG, Tan NS, Wahli W, Ferenczi MA, Song W, Hong W, and Wang X
- Subjects
- Adult, Aged, Animals, Cells, Cultured, Chronic Disease, Disease Models, Animal, Female, Fibroblasts pathology, Fibrosis, Glycoproteins deficiency, Glycoproteins genetics, Heart Diseases pathology, Heart Diseases physiopathology, Humans, Male, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Myocardium pathology, Nuclear Receptor Co-Repressor 2 metabolism, PPAR gamma deficiency, PPAR gamma genetics, PPAR-beta deficiency, PPAR-beta genetics, Signal Transduction, Fibroblasts metabolism, Glycoproteins metabolism, Heart Diseases metabolism, Myocardium metabolism, PPAR gamma metabolism, PPAR-beta metabolism, Transforming Growth Factor beta1 metabolism, Ventricular Function, Left, Ventricular Remodeling
- Abstract
Background: Despite its established significance in fibrotic cardiac remodeling, clinical benefits of global inhibition of TGF (transforming growth factor)-β1 signaling remain controversial. LRG1 (leucine-rich-α2 glycoprotein 1) is known to regulate endothelial TGFβ signaling. This study evaluated the role of LRG1 in cardiac fibrosis and its transcriptional regulatory network in cardiac fibroblasts., Methods: Pressure overload-induced heart failure was established by transverse aortic constriction. Western blot, quantitative reverse transcription polymerase chain reaction, immunofluorescence, and immunohistochemistry were used to evaluate the expression level and pattern of interested targets or pathology during fibrotic cardiac remodeling. Cardiac function was assessed by pressure-volume loop analysis., Results: LRG1 expression was significantly suppressed in left ventricle of mice with transverse aortic constriction-induced fibrotic cardiac remodeling (mean difference, -0.00085 [95% CI, -0.0013 to -0.00043]; P =0.005) and of patients with end-stage ischemic-dilated cardiomyopathy (mean difference, 0.13 [95% CI, 0.012-0.25]; P =0.032). More profound cardiac fibrosis (mean difference, -0.014% [95% CI, -0.029% to -0.00012%]; P =0.048 for interstitial fibrosis; mean difference, -1.3 [95% CI, -2.5 to -0.2]; P =0.016 for perivascular fibrosis), worse cardiac dysfunction (mean difference, -2.5 ms [95% CI, -4.5 to -0.4 ms]; P =0.016 for Tau-g; mean difference, 13% [95% CI, 2%-24%]; P =0.016 for ejection fraction), and hyperactive TGFβ signaling in transverse aortic constriction-operated Lrg1 -deficient mice (mean difference, -0.27 [95% CI, -0.47 to -0.07]; P <0.001), which could be reversed by cardiac-specific Lrg1 delivery mediated by adeno-associated virus 9. Mechanistically, LRG1 inhibits cardiac fibroblast activation by competing with TGFβ1 for receptor binding, while PPAR (peroxisome proliferator-activated receptor)-β/δ and TGFβ1 collaboratively regulate LRG1 expression via SMRT (silencing mediator for retinoid and thyroid hormone receptor). We further demonstrated functional interactions between LRG1 and PPARβ/δ in cardiac fibroblast activation., Conclusions: Our results established a highly complex molecular network involving LRG1, TGFβ1, PPARβ/δ, and SMRT in regulating cardiac fibroblast activation and cardiac fibrosis. Targeting LRG1 or PPARβ/δ represents a promising strategy to control pathological cardiac remodeling in response to chronic pressure overload.
- Published
- 2019
- Full Text
- View/download PDF
23. Prelamin A mediates myocardial inflammation in dilated and HIV-associated cardiomyopathies.
- Author
-
Brayson D, Frustaci A, Verardo R, Chimenti C, Russo MA, Hayward R, Ahmad S, Vizcay-Barrena G, Protti A, Zammit PS, dos Remedios CG, Ehler E, Shah AM, and Shanahan CM
- Subjects
- Adult, Animals, Disease Models, Animal, Female, Heart physiopathology, Humans, Male, Mice, Middle Aged, Myocardium metabolism, Myocardium pathology, Cardiomyopathy, Dilated metabolism, Cardiomyopathy, Dilated virology, HIV Infections complications, HIV Infections metabolism, Inflammation metabolism, Lamin Type A genetics, Lamin Type A metabolism
- Abstract
Cardiomyopathies are complex heart muscle diseases that can be inherited or acquired. Dilated cardiomyopathy can result from mutations in LMNA, encoding the nuclear intermediate filament proteins lamin A/C. Some LMNA mutations lead to accumulation of the lamin A precursor, prelamin A, which is disease causing in a number of tissues, yet its impact upon the heart is unknown. Here, we discovered myocardial prelamin A accumulation occurred in a case of dilated cardiomyopathy, and we show that a potentially novel mouse model of cardiac-specific prelamin A accumulation exhibited a phenotype consistent with inflammatory cardiomyopathy, which we observed to be similar to HIV-associated cardiomyopathy, an acquired disease state. Numerous HIV protease therapies are known to inhibit ZMPSTE24, the enzyme responsible for prelamin A processing, and we confirmed that accumulation of prelamin A occurred in HIV+ patient cardiac biopsies. These findings (a) confirm a unifying pathological role for prelamin A common to genetic and acquired cardiomyopathies; (b) have implications for the management of HIV patients with cardiac disease, suggesting protease inhibitors should be replaced with alternative therapies (i.e., nonnucleoside reverse transcriptase inhibitors); and (c) suggest that targeting inflammation may be a useful treatment strategy for certain forms of inherited cardiomyopathy.
- Published
- 2019
- Full Text
- View/download PDF
24. Enhanced cardiac repair by telomerase reverse transcriptase over-expression in human cardiac mesenchymal stromal cells.
- Author
-
Le TYL, Pickett HA, Yang A, Ho JWK, Thavapalachandran S, Igoor S, Yang SF, Farraha M, Voges HK, Hudson JE, Dos Remedios CG, Bryan TM, Kizana E, and Chong JJH
- Subjects
- Animals, Child, Humans, Middle Aged, Myocytes, Cardiac transplantation, Rats, Receptor, Platelet-Derived Growth Factor alpha metabolism, Mesenchymal Stem Cell Transplantation methods, Mesenchymal Stem Cells enzymology, Myocardial Infarction therapy, Myocytes, Cardiac enzymology, Telomerase metabolism
- Abstract
We have previously reported a subpopulation of mesenchymal stromal cells (MSCs) within the platelet-derived growth factor receptor-alpha (PDGFRα)/CD90 co-expressing cardiac interstitial and adventitial cell fraction. Here we further characterise PDGFRα/CD90-expressing cardiac MSCs (PDGFRα + cMSCs) and use human telomerase reverse transcriptase (hTERT) over-expression to increase cMSCs ability to repair the heart after induced myocardial infarction. hTERT over-expression in PDGFRα + cardiac MSCs (hTERT + PDGFRα + cMSCs) modulates cell differentiation, proliferation, survival and angiogenesis related genes. In vivo, transplantation of hTERT + PDGFRα + cMSCs in athymic rats significantly increased left ventricular function, reduced scar size, increased angiogenesis and proliferation of both cardiomyocyte and non-myocyte cell fractions four weeks after myocardial infarction. In contrast, transplantation of mutant hTERT + PDGFRα + cMSCs (which generate catalytically-inactive telomerase) failed to replicate this cardiac functional improvement, indicating a telomerase-dependent mechanism. There was no hTERT + PDGFRα + cMSCs engraftment 14 days after transplantation indicating functional improvement occurred by paracrine mechanisms. Mass spectrometry on hTERT + PDGFRα + cMSCs conditioned media showed increased proteins associated with matrix modulation, angiogenesis, cell proliferation/survival/adhesion and innate immunity function. Our study shows that hTERT can activate pro-regenerative signalling within PDGFRα + cMSCs and enhance cardiac repair after myocardial infarction. An increased understanding of hTERT's role in mesenchymal stromal cells from various organs will favourably impact clinical regenerative and anti-cancer therapies.
- Published
- 2019
- Full Text
- View/download PDF
25. Protein Quality Control Activation and Microtubule Remodeling in Hypertrophic Cardiomyopathy.
- Author
-
Dorsch LM, Schuldt M, dos Remedios CG, Schinkel AFL, de Jong PL, Michels M, Kuster DWD, Brundel BJJM, and van der Velden J
- Subjects
- Adult, Aged, Cardiomyopathy, Hypertrophic genetics, Carrier Proteins genetics, Carrier Proteins metabolism, Chaperonin 60 metabolism, Female, HSP70 Heat-Shock Proteins metabolism, Humans, Male, Microtubules genetics, Middle Aged, Mitochondrial Proteins metabolism, Mutation, Myocytes, Cardiac metabolism, Sarcomeres genetics, Sarcomeres metabolism, Tubulin metabolism, Cardiomyopathy, Hypertrophic metabolism, Microtubules metabolism, Unfolded Protein Response
- Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac disorder. It is mainly caused by mutations in genes encoding sarcomere proteins. Mutant forms of these highly abundant proteins likely stress the protein quality control (PQC) system of cardiomyocytes. The PQC system, together with a functional microtubule network, maintains proteostasis. We compared left ventricular (LV) tissue of nine donors (controls) with 38 sarcomere mutation-positive (HCM
SMP ) and 14 sarcomere mutation-negative (HCMSMN ) patients to define HCM and mutation-specific changes in PQC. Mutations in HCMSMP result in poison polypeptides or reduced protein levels (haploinsufficiency, HI). The main findings were 1) several key PQC players were more abundant in HCM compared to controls, 2) after correction for sex and age, stabilizing heat shock protein (HSP)B1, and refolding, HSPD1 and HSPA2 were increased in HCMSMP compared to controls, 3) α-tubulin and acetylated α-tubulin levels were higher in HCM compared to controls, especially in HCMHI , 4) myosin-binding protein-C (cMyBP-C) levels were inversely correlated with α-tubulin, and 5) α-tubulin levels correlated with acetylated α-tubulin and HSPs. Overall, carrying a mutation affects PQC and α-tubulin acetylation. The haploinsufficiency of cMyBP-C may trigger HSPs and α-tubulin acetylation. Our study indicates that proliferation of the microtubular network may represent a novel pathomechanism in cMyBP-C haploinsufficiency-mediated HCM., Competing Interests: The authors declare no conflict of interest.- Published
- 2019
- Full Text
- View/download PDF
26. A step towards understanding the molecular nature of human heart failure: advances using the Sydney Heart Bank collection.
- Author
-
Li A, Lal S, and Dos Remedios CG
- Published
- 2019
- Full Text
- View/download PDF
27. Letter from the Editor.
- Author
-
Dos Remedios CG
- Published
- 2019
- Full Text
- View/download PDF
28. PKC and PKN in heart disease.
- Author
-
Marrocco V, Bogomolovas J, Ehler E, Dos Remedios CG, Yu J, Gao C, and Lange S
- Subjects
- Cyclic AMP-Dependent Protein Kinases genetics, Heart Diseases enzymology, Heart Diseases pathology, Humans, Myocardium pathology, Proto-Oncogene Proteins c-akt genetics, Signal Transduction genetics, Heart Diseases genetics, Myocardium enzymology, Protein Kinase C genetics
- Abstract
The protein kinase C (PKC) and closely related protein kinase N (PKN) families of serine/threonine protein kinases play crucial cellular roles. Both kinases belong to the AGC subfamily of protein kinases that also include the cAMP dependent protein kinase (PKA), protein kinase B (PKB/AKT), protein kinase G (PKG) and the ribosomal protein S6 kinase (S6K). Involvement of PKC family members in heart disease has been well documented over the years, as their activity and levels are mis-regulated in several pathological heart conditions, such as ischemia, diabetic cardiomyopathy, as well as hypertrophic or dilated cardiomyopathy. This review focuses on the regulation of PKCs and PKNs in different pathological heart conditions and on the influences that PKC/PKN activation has on several physiological processes. In addition, we discuss mechanisms by which PKCs and the closely related PKNs are activated and turned-off in hearts, how they regulate cardiac specific downstream targets and pathways, and how their inhibition by small molecules is explored as new therapeutic target to treat cardiomyopathies and heart failure., (Copyright © 2019 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
29. A review of heavy metal cation binding to deoxyribonucleic acids for the creation of chemical sensors.
- Author
-
Kanellis VG and Dos Remedios CG
- Abstract
Various human activities lead to the pollution of ground, drinking, and wastewater with toxic metals. It is well known that metal ions preferentially bind to DNA phosphate backbones or DNA nucleobases, or both. Foreman et al. (Environ Toxicol Chem 30(8):1810-1818, 2011) reported the use of a DNA-dye based assay suitable for use as a toxicity test for potable environmental water. They compared the results of this test with the responses of live-organism bioassays. The DNA-based demonstrated that the loss of SYBR Green I fluorescence dye bound to calf thymus DNA was proportional to the toxicity of the water sample. However, this report raised questions about the mechanism that formed the basis of this quasi-quantitatively test. In this review, we identify the unique and preferred DNA-binding sites of individual metals. We show how highly sensitive and selective DNA-based sensors can be designed that contain multiple binding sites for 21 heavy metal cations that bind to DNA and change its structure, consistent with the release of the DNA-bound dye.
- Published
- 2018
- Full Text
- View/download PDF
30. Reviews arising from the 2017 conference of the Australian Society for Biophysics and the Japanese Society for Biophysics.
- Author
-
Dos Remedios CG
- Published
- 2018
- Full Text
- View/download PDF
31. Author Correction: Abnormal contractility in human heart myofibrils from patients with dilated cardiomyopathy due to mutations in TTN and contractile protein genes.
- Author
-
Vikhorev PG, Smoktunowicz N, Munster AB, Copeland ON, Kostin S, Montgiraud C, Messer AE, Toliat MR, Li A, Dos Remedios CG, Lal S, Blair CA, Campbell KS, Guglin M, Richter M, Knöll R, and Marston SB
- Abstract
A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.
- Published
- 2018
- Full Text
- View/download PDF
32. Non-sarcomeric causes of heart failure: a Sydney Heart Bank perspective.
- Author
-
Dos Remedios CG, Li A, and Lal S
- Published
- 2018
- Full Text
- View/download PDF
33. An historical perspective of the discovery of titin filaments -Part 2.
- Author
-
Dos Remedios CG
- Abstract
In 2017, a Special Issue of Biophysical Reviews was devoted to "Titin and Its Binding Partners. The issue contained a review: "An historical perspective of the discovery of titin filaments" by dos Remedios and Gilmour that was intended to be a history of the discovery of the giant protein titin, previously named connectin. The review took readers back to the earliest discovery of the so-called third filament component of skeletal and cardiac muscle sarcomeres and ended in 1969. Recently, my colleague Shin'ichi Ishiwata gently reminded me of two papers published in 1990 and 1993 that were unwittingly omitted from the original historical perspective. In the first paper (J Cell Biol 110:53-62, 1990), Funatsu et al. examined the elastic filaments in skeletal muscle using a combination of light and electron microscopy, but they also measured resting as well as passive stiffness mechanical measurements to establish that connectin (titin) is responsible for both stiffness and fiber tension. In the second paper (J Cell Biol 120:711-724, 1993), Funatsu et al. used permeabilised cardiac muscle myocytes (from rabbit papillary muscles) and focussed on filament ultrastructure using either freeze-substitution or deep-etched replica methods to visualise connectin/titin filaments in fibers with and without actin and myosin filaments.
- Published
- 2018
- Full Text
- View/download PDF
34. Alterations in Titin Properties and Myocardial Fibrosis Correlate With Clinical Phenotypes in Hemodynamic Subgroups of Severe Aortic Stenosis.
- Author
-
Gotzmann M, Grabbe S, Schöne D, von Frieling-Salewsky M, Dos Remedios CG, Strauch J, Bechtel M, Dietrich JW, Tannapfel A, Mügge A, and Linke WA
- Abstract
Titin-isoform expression, titin phosphorylation, and myocardial fibrosis were studied in 30 patients with severe symptomatic aortic stenosis (AS). Patients were grouped into "classical" high-gradient, normal-flow AS with preserved ejection fraction (EF); "paradoxical" low-flow, low-gradient AS with preserved EF; and AS with reduced EF. Nonfailing donor hearts served as controls. AS was associated with increased fibrosis, titin-isoform switch toward compliant N2BA, and both total and site-specific titin hypophosphorylation compared with control hearts. All AS subtypes revealed titin and matrix alterations. The extent of myocardial remodeling in "paradoxical" AS was no less severe than in other AS subtypes, thus explaining the unfavorable prognosis.
- Published
- 2018
- Full Text
- View/download PDF
35. Monophosphorylation of cardiac troponin-I at Ser-23/24 is sufficient to regulate cardiac myofibrillar Ca 2+ sensitivity and calpain-induced proteolysis.
- Author
-
Martin-Garrido A, Biesiadecki BJ, Salhi HE, Shaifta Y, Dos Remedios CG, Ayaz-Guner S, Cai W, Ge Y, Avkiran M, and Kentish JC
- Subjects
- Animals, Cells, Cultured, Cyclic AMP-Dependent Protein Kinases metabolism, Humans, Mice, Myocytes, Cardiac cytology, Myocytes, Cardiac drug effects, Myofibrils drug effects, Phosphorylation, Protein Kinase C metabolism, Rats, Serine chemistry, Calcium metabolism, Calpain pharmacology, Myocytes, Cardiac physiology, Myofibrils physiology, Proteolysis drug effects, Serine metabolism, Troponin I metabolism
- Abstract
The acceleration of myocardial relaxation produced by β-adrenoreceptor stimulation is mediated in part by protein kinase A (PKA)-mediated phosphorylation of cardiac troponin-I (cTnI), which decreases myofibrillar Ca
2+ sensitivity. Previous evidence suggests that phosphorylation of both Ser-23 and Ser-24 in cTnI is required for this Ca2+ desensitization. PKA-mediated phosphorylation also partially protects cTnI from proteolysis by calpain. Here we report that protein kinase D (PKD) phosphorylates only one serine of cTnI Ser-23/24. To explore the functional consequences of this monophosphorylation, we examined the Ca2+ sensitivity of force production and susceptibility of cTnI to calpain-mediated proteolysis when Ser-23/24 of cTnI in mouse cardiac myofibrils was nonphosphorylated, mono-phosphorylated, or bisphosphorylated (using sequential incubations in λ-phosphatase, PKD, and PKA, respectively). Phos-tag gels, Western blotting, and high-resolution MS revealed that PKD produced >90% monophosphorylation of cTnI, primarily at Ser-24, whereas PKA led to cTnI bisphosphorylation exclusively. PKD markedly decreased the Ca2+ sensitivity of force production in detergent-permeabilized ventricular trabeculae, whereas subsequent incubation with PKA produced only a small further fall of Ca2+ sensitivity. Unlike PKD, PKA also substantially phosphorylated myosin-binding protein-C and significantly accelerated cross-bridge kinetics ( ktr ). After phosphorylation by PKD or PKA, cTnI in isolated myofibrils was partially protected from calpain-mediated degradation. We conclude that cTnI monophosphorylation at Ser-23/24 decreases myofibrillar Ca2+ sensitivity and partially protects cTnI from calpain-induced proteolysis. In healthy cardiomyocytes, the basal monophosphorylation of cTnI may help tonically regulate myofibrillar Ca2+ sensitivity., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2018
- Full Text
- View/download PDF
36. Correction to: The Sydney Heart Bank: improving translational research while eliminating or reducing the use of animal models of human heart disease.
- Author
-
Dos Remedios CG, Lal SP, Li A, McNamara J, Keogh A, Macdonald PS, Cooke R, Ehler E, Knöll R, Marston SB, Stelzer J, Granzier H, Bezzina C, van Dijk S, De Man F, Stienen GJM, Odeberg J, Pontén F, Linke WA, and van der Velden J
- Abstract
In the original version of this article, the name of one of the authors is not correct. The correct name should be W. A. Linke, which is shown correctly in the authorgroup section above.
- Published
- 2018
- Full Text
- View/download PDF
37. Burst-Like Transcription of Mutant and Wildtype MYH7 -Alleles as Possible Origin of Cell-to-Cell Contractile Imbalance in Hypertrophic Cardiomyopathy.
- Author
-
Montag J, Kowalski K, Makul M, Ernstberger P, Radocaj A, Beck J, Becker E, Tripathi S, Keyser B, Mühlfeld C, Wissel K, Pich A, van der Velden J, Dos Remedios CG, Perrot A, Francino A, Navarro-López F, Brenner B, and Kraft T
- Abstract
Hypertrophic Cardiomyopathy (HCM) has been related to many different mutations in more than 20 different, mostly sarcomeric proteins. While development of the HCM-phenotype is thought to be triggered by the different mutations, a common mechanism remains elusive. Studying missense-mutations in the ventricular beta-myosin heavy chain (β-MyHC, MYH7 ) we hypothesized that significant contractile heterogeneity exists among individual cardiomyocytes of HCM-patients that results from cell-to-cell variation in relative expression of mutated vs. wildtype β-MyHC. To test this hypothesis, we measured force-calcium-relationships of cardiomyocytes isolated from myocardium of heterozygous HCM-patients with either β-MyHC-mutation Arg723Gly or Arg200Val, and from healthy controls. From the myocardial samples of the HCM-patients we also obtained cryo-sections, and laser-microdissected single cardiomyocytes for quantification of mutated vs. wildtype MYH7 -mRNA using a single cell RT-qPCR and restriction digest approach. We characterized gene transcription by visualizing active transcription sites by fluorescence in situ hybridization of intronic and exonic sequences of MYH7 -pre-mRNA. For both mutations, cardiomyocytes showed large cell-to-cell variation in Ca
++ -sensitivity. Interestingly, some cardiomyocytes were essentially indistinguishable from controls what might indicate that they had no mutant β-MyHC while others had highly reduced Ca++ -sensitivity suggesting substantial fractions of mutant β-MyHC. Single-cell MYH7 -mRNA-quantification in cardiomyocytes of the same patients revealed high cell-to-cell variability of mutated vs. wildtype mRNA, ranging from essentially pure mutant to essentially pure wildtype MYH7 -mRNA. We found 27% of nuclei without active transcription sites which is inconsistent with continuous gene transcription but suggests burst-like transcription of MYH7 . Model simulations indicated that burst-like, stochastic on/off-switching of MYH7 transcription, which is independent for mutant and wildtype alleles, could generate the observed cell-to-cell variation in the fraction of mutant vs. wildtype MYH7 -mRNA, a similar variation in β-MyHC-protein, and highly heterogeneous Ca++ -sensitivity of individual cardiomyocytes. In the long run, such contractile imbalance in the myocardium may well induce progressive structural distortions like cellular and myofibrillar disarray and interstitial fibrosis, as they are typically observed in HCM.- Published
- 2018
- Full Text
- View/download PDF
38. Orphan receptor GPR37L1 contributes to the sexual dimorphism of central cardiovascular control.
- Author
-
Coleman JLJ, Mouat MA, Wu J, Jancovski N, Bassi JK, Chan AY, Humphreys DT, Mrad N, Yu ZY, Ngo T, Iismaa S, Dos Remedios CG, Feneley MP, Allen AM, Graham RM, and Smith NJ
- Subjects
- Animals, Brain metabolism, Female, Fibrosis, Kidney metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Myocardium metabolism, Myocardium pathology, Sex Characteristics, Blood Pressure, Receptors, G-Protein-Coupled physiology
- Abstract
Background: Over 100 mammalian G protein-coupled receptors are yet to be matched with endogenous ligands; these so-called orphans are prospective drug targets for the treatment of disease. GPR37L1 is one such orphan, abundant in the brain and detectable as mRNA in the heart and kidney. GPR37L1 ablation was reported to cause hypertension and left ventricular hypertrophy, and thus, we sought to further define the role of GPR37L1 in blood pressure homeostasis., Methods: We investigated the cardiovascular effects of GPR37L1 using wild-type (GPR37L1
wt/wt ) and null (GPR37L1KO/KO ) mice established on a C57BL/6J background, both under baseline conditions and during AngII infusion. We profiled GPR37L1 tissue expression, examining the endogenous receptor by immunoblotting and a β-galactosidase reporter mouse by immunohistochemistry., Results: GPR37L1 protein was abundant in the brain but not detectable in the heart and kidney. We measured blood pressure in GPR37L1wt/wt and GPR37L1KO/KO mice and found that deletion of GPR37L1 causes a female-specific increase in systolic, diastolic, and mean arterial pressures. When challenged with short-term AngII infusion, only male GPR37L1KO/KO mice developed exacerbated left ventricular hypertrophy and evidence of heart failure, while the female GPR37L1KO/KO mice were protected from cardiac fibrosis., Conclusions: Despite its absence in the heart and kidney, GPR37L1 regulates baseline blood pressure in female mice and is crucial for cardiovascular compensatory responses in males. The expression of GPR37L1 in the brain, yet absence from peripheral cardiovascular tissues, suggests this orphan receptor is a hitherto unknown contributor to central cardiovascular control.- Published
- 2018
- Full Text
- View/download PDF
39. Skeletal myosin binding protein-C isoforms regulate thin filament activity in a Ca 2+ -dependent manner.
- Author
-
Lin BL, Li A, Mun JY, Previs MJ, Previs SB, Campbell SG, Dos Remedios CG, Tombe PP, Craig R, Warshaw DM, and Sadayappan S
- Subjects
- Actins metabolism, Animals, Calcium metabolism, Male, Muscle Contraction, Protein Isoforms physiology, Rats, Sprague-Dawley, Recombinant Proteins pharmacology, Tropomyosin metabolism, Carrier Proteins physiology, Muscle, Skeletal physiology, Myocardial Contraction, Myocardium
- Abstract
Muscle contraction, which is initiated by Ca
2+ , results in precise sliding of myosin-based thick and actin-based thin filament contractile proteins. The interactions between myosin and actin are finely tuned by three isoforms of myosin binding protein-C (MyBP-C): slow-skeletal, fast-skeletal, and cardiac (ssMyBP-C, fsMyBP-C and cMyBP-C, respectively), each with distinct N-terminal regulatory regions. The skeletal MyBP-C isoforms are conditionally coexpressed in cardiac muscle, but little is known about their function. Therefore, to characterize the functional differences and regulatory mechanisms among these three isoforms, we expressed recombinant N-terminal fragments and examined their effect on contractile properties in biophysical assays. Addition of the fragments to in vitro motility assays demonstrated that ssMyBP-C and cMyBP-C activate thin filament sliding at low Ca2+ . Corresponding 3D electron microscopy reconstructions of native thin filaments suggest that graded shifts of tropomyosin on actin are responsible for this activation (cardiac > slow-skeletal > fast-skeletal). Conversely, at higher Ca2+ , addition of fsMyBP-C and cMyBP-C fragments reduced sliding velocities in the in vitro motility assays and increased force production in cardiac muscle fibers. We conclude that due to the high frequency of Ca2+ cycling in cardiac muscle, cardiac MyBP-C may play dual roles at both low and high Ca2+ . However, skeletal MyBP-C isoforms may be tuned to meet the needs of specific skeletal muscles.- Published
- 2018
- Full Text
- View/download PDF
40. Editorial for issue #1 2018.
- Author
-
Dos Remedios CG and Li A
- Published
- 2018
- Full Text
- View/download PDF
41. Platelet-Derived Growth Factor Receptor-Alpha Expressing Cardiac Progenitor Cells Can Be Derived from Previously Cryopreserved Human Heart Samples.
- Author
-
Le TYL, Pickett HA, Dos Remedios CG, Barbaro PM, Kizana E, and Chong JJH
- Subjects
- Cells, Cultured, Humans, Kruppel-Like Factor 4, Myoblasts, Cardiac cytology, Myoblasts, Cardiac metabolism, Antigens, Differentiation biosynthesis, Cell Separation, Cryopreservation, Gene Expression Regulation, Myocardium cytology, Myocardium metabolism, Receptor, Platelet-Derived Growth Factor alpha biosynthesis
- Abstract
Cardiac progenitor cells (CPCs) are being developed as a promising treatment for heart failure. Although clinical trials have predominantly used donor cardiac biopsies to derive CPCs, a better solution could be to use previously cryopreserved human heart tissue. This would enable timely and convenient access to healthy and young heart samples for CPC production. However, few studies have attempted to isolate CPCs from previously cryopreserved heart tissue. In this study, we isolated CPCs from eight nondiseased human heart samples previously cryopreserved as part of the Sydney Heart Bank. Resulting cells were strongly positive for known fibroblast (DDR2, Vimentin), mesenchymal/CPC (PDGFRα, CD90) markers, and for pluripotency genes (SOX2, NANOG, MYC, KLF4), whereas being negative for the pan-hematopoietic marker (CD45). Outgrowth cells from aged hearts had decreased proliferative and self-renewing capacity that correlated with shorter telomere lengths compared with cells from young hearts. No telomerase activity was detected in any cells isolated. Colony-forming assays and fluorescence-activated cell sorting were used to enrich PDGFRα
+ /CD90+ /CD31- CPCs. Multipotent potential was confirmed using in vitro differentiation assays with smooth muscle (MYH11+ ), endothelial cell (vWF+ ), and cardiomyocyte-like (cTnT+ , α-actinin+ ) cell formation. Single cell assays demonstrated clonogenicity of PDGFRα+ CPCs with maintenance of prolonged self-renewing capacity (>2 months), and pluripotency gene expression at both early and late culture passages. Our results demonstrate that multipotent PDGFRα+ CPCs can be harvested and expanded from previously banked cryopreserved human heart samples. These data support cardiac tissue banking as a strategy for improved access to CPCs for future clinical therapies.- Published
- 2018
- Full Text
- View/download PDF
42. Differences in Contractile Function of Myofibrils within Human Embryonic Stem Cell-Derived Cardiomyocytes vs. Adult Ventricular Myofibrils Are Related to Distinct Sarcomeric Protein Isoforms.
- Author
-
Iorga B, Schwanke K, Weber N, Wendland M, Greten S, Piep B, Dos Remedios CG, Martin U, Zweigerdt R, Kraft T, and Brenner B
- Abstract
Characterizing the contractile function of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is key for advancing their utility for cellular disease models, promoting cell based heart repair, or developing novel pharmacological interventions targeting cardiac diseases. The aim of the present study was to understand whether steady-state and kinetic force parameters of β-myosin heavy chain (βMyHC) isoform-expressing myofibrils within human embryonic stem cell-derived cardiomyocytes (hESC-CMs) differentiated in vitro resemble those of human ventricular myofibrils (hvMFs) isolated from adult donor hearts. Contractile parameters were determined using the same micromechanical method and experimental conditions for both types of myofibrils. We identified isoforms and phosphorylation of main sarcomeric proteins involved in the modulation of force generation of both, chemically demembranated hESC-CMs (d-hESC-CMs) and hvMFs. Our results indicate that at saturating Ca
2+ concentration, both human-derived contractile systems developed forces with similar rate constants (0.66 and 0.68 s-1 ), reaching maximum isometric force that was significantly smaller for d-hESC-CMs (42 kPa) than for hvMFs (94 kPa). At submaximal Ca2+ -activation, where intact cardiomyocytes normally operate, contractile parameters of d-hESC-CMs and hvMFs exhibited differences. Ca2+ sensitivity of force was higher for d-hESC-CMs (pCa50 = 6.04) than for hvMFs (pCa50 = 5.80). At half-maximum activation, the rate constant for force redevelopment was significantly faster for d-hESC-CMs (0.51 s-1 ) than for hvMFs (0.28 s-1 ). During myofibril relaxation, kinetics of the slow force decay phase were significantly faster for d-hESC-CMs (0.26 s-1 ) than for hvMFs (0.21 s-1 ), while kinetics of the fast force decay were similar and ~20x faster. Protein analysis revealed that hESC-CMs had essentially no cardiac troponin-I, and partially non-ventricular isoforms of some other sarcomeric proteins, explaining the functional discrepancies. The sarcomeric protein isoform pattern of hESC-CMs had features of human cardiomyocytes at an early developmental stage. The study indicates that morphological and ultrastructural maturation of βMyHC isoform-expressing hESC-CMs is not necessarily accompanied by ventricular-like expression of all sarcomeric proteins. Our data suggest that hPSC-CMs could provide useful tools for investigating inherited cardiac diseases affecting contractile function during early developmental stages.- Published
- 2018
- Full Text
- View/download PDF
43. Protein phosphatase 5 regulates titin phosphorylation and function at a sarcomere-associated mechanosensor complex in cardiomyocytes.
- Author
-
Krysiak J, Unger A, Beckendorf L, Hamdani N, von Frieling-Salewsky M, Redfield MM, Dos Remedios CG, Sheikh F, Gergs U, Boknik P, and Linke WA
- Subjects
- Animals, Cardiomyopathy, Dilated metabolism, Dogs, Heart Failure, Diastolic metabolism, Humans, MAP Kinase Signaling System, Mice, Mice, Transgenic, Nuclear Proteins genetics, Phosphoprotein Phosphatases genetics, Phosphorylation, Sarcomeres, Connectin metabolism, Mechanotransduction, Cellular, Myocytes, Cardiac metabolism, Nuclear Proteins metabolism, Phosphoprotein Phosphatases metabolism
- Abstract
Serine/threonine protein phosphatase 5 (PP5) is ubiquitously expressed in eukaryotic cells; however, its function in cardiomyocytes is unknown. Under basal conditions, PP5 is autoinhibited, but enzymatic activity rises upon binding of specific factors, such as the chaperone Hsp90. Here we show that PP5 binds and dephosphorylates the elastic N2B-unique sequence (N2Bus) of titin in cardiomyocytes. Using various binding and phosphorylation tests, cell-culture manipulation, and transgenic mouse hearts, we demonstrate that PP5 associates with N2Bus in vitro and in sarcomeres and is antagonistic to several protein kinases, which phosphorylate N2Bus and lower titin-based passive tension. PP5 is pathologically elevated and likely contributes to hypo-phosphorylation of N2Bus in failing human hearts. Furthermore, Hsp90-activated PP5 interacts with components of a sarcomeric, N2Bus-associated, mechanosensor complex, and blocks mitogen-activated protein-kinase signaling in this complex. Our work establishes PP5 as a compartmentalized, well-controlled phosphatase in cardiomyocytes, which regulates titin properties and kinase signaling at the myofilaments.
- Published
- 2018
- Full Text
- View/download PDF
44. Myofilament Remodeling and Function Is More Impaired in Peripartum Cardiomyopathy Compared with Dilated Cardiomyopathy and Ischemic Heart Disease.
- Author
-
Bollen IAE, Ehler E, Fleischanderl K, Bouwman F, Kempers L, Ricke-Hoch M, Hilfiker-Kleiner D, Dos Remedios CG, Krüger M, Vink A, Asselbergs FW, van Spaendonck-Zwarts KY, Pinto YM, Kuster DWD, and van der Velden J
- Subjects
- Adult, Female, Humans, Male, Middle Aged, Myocardial Ischemia physiopathology, Myocytes, Cardiac metabolism, Myofibrils metabolism, Pregnancy, Cardiomyopathies physiopathology, Cardiomyopathy, Dilated physiopathology, Myocytes, Cardiac pathology, Myofibrils pathology, Peripartum Period
- Abstract
Peripartum cardiomyopathy (PPCM) and dilated cardiomyopathy (DCM) show similarities in clinical presentation. However, although DCM patients do not recover and slowly deteriorate further, PPCM patients show either a fast cardiac deterioration or complete recovery. The aim of this study was to assess if underlying cellular changes can explain the clinical similarities and differences in the two diseases. We, therefore, assessed sarcomeric protein expression, modification, titin isoform shift, and contractile behavior of cardiomyocytes in heart tissue of PPCM and DCM patients and compared these with nonfailing controls. Heart samples from ischemic heart disease (ISHD) patients served as heart failure control samples. Passive force was only increased in PPCM samples compared with controls, whereas PPCM, DCM, and ISHD samples all showed increased myofilament Ca
2+ sensitivity. Length-dependent activation was significantly impaired in PPCM compared with controls, no impairment was observed in ISHD samples, and DCM samples showed an intermediate response. Contractile impairments were caused by impaired protein kinase A (PKA)-mediated phosphorylation because exogenous PKA restored all parameters to control levels. Although DCM samples showed reexpression of EH-myomesin, an isoform usually only expressed in the heart before birth, PPCM and ISHD did not. The lack of EH-myomesin, combined with low PKA-mediated phosphorylation of myofilament proteins and increased compliant titin isoform, may explain the increase in passive force and blunted length-dependent activation of myofilaments in PPCM samples., (Copyright © 2017 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
45. Abnormal contractility in human heart myofibrils from patients with dilated cardiomyopathy due to mutations in TTN and contractile protein genes.
- Author
-
Vikhorev PG, Smoktunowicz N, Munster AB, Copeland O, Kostin S, Montgiraud C, Messer AE, Toliat MR, Li A, Dos Remedios CG, Lal S, Blair CA, Campbell KS, Guglin M, Richter M, Knöll R, and Marston SB
- Subjects
- Biomechanical Phenomena, Cardiomyopathy, Dilated physiopathology, Heart physiopathology, Humans, Myocardial Contraction, Myofibrils genetics, Point Mutation, Cardiomyopathy, Dilated genetics, Cardiomyopathy, Dilated pathology, Connectin genetics, Mutation, Myofibrils pathology
- Abstract
Dilated cardiomyopathy (DCM) is an important cause of heart failure. Single gene mutations in at least 50 genes have been proposed to account for 25-50% of DCM cases and up to 25% of inherited DCM has been attributed to truncating mutations in the sarcomeric structural protein titin (TTNtv). Whilst the primary molecular mechanism of some DCM-associated mutations in the contractile apparatus has been studied in vitro and in transgenic mice, the contractile defect in human heart muscle has not been studied. In this study we isolated cardiac myofibrils from 3 TTNtv mutants, and 3 with contractile protein mutations (TNNI3 K36Q, TNNC1 G159D and MYH7 E1426K) and measured their contractility and passive stiffness in comparison with donor heart muscle as a control. We found that the three contractile protein mutations but not the TTNtv mutations had faster relaxation kinetics. Passive stiffness was reduced about 38% in all the DCM mutant samples. However, there was no change in maximum force or the titin N2BA/N2B isoform ratio and there was no titin haploinsufficiency. The decrease in myofibril passive stiffness was a common feature in all hearts with DCM-associated mutations and may be causative of DCM.
- Published
- 2017
- Full Text
- View/download PDF
46. Dose-Dependent Effects of the Myosin Activator Omecamtiv Mecarbil on Cross-Bridge Behavior and Force Generation in Failing Human Myocardium.
- Author
-
Mamidi R, Li J, Gresham KS, Verma S, Doh CY, Li A, Lal S, Dos Remedios CG, and Stelzer JE
- Subjects
- Cardiotonic Agents metabolism, Carrier Proteins metabolism, Case-Control Studies, Dose-Response Relationship, Drug, Heart Failure metabolism, Heart Failure physiopathology, Humans, In Vitro Techniques, Phosphorylation, Protein Binding, Sarcomeres metabolism, Signal Transduction drug effects, Time Factors, Troponin I metabolism, Troponin T metabolism, Urea metabolism, Urea pharmacology, Ventricular Remodeling, Cardiotonic Agents pharmacology, Heart Failure drug therapy, Muscle Strength drug effects, Myocardial Contraction drug effects, Myosins metabolism, Urea analogs & derivatives
- Abstract
Background: Omecamtiv mecarbil (OM) enhances systolic function in vivo by directly binding the myosin cross-bridges (XBs) in the sarcomere. However, the mechanistic details governing OM-induced modulation of XB behavior in failing human myocardium are unclear., Methods and Results: The effects of OM on steady state and dynamic XB behavior were measured in chemically skinned myocardial preparations isolated from human donor and heart failure (HF) left ventricle. HF myocardium exhibited impaired contractile function as evidenced by reduced maximal force, magnitude of XB recruitment ( P
df ), and a slowed rate of XB detachment ( krel ) at submaximal Ca2+ activations. Ca2+ sensitivity of force generation (pCa50 ) was higher in HF myocardium when compared with donor myocardium, both prior to and after OM incubations. OM incubation (0.5 and 1.0 μmol/L) enhanced force generation at submaximal Ca2+ activations in a dose-dependent manner. Notably, OM induced a slowing in krel with 1.0 μmol/L OM but not with 0.5 μmol/L OM in HF myocardium. Additionally, OM exerted other differential effects on XB behavior in HF myocardium as evidenced by a greater enhancement in Pdf and slowing in the time course of cooperative XB recruitment ( Trec ), which collectively prolonged achievement of peak force development ( Tpk ), compared with donor myocardium., Conclusions: Our findings demonstrate that OM augments force generation but also prolongs the time course of XB transitions to force-bearing states in remodeled HF myocardium, which may extend the systolic ejection time in vivo. Optimal OM dosing is critical for eliciting enhanced systolic function without excessive prolongation of systolic ejection time, which may compromise diastolic filling., (© 2017 American Heart Association, Inc.)- Published
- 2017
- Full Text
- View/download PDF
47. Special Issue of Biophysical Reviews dedicated to the 19 th IUPAB Conference in Edinburgh, Scotland (July 2017).
- Author
-
Hall D and Dos Remedios CG
- Abstract
From the 16th to the 20th of July 2017, the 19th IUPAB (International Union of Pure and Applied Biophysics) world congress was held in Edinburgh, Scotland. Organized in conjunction with the 11th EBSA (European Biophysical Societies' Association) the meeting attracted over 1430 participants, had 35 scientific sessions and 10 plenary speakers. Each invited speaker was invited to contribute either a short review or a letter article that summarized the contents of their session. This special issue of Biophysical Reviews is dedicated to providing contextual background to some of the many talks presented at the meeting in Edinburgh. The reader is referred to supplementary issue 1 of volume 46 of the European Biophysics Journal for a complete listing of abstracts for all presented talks and posters.
- Published
- 2017
- Full Text
- View/download PDF
48. The Sydney Heart Bank: improving translational research while eliminating or reducing the use of animal models of human heart disease.
- Author
-
Dos Remedios CG, Lal SP, Li A, McNamara J, Keogh A, Macdonald PS, Cooke R, Ehler E, Knöll R, Marston SB, Stelzer J, Granzier H, Bezzina C, van Dijk S, De Man F, Stienen GJM, Odeberg J, Pontén F, Linke, Linke W, and van der Velden J
- Abstract
The Sydney Heart Bank (SHB) is one of the largest human heart tissue banks in existence. Its mission is to provide high-quality human heart tissue for research into the molecular basis of human heart failure by working collaboratively with experts in this field. We argue that, by comparing tissues from failing human hearts with age-matched non-failing healthy donor hearts, the results will be more relevant than research using animal models, particularly if their physiology is very different from humans. Tissue from heart surgery must generally be used soon after collection or it significantly deteriorates. Freezing is an option but it raises concerns that freezing causes substantial damage at the cellular and molecular level. The SHB contains failing samples from heart transplant patients and others who provided informed consent for the use of their tissue for research. All samples are cryopreserved in liquid nitrogen within 40 min of their removal from the patient, and in less than 5-10 min in the case of coronary arteries and left ventricle samples. To date, the SHB has collected tissue from about 450 failing hearts (>15,000 samples) from patients with a wide range of etiologies as well as increasing numbers of cardiomyectomy samples from patients with hypertrophic cardiomyopathy. The Bank also has hearts from over 120 healthy organ donors whose hearts, for a variety of reasons (mainly tissue-type incompatibility with waiting heart transplant recipients), could not be used for transplantation. Donor hearts were collected by the St Vincent's Hospital Heart and Lung transplantation team from local hospitals or within a 4-h jet flight from Sydney. They were flushed with chilled cardioplegic solution and transported to Sydney where they were quickly cryopreserved in small samples. Failing and/or donor samples have been used by more than 60 research teams around the world, and have resulted in more than 100 research papers. The tissues most commonly requested are from donor left ventricles, but right ventricles, atria, interventricular system, and coronary arteries vessels have also been reported. All tissues are stored for long-term use in liquid N or vapor (170-180 °C), and are shipped under nitrogen vapor to avoid degradation of sensitive molecules such as RNAs and giant proteins. We present evidence that the availability of these human heart samples has contributed to a reduction in the use of animal models of human heart failure.
- Published
- 2017
- Full Text
- View/download PDF
49. Increased collagen within the transverse tubules in human heart failure.
- Author
-
Crossman DJ, Shen X, Jüllig M, Munro M, Hou Y, Middleditch M, Shrestha D, Li A, Lal S, Dos Remedios CG, Baddeley D, Ruygrok PN, and Soeller C
- Subjects
- Adult, Dystrophin metabolism, Female, Heart Failure pathology, Heart Ventricles metabolism, Humans, Male, Middle Aged, Myocytes, Cardiac pathology, Proteomics methods, Sarcolemma pathology, Collagen metabolism, Heart Failure metabolism, Myocardium metabolism, Myocytes, Cardiac metabolism, Sarcolemma metabolism
- Abstract
Aims: In heart failure transverse-tubule (t-tubule) remodelling disrupts calcium release, and contraction. T-tubules in human failing hearts exhibit increased labelling by wheat germ agglutinin (WGA), a lectin that binds to the dystrophin-associated glycoprotein complex. We hypothesized changes in this complex may explain the increased WGA labelling and contribute to t-tubule remodelling in the failing human heart. In this study we sought to identify the molecules responsible for this increased WGA labelling., Methods and Results: Confocal and super-resolution fluorescence microscopy and proteomic analyses were used to quantify left ventricle samples from healthy donors and patients with idiopathic dilated cardiomyopathy (IDCM). Confocal microscopy demonstrated both WGA and dystrophin were located at t-tubules. Super-resolution microscopy revealed that WGA labelling of t-tubules is largely located within the lumen while dystrophin was restricted to near the sarcolemma. Western blots probed with WGA reveal a 5.7-fold increase in a 140 kDa band in IDCM. Mass spectrometry identified this band as type VI collagen (Col-VI) comprised of α1(VI), α2(VI), and α3(VI) chains. Pertinently, mutations in Col-VI cause muscular dystrophy. Western blotting identified a 2.4-fold increased expression and 3.2-fold increased WGA binding of Col-VI in IDCM. Confocal images showed that Col-VI is located in the t-tubules and that their diameter increased in the IDCM samples. Super-resolution imaging revealed Col-VI was restricted to the t-tubule lumen where increases were associated with displacement in the sarcolemma as identified from dystrophin labelling. Samples were also labelled for type I, III, and IV collagen. Both confocal and super-resolution imaging identified that these collagens were also present within t-tubule lumen., Conclusion: Increased expression and labelling of collagen in IDCM samples indicates fibrosis may contribute to t-tubule remodelling in human heart failure., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2017. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
50. MYBPC3 mutations are associated with a reduced super-relaxed state in patients with hypertrophic cardiomyopathy.
- Author
-
McNamara JW, Li A, Lal S, Bos JM, Harris SP, van der Velden J, Ackerman MJ, Cooke R, and Dos Remedios CG
- Subjects
- Adolescent, Adult, Carrier Proteins physiology, Female, Genotyping Techniques, Humans, Male, Middle Aged, Muscle Relaxation genetics, Muscle Relaxation physiology, Mutation genetics, Myocardial Contraction genetics, Myocardial Contraction physiology, Myocytes, Cardiac physiology, Myosins metabolism, Myosins physiology, Young Adult, Cardiomyopathy, Hypertrophic genetics, Carrier Proteins genetics
- Abstract
The "super-relaxed state" (SRX) of myosin represents a 'reserve' of motors in the heart. Myosin heads in the SRX are bound to the thick filament and have a very low ATPase rate. Changes in the SRX are likely to modulate cardiac contractility. We previously demonstrated that the SRX is significantly reduced in mouse cardiomyocytes lacking cardiac myosin binding protein-C (cMyBP-C). Here, we report the effect of mutations in the cMyBP-C gene (MYBPC3) using samples from human patients with hypertrophic cardiomyopathy (HCM). Left ventricular (LV) samples from 11 HCM patients were obtained following myectomy surgery to relieve LV outflow tract obstruction. HCM samples were genotyped as either MYBPC3 mutation positive (MYBPC3mut) or negative (HCMsmn) and were compared to eight non-failing donor hearts. Compared to donors, only MYBPC3mut samples display a significantly diminished SRX, characterised by a decrease in both the number of myosin heads in the SRX and the lifetime of ATP turnover. These changes were not observed in HCMsmn samples. There was a positive correlation (p < 0.01) between the expression of cMyBP-C and the proportion of myosin heads in the SRX state, suggesting cMyBP-C modulates and maintains the SRX. Phosphorylation of the myosin regulatory light chain in MYBPC3mut samples was significantly decreased compared to the other groups, suggesting a potential mechanism to compensate for the diminished SRX. We conclude that by altering both contractility and sarcomeric energy requirements, a reduced SRX may be an important disease mechanism in patients with MYBPC3 mutations.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.