Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 365
Filtrer
1.
Sci Rep ; 14(1): 13727, 2024 06 14.
Article de Anglais | MEDLINE | ID: mdl-38877142

RÉSUMÉ

Connectin (also known as titin) is a giant striated muscle protein that functions as a molecular spring by providing elasticity to the sarcomere. Novex-3 is a short splice variant of connectin whose physiological function remains unknown. We have recently demonstrated using in vitro analyses that in addition to sarcomere expression, novex-3 was also expressed in cardiomyocyte nuclei exclusively during fetal life, where it provides elasticity/compliance to cardiomyocyte nuclei and promotes cardiomyocyte proliferation in the fetus, suggesting a non-sarcomeric function. Here, we analyzed novex-3 knockout mice to assess the involvement of this function in cardiac pathophysiology in vivo. Deficiency of novex-3 compromised fetal cardiomyocyte proliferation and induced the enlargement of individual cardiomyocytes in neonates. In adults, novex-3 deficiency resulted in chamber dilation and systolic dysfunction, associated with Ca2+ dysregulation, resulting in a reduced life span. Mechanistic analyses revealed a possible association between impaired proliferation and abnormal nuclear mechanics, including stiffer nuclei positioned peripherally with stabilized circumnuclear microtubules in knockout cardiomyocytes. Although the underlying causal relationships were not fully elucidated, these data show that novex-3 has a vital non-sarcomeric function in cardiac pathophysiology and serves as an early contributor to cardiomyocyte proliferation.


Sujet(s)
Noyau de la cellule , Prolifération cellulaire , Connectine , Souris knockout , Myocytes cardiaques , Animaux , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Souris , Noyau de la cellule/métabolisme , Connectine/génétique , Connectine/métabolisme , Sarcomères/métabolisme , Protéines du muscle/métabolisme , Protéines du muscle/génétique , Protéines du muscle/déficit , Calcium/métabolisme
2.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-38928324

RÉSUMÉ

Heart failure with preserved ejection fraction (HFpEF) is characterized by biomechanically dysfunctional cardiomyocytes. Underlying cellular changes include perturbed myocardial titin expression and titin hypophosphorylation leading to titin filament stiffening. Beside these well-studied alterations at the cardiomyocyte level, exercise intolerance is another hallmark of HFpEF caused by molecular alterations in skeletal muscle (SKM). Currently, there is a lack of data regarding titin modulation in the SKM of HFpEF. Therefore, the aim of the present study was to analyze molecular alterations in limb SKM (tibialis anterior (TA)) and in the diaphragm (Dia), as a more central SKM, with a focus on titin, titin phosphorylation, and contraction-regulating proteins. This study was performed with muscle tissue, obtained from 32-week old female ZSF-1 rats, an established a HFpEF rat model. Our results showed a hyperphosphorylation of titin in limb SKM, based on enhanced phosphorylation at the PEVK region, which is known to lead to titin filament stiffening. This hyperphosphorylation could be reversed by high-intensity interval training (HIIT). Additionally, a negative correlation occurring between the phosphorylation state of titin and the muscle force in the limb SKM was evident. For the Dia, no alterations in the phosphorylation state of titin could be detected. Supported by data of previous studies, this suggests an exercise effect of the Dia in HFpEF. Regarding the expression of contraction regulating proteins, significant differences between Dia and limb SKM could be detected, supporting muscle atrophy and dysfunction in limb SKM, but not in the Dia. Altogether, these data suggest a correlation between titin stiffening and the appearance of exercise intolerance in HFpEF, as well as a differential regulation between different SKM groups.


Sujet(s)
Connectine , Muscle diaphragme , Modèles animaux de maladie humaine , Défaillance cardiaque , Muscles squelettiques , Animaux , Défaillance cardiaque/métabolisme , Défaillance cardiaque/physiopathologie , Défaillance cardiaque/anatomopathologie , Rats , Muscle diaphragme/métabolisme , Muscle diaphragme/physiopathologie , Muscle diaphragme/anatomopathologie , Connectine/métabolisme , Phosphorylation , Femelle , Muscles squelettiques/métabolisme , Muscles squelettiques/physiopathologie , Muscles squelettiques/anatomopathologie , Débit systolique , Contraction musculaire , Conditionnement physique d'animal , Protéines du muscle/métabolisme
3.
Nat Commun ; 15(1): 4496, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38802383

RÉSUMÉ

Titin N2B unique sequence (N2B-us) is a 572 amino acid sequence that acts as an elastic spring to regulate muscle passive elasticity. It is thought to lack stable tertiary structures and is a force-bearing region that is regulated by mechanical stretching. In this study, the conformation of N2B-us and its interaction with four-and-a-half LIM domain protein 2 (FHL2) are investigated using AlphaFold2 predictions and single-molecule experimental validation. Surprisingly, a stable alpha/beta structural domain is predicted and confirmed in N2B-us that can be mechanically unfolded at forces of a few piconewtons. Additionally, more than twenty FHL2 LIM domain binding sites are predicted to spread throughout N2B-us. Single-molecule manipulation experiments reveals the force-dependent binding of FHL2 to the N2B-us structural domain. These findings provide insights into the mechano-sensing functions of N2B-us and its interactions with FHL2.


Sujet(s)
Connectine , Protéines à homéodomaine LIM , Liaison aux protéines , Domaines protéiques , Facteurs de transcription , Protéines à homéodomaine LIM/métabolisme , Protéines à homéodomaine LIM/composition chimique , Protéines à homéodomaine LIM/génétique , Connectine/métabolisme , Connectine/composition chimique , Connectine/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/composition chimique , Facteurs de transcription/génétique , Sites de fixation , Humains , Animaux , Protéines du muscle/métabolisme , Protéines du muscle/composition chimique , Protéines du muscle/génétique , Séquence d'acides aminés
4.
J Physiol ; 602(12): 2751-2762, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38695322

RÉSUMÉ

There is a growing appreciation that regulation of muscle contraction requires both thin filament and thick filament activation in order to fully activate the sarcomere. The prevailing mechano-sensing model for thick filament activation was derived from experiments on fast-twitch muscle. We address the question whether, or to what extent, this mechanism can be extrapolated to the slow muscle in the hearts of large mammals, including humans. We investigated the similarities and differences in structural signatures of thick filament activation in porcine myocardium as compared to fast rat extensor digitorum longus (EDL) skeletal muscle under relaxed conditions and sub-maximal contraction using small angle X-ray diffraction. Thick and thin filaments were found to adopt different structural configurations under relaxing conditions, and myosin heads showed different changes in configuration upon sub-maximal activation, when comparing the two muscle types. Titin was found to have an X-ray diffraction signature distinct from those of the overall thick filament backbone, and its spacing change appeared to be positively correlated to the force exerted on the thick filament. Structural changes in fast EDL muscle were found to be consistent with the mechano-sensing model. In porcine myocardium, however, the structural basis of mechano-sensing is blunted suggesting the need for additional activation mechanism(s) in slow cardiac muscle. These differences in thick filament regulation can be related to their different physiological roles where fast muscle is optimized for rapid, burst-like, contractions, and the slow cardiac muscle in large mammalian hearts adopts a more finely tuned, graded response to allow for their substantial functional reserve. KEY POINTS: Both thin filament and thick filament activation are required to fully activate the sarcomere. Thick and thin filaments adopt different structural configurations under relaxing conditions, and myosin heads show different changes in configuration upon sub-maximal activation in fast extensor digitorum longus muscle and slow porcine cardiac muscle. Titin has an X-ray diffraction signature distinct from those of the overall thick filament backbone and this titin reflection spacing change appeared to be directly proportional to the force exerted on the thick filament. Mechano-sensing is blunted in porcine myocardium suggesting the need for additional activation mechanism(s) in slow cardiac muscle. Fast skeletal muscle is optimized for rapid, burst-like contractions, and the slow cardiac muscle in large mammalian hearts adopts a more finely tuned graded response to allow for their substantial functional reserve.


Sujet(s)
Myocarde , Animaux , Suidae , Myocarde/métabolisme , Connectine/métabolisme , Rats , Mâle , Fibres musculaires à contraction rapide/physiologie , Fibres musculaires à contraction rapide/métabolisme , Sarcomères/physiologie , Sarcomères/métabolisme , Fibres musculaires à contraction lente/physiologie , Fibres musculaires à contraction lente/métabolisme , Muscles squelettiques/physiologie , Muscles squelettiques/métabolisme , Diffraction des rayons X , Contraction musculaire/physiologie , Myosines/métabolisme , Myosines/physiologie
5.
J Biol Chem ; 300(5): 107254, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38569934

RÉSUMÉ

Nesprins comprise a family of multi-isomeric scaffolding proteins, forming the linker of nucleoskeleton-and-cytoskeleton complex with lamin A/C, emerin and SUN1/2 at the nuclear envelope. Mutations in nesprin-1/-2 are associated with Emery-Dreifuss muscular dystrophy (EDMD) with conduction defects and dilated cardiomyopathy (DCM). We have previously observed sarcomeric staining of nesprin-1/-2 in cardiac and skeletal muscle, but nesprin function in this compartment remains unknown. In this study, we show that specific nesprin-2 isoforms are highly expressed in cardiac muscle and localize to the Z-disc and I band of the sarcomere. Expression of GFP-tagged nesprin-2 giant spectrin repeats 52 to 53, localized to the sarcomere of neonatal rat cardiomyocytes. Yeast two-hybrid screening of a cardiac muscle cDNA library identified telethonin and four-and-half LIM domain (FHL)-2 as potential nesprin-2 binding partners. GST pull-down and immunoprecipitation confirmed the individual interactions between nesprin-2/telethonin and nesprin-2/FHL-2, and showed that nesprin-2 and telethonin binding was dependent on telethonin phosphorylation status. Importantly, the interactions between these binding partners were impaired by mutations in nesprin-2, telethonin, and FHL-2 identified in EDMD with DCM and hypertrophic cardiomyopathy patients. These data suggest that nesprin-2 is a novel sarcomeric scaffold protein that may potentially participate in the maintenance and/or regulation of sarcomeric organization and function.


Sujet(s)
Connectine , Protéines à domaine LIM , Protéines du muscle , Myocytes cardiaques , Protéines de tissu nerveux , Protéines nucléaires , Sarcomères , Animaux , Humains , Souris , Rats , Connectine/métabolisme , Connectine/génétique , Protéines du cytosquelette/métabolisme , Protéines du cytosquelette/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Protéines à homéodomaine LIM , Protéines des microfilaments/métabolisme , Protéines des microfilaments/génétique , Protéines du muscle/métabolisme , Protéines du muscle/génétique , Myocytes cardiaques/métabolisme , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Liaison aux protéines , Sarcomères/métabolisme , Facteurs de transcription
6.
J Mol Cell Cardiol ; 191: 40-49, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38604403

RÉSUMÉ

The heart has the ability to detect and respond to changes in mechanical load through a process called mechanotransduction. In this study, we focused on investigating the role of the cardiac-specific N2B element within the spring region of titin, which has been proposed to function as a mechanosensor. To assess its significance, we conducted experiments using N2B knockout (KO) mice and wildtype (WT) mice, subjecting them to three different conditions: 1) cardiac pressure overload induced by transverse aortic constriction (TAC), 2) volume overload caused by aortocaval fistula (ACF), and 3) exercise-induced hypertrophy through swimming. Under conditions of pressure overload (TAC), both genotypes exhibited similar hypertrophic responses. In contrast, WT mice displayed robust left ventricular hypertrophy after one week of volume overload (ACF), while the KO mice failed to undergo hypertrophy and experienced a high mortality rate. Similarly, swim exercise-induced hypertrophy was significantly reduced in the KO mice. RNA-Seq analysis revealed an abnormal ß-adrenergic response to volume overload in the KO mice, as well as a diminished response to isoproterenol-induced hypertrophy. Because it is known that the N2B element interacts with the four-and-a-half LIM domains 1 and 2 (FHL1 and FHL2) proteins, both of which have been associated with mechanotransduction, we evaluated these proteins. Interestingly, while volume-overload resulted in FHL1 protein expression levels that were comparable between KO and WT mice, FHL2 protein levels were reduced by over 90% in the KO mice compared to WT. This suggests that in response to volume overload, FHL2 might act as a signaling mediator between the N2B element and downstream signaling pathways. Overall, our study highlights the importance of the N2B element in mechanosensing during volume overload, both in physiological and pathological settings.


Sujet(s)
Connectine , Mécanotransduction cellulaire , Souris knockout , Animaux , Souris , Connectine/métabolisme , Connectine/génétique , Hypertrophie ventriculaire gauche/métabolisme , Hypertrophie ventriculaire gauche/physiopathologie , Hypertrophie ventriculaire gauche/génétique , Myocarde/métabolisme , Myocarde/anatomopathologie , Mâle , Conditionnement physique d'animal , Protéines à homéodomaine LIM/métabolisme , Protéines à homéodomaine LIM/génétique , Modèles animaux de maladie humaine , Protéines du muscle/métabolisme , Protéines du muscle/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Protein kinases , Protéines et peptides de signalisation intracellulaire
7.
J Nanobiotechnology ; 22(1): 191, 2024 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-38637832

RÉSUMÉ

BACKGROUND: Exosomes assume a pivotal role as essential mediators of intercellular communication within tumor microenvironments. Within this context, long noncoding RNAs (LncRNAs) have been observed to be preferentially sorted into exosomes, thus exerting regulatory control over the initiation and progression of cancer through diverse mechanisms. RESULTS: Exosomes were successfully isolated from cholangiocarcinoma (CCA) CTCs organoid and healthy human serum. Notably, the LncRNA titin-antisense RNA1 (TTN-AS1) exhibited a conspicuous up-regulation within CCA CTCs organoid derived exosomes. Furthermore, a significant elevation of TTN-AS1 expression was observed in tumor tissues, as well as in blood and serum exosomes from patients afflicted with CCA. Importantly, this hightened TTN-AS1 expression in serum exosomes of CCA patients manifested a strong correlation with both lymph node metastasis and TNM staging. Remarkably, both CCA CTCs organoid-derived exosomes and CCA cells-derived exosomes featuring pronounced TTN-AS1 expression demonstrated the capability to the proliferation and migratory potential of CCA cells. Validation of these outcomes was conducted in vivo experiments. CONCLUSIONS: In conclusion, our study elucidating that CCA CTCs-derived exosomes possess the capacity to bolster the metastasis tendencies of CCA cells by transporting TTN-AS1. These observations underscore the potential of TTN-AS1 within CTCs-derived exosomes to serve as a promising biomarker for the diagnosis and therapeutic management of CCA.


Sujet(s)
Cholangiocarcinome , Exosomes , microARN , Cellules tumorales circulantes , ARN bactérien , ARN long non codant , Humains , microARN/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , Exosomes/métabolisme , Connectine/génétique , Connectine/métabolisme , Lignée cellulaire tumorale , Évolution de la maladie , Prolifération cellulaire , Mouvement cellulaire , Cholangiocarcinome/génétique , Cholangiocarcinome/métabolisme , Cholangiocarcinome/anatomopathologie , Régulation de l'expression des gènes tumoraux , Microenvironnement tumoral
8.
Nat Genet ; 56(3): 395-407, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38429495

RÉSUMÉ

In digenic inheritance, pathogenic variants in two genes must be inherited together to cause disease. Only very few examples of digenic inheritance have been described in the neuromuscular disease field. Here we show that predicted deleterious variants in SRPK3, encoding the X-linked serine/argenine protein kinase 3, lead to a progressive early onset skeletal muscle myopathy only when in combination with heterozygous variants in the TTN gene. The co-occurrence of predicted deleterious SRPK3/TTN variants was not seen among 76,702 healthy male individuals, and statistical modeling strongly supported digenic inheritance as the best-fitting model. Furthermore, double-mutant zebrafish (srpk3-/-; ttn.1+/-) replicated the myopathic phenotype and showed myofibrillar disorganization. Transcriptome data suggest that the interaction of srpk3 and ttn.1 in zebrafish occurs at a post-transcriptional level. We propose that digenic inheritance of deleterious changes impacting both the protein kinase SRPK3 and the giant muscle protein titin causes a skeletal myopathy and might serve as a model for other genetic diseases.


Sujet(s)
Maladies musculaires , Danio zébré , Animaux , Humains , Mâle , Connectine/génétique , Connectine/métabolisme , Muscles squelettiques , Maladies musculaires/génétique , Maladies musculaires/métabolisme , Maladies musculaires/anatomopathologie , Mutation , Danio zébré/génétique
9.
J Mol Cell Cardiol ; 190: 13-23, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38462126

RÉSUMÉ

Mutations in cardiac myosin-binding protein C (cMyBP-C) or titin may respectively lead to hypertrophic (HCM) or dilated (DCM) cardiomyopathies. The mechanisms leading to these phenotypes remain unclear because of the challenge of translating cellular abnormalities to whole-heart and system function. We developed and validated a novel computer model of calcium-contraction coupling incorporating the role of cMyBP-C and titin based on the key assumptions: 1) tension in the thick filament promotes cross-bridge attachment mechanochemically, 2) with increasing titin tension, more myosin heads are unlocked for attachment, and 3) cMyBP-C suppresses cross-bridge attachment. Simulated stationary calcium-tension curves, isotonic and isometric contractions, and quick release agreed with experimental data. The model predicted that a loss of cMyBP-C function decreases the steepness of the calcium-tension curve, and that more compliant titin decreases the level of passive and active tension and its dependency on sarcomere length. Integrating this cellular model in the CircAdapt model of the human heart and circulation showed that a loss of cMyBP-C function resulted in HCM-like hemodynamics with higher left ventricular end-diastolic pressures and smaller volumes. More compliant titin led to higher diastolic pressures and ventricular dilation, suggesting DCM-like hemodynamics. The novel model of calcium-contraction coupling incorporates the role of cMyBP-C and titin. Its coupling to whole-heart mechanics translates changes in cellular calcium-contraction coupling to changes in cardiac pump and circulatory function and identifies potential mechanisms by which cMyBP-C and titin abnormalities may develop into HCM and DCM phenotypes. This modeling platform may help identify distinct mechanisms underlying clinical phenotypes in cardiac diseases.


Sujet(s)
Calcium , Protéines de transport , Connectine , Contraction myocardique , Humains , Connectine/métabolisme , Connectine/génétique , Protéines de transport/métabolisme , Calcium/métabolisme , Sarcomères/métabolisme , Modèles cardiovasculaires , Simulation numérique , Animaux , Coeur/physiopathologie , Coeur/physiologie
10.
Int J Mol Sci ; 25(5)2024 Feb 28.
Article de Anglais | MEDLINE | ID: mdl-38474037

RÉSUMÉ

Protein kinase D (PKD) enzymes play important roles in regulating myocardial contraction, hypertrophy, and remodeling. One of the proteins phosphorylated by PKD is titin, which is involved in myofilament function. In this study, we aimed to investigate the role of PKD in cardiomyocyte function under conditions of oxidative stress. To do this, we used mice with a cardiomyocyte-specific knock-out of Prkd1, which encodes PKD1 (Prkd1loxP/loxP; αMHC-Cre; PKD1 cKO), as well as wild type littermate controls (Prkd1loxP/loxP; WT). We isolated permeabilized cardiomyocytes from PKD1 cKO mice and found that they exhibited increased passive stiffness (Fpassive), which was associated with increased oxidation of titin, but showed no change in titin ubiquitination. Additionally, the PKD1 cKO mice showed increased myofilament calcium (Ca2+) sensitivity (pCa50) and reduced maximum Ca2+-activated tension. These changes were accompanied by increased oxidation and reduced phosphorylation of the small myofilament protein cardiac myosin binding protein C (cMyBPC), as well as altered phosphorylation levels at different phosphosites in troponin I (TnI). The increased Fpassive and pCa50, and the reduced maximum Ca2+-activated tension were reversed when we treated the isolated permeabilized cardiomyocytes with reduced glutathione (GSH). This indicated that myofilament protein oxidation contributes to cardiomyocyte dysfunction. Furthermore, the PKD1 cKO mice exhibited increased oxidative stress and increased expression of pro-inflammatory markers interleukin (IL)-6, IL-18, and tumor necrosis factor alpha (TNF-α). Both oxidative stress and inflammation contributed to an increase in microtubule-associated protein 1 light chain 3 (LC3)-II levels and heat shock response by inhibiting the mammalian target of rapamycin (mTOR) in the PKD1 cKO mouse myocytes. These findings revealed a previously unknown role for PKD1 in regulating diastolic passive properties, myofilament Ca2+ sensitivity, and maximum Ca2+-activated tension under conditions of oxidative stress. Finally, we emphasized the importance of PKD1 in maintaining the balance of oxidative stress and inflammation in the context of autophagy, as well as cardiomyocyte function.


Sujet(s)
Myofibrilles , Protéine kinase C , Maturation post-traductionnelle des protéines , Souris , Animaux , Connectine/métabolisme , Myofibrilles/métabolisme , Myocytes cardiaques/métabolisme , Phosphorylation , Protéines des microfilaments/métabolisme , Homéostasie , Inflammation/métabolisme , Calcium/métabolisme , Mammifères/métabolisme
11.
Biosci Trends ; 18(1): 105-107, 2024 Mar 19.
Article de Anglais | MEDLINE | ID: mdl-38325822

RÉSUMÉ

Lactylation of α-myosin heavy chain (α-MHC) has recently been reported to preserve sarcomeric structure and function and attenuate the development of heart failure. Specifically, lactylation enhanced the interaction of α-MHC with the sarcomeric protein Titin, thereby maintaining normal sarcomeric structure and myocardial contractile function. Furthermore, the administration of lactate or inhibition of lactate efflux potentially treats heart failure by restoring lactylation of α-MHC and the interaction of α-MHC with Titin. This finding highlights the significant role of α-MHC lactylation in myocardial diseases and presents a new therapeutic target for the treatment of heart failure.


Sujet(s)
Défaillance cardiaque , Acide lactique , Humains , Connectine/métabolisme , Acide lactique/métabolisme , Défaillance cardiaque/métabolisme , Myocarde/métabolisme , Protéines/métabolisme , Chaînes lourdes de myosine/métabolisme
12.
J Clin Invest ; 134(2)2024 Jan 16.
Article de Anglais | MEDLINE | ID: mdl-38226618

RÉSUMÉ

Titin (TTN) is one of the largest and most complex proteins expressed in humans, and truncation variants are the most prevalent genetic lesion identified in individuals with dilated cardiomyopathy (DCM) or other disorders of impaired cardiac contractility. Two reports in this issue of the JCI shed light on a potential mechanism involving truncated TTN sarcomere integration and the potential for disruption of sarcomere structural integrity. Kellermayer, Tordai, and colleagues confirmed the presence of truncated TTN protein in human DCM samples. McAfee and authors developed a patient-specific TTN antibody to study truncated TTN subcellular localization and to explore its functional consequences. A "poison peptide" mechanism emerges that inspires alternative therapeutic approaches while opening new lines for inquiry, such as the role of haploinsufficiency of full-length TTN protein, mechanisms explaining sarcomere dysfunction, and explanations for variable penetrance.


Sujet(s)
Cardiomyopathie dilatée , Sarcomères , Humains , Connectine/génétique , Connectine/métabolisme , Sarcomères/métabolisme , Cardiomyopathie dilatée/métabolisme , Pénétrance , Mutation
13.
J Clin Invest ; 134(2)2024 Jan 16.
Article de Anglais | MEDLINE | ID: mdl-37962957

RÉSUMÉ

Heterozygous (HET) truncating variant mutations in the TTN gene (TTNtvs), encoding the giant titin protein, are the most common genetic cause of dilated cardiomyopathy (DCM). However, the molecular mechanisms by which TTNtv mutations induce DCM are controversial. Here, we studied 127 clinically identified DCM human cardiac samples with next-generation sequencing (NGS), high-resolution gel electrophoresis, Western blot analysis, and super-resolution microscopy in order to dissect the structural and functional consequences of TTNtv mutations. The occurrence of TTNtv was found to be 15% in the DCM cohort. Truncated titin proteins matching, by molecular weight, the gene sequence predictions were detected in the majority of the TTNtv+ samples. Full-length titin was reduced in TTNtv+ compared with TTNtv- samples. Proteomics analysis of washed myofibrils and stimulated emission depletion (STED) super-resolution microscopy of myocardial sarcomeres labeled with sequence-specific anti-titin antibodies revealed that truncated titin was structurally integrated into the sarcomere. Sarcomere length-dependent anti-titin epitope position, shape, and intensity analyses pointed at possible structural defects in the I/A junction and the M-band of TTNtv+ sarcomeres, which probably contribute, possibly via faulty mechanosensor function, to the development of manifest DCM.


Sujet(s)
Cardiomyopathie dilatée , Connectine , Humains , Cardiomyopathie dilatée/génétique , Connectine/génétique , Connectine/métabolisme , Coeur , Sarcomères/génétique , Sarcomères/métabolisme
15.
Cardiovasc Res ; 120(1): 56-68, 2024 02 27.
Article de Anglais | MEDLINE | ID: mdl-37890031

RÉSUMÉ

AIMS: RNA binding proteins play essential roles in mediating RNA splicing and are key post-transcriptional regulators in the heart. Our recent study demonstrated that RBPMS (RNA binding protein with multiple splicing) is crucial for cardiac development through modulating mRNA splicing, but little is known about its functions in the adult heart. In this study, we aim to characterize the post-natal cardiac function of Rbpms and its mechanism of action. METHODS AND RESULTS: We generated a cardiac-specific knockout mouse line and found that cardiac-specific loss of Rbpms caused severe cardiomyocyte contractile defects, leading to dilated cardiomyopathy and early lethality in adult mice. We showed by proximity-dependent biotin identification assay and mass spectrometry that RBPMS associates with spliceosome factors and other RNA binding proteins, such as RBM20, that are important in cardiac function. We performed paired-end RNA sequencing and RT-PCR and found that RBPMS regulates mRNA alternative splicing of genes associated with sarcomere structure and function, such as Ttn, Pdlim5, and Nexn, generating new protein isoforms. Using a minigene splicing reporter assay, we determined that RBPMS regulates target gene splicing through recognizing tandem intronic CAC motifs. We also showed that RBPMS knockdown in human induced pluripotent stem cell-derived cardiomyocytes impaired cardiomyocyte contraction. CONCLUSION: This study identifies RBPMS as an important regulator of cardiomyocyte contraction and cardiac function by modulating sarcomeric gene alternative splicing.


Sujet(s)
Épissage alternatif , Cellules souches pluripotentes induites , Animaux , Humains , Souris , Connectine/métabolisme , Cellules souches pluripotentes induites/métabolisme , Souris knockout , Myocytes cardiaques/métabolisme , ARN/métabolisme , Épissage des ARN , ARN messager/génétique , ARN messager/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme
16.
Adv Physiol Educ ; 48(1): 92-96, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38059284

RÉSUMÉ

Given the recently proposed three-filament theory of muscle contraction, we present a low-cost physical sarcomere model aimed at illustrating the role of titin in the production of active force in skeletal muscle. With inexpensive materials, it is possible to illustrate actin-myosin cross-bridge interactions between the thick and thin filaments and demonstrate the two different mechanisms by which titin is thought to contribute to active and passive muscle force. Specifically, the model illustrates how titin, a molecule with springlike properties, may increase its stiffness by binding free calcium upon muscle activation and reducing its extensible length by attaching itself to actin, resulting in the greater force-generating capacity after an active than a passive elongation that has been observed experimentally. The model is simple to build and manipulate, and demonstration to high school students was shown to result in positive perception and improved understanding of the otherwise complex titin-related mechanisms of force production in skeletal and cardiac muscles.NEW & NOTEWORTHY Our physical sarcomere model illustrates not only the classic view of muscle contraction, the sliding filament and cross-bridge theories, but also the newly discovered role of titin in force regulation, called the three-filament theory. The model allows for easy visualization of the role of titin in muscle contraction and aids in explaining complex muscle properties that are not captured by the traditional cross-bridge theory.


Sujet(s)
Actines , Sarcomères , Humains , Sarcomères/physiologie , Connectine/métabolisme , Contraction musculaire/physiologie , Muscles squelettiques
17.
Nature ; 623(7988): 863-871, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37914933

RÉSUMÉ

The thick filament is a key component of sarcomeres, the basic units of striated muscle1. Alterations in thick filament proteins are associated with familial hypertrophic cardiomyopathy and other heart and muscle diseases2. Despite the central importance of the thick filament, its molecular organization remains unclear. Here we present the molecular architecture of native cardiac sarcomeres in the relaxed state, determined by cryo-electron tomography. Our reconstruction of the thick filament reveals the three-dimensional organization of myosin, titin and myosin-binding protein C (MyBP-C). The arrangement of myosin molecules is dependent on their position along the filament, suggesting specialized capacities in terms of strain susceptibility and force generation. Three pairs of titin-α and titin-ß chains run axially along the filament, intertwining with myosin tails and probably orchestrating the length-dependent activation of the sarcomere. Notably, whereas the three titin-α chains run along the entire length of the thick filament, titin-ß chains do not. The structure also demonstrates that MyBP-C bridges thin and thick filaments, with its carboxy-terminal region binding to the myosin tails and directly stabilizing the OFF state of the myosin heads in an unforeseen manner. These results provide a foundation for future research investigating muscle disorders involving sarcomeric components.


Sujet(s)
Myosines cardiaques , Myocarde , Sarcomères , Connectine/composition chimique , Connectine/métabolisme , Connectine/ultrastructure , Cryomicroscopie électronique , Tomographie en microscopie électronique , Myocarde/composition chimique , Myocarde/cytologie , Myocarde/ultrastructure , Sarcomères/composition chimique , Sarcomères/métabolisme , Sarcomères/ultrastructure , Myosines cardiaques/composition chimique , Myosines cardiaques/métabolisme , Myosines cardiaques/ultrastructure
18.
Nature ; 623(7988): 853-862, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37914935

RÉSUMÉ

Pumping of the heart is powered by filaments of the motor protein myosin that pull on actin filaments to generate cardiac contraction. In addition to myosin, the filaments contain cardiac myosin-binding protein C (cMyBP-C), which modulates contractility in response to physiological stimuli, and titin, which functions as a scaffold for filament assembly1. Myosin, cMyBP-C and titin are all subject to mutation, which can lead to heart failure. Despite the central importance of cardiac myosin filaments to life, their molecular structure has remained a mystery for 60 years2. Here we solve the structure of the main (cMyBP-C-containing) region of the human cardiac filament using cryo-electron microscopy. The reconstruction reveals the architecture of titin and cMyBP-C and shows how myosin's motor domains (heads) form three different types of motif (providing functional flexibility), which interact with each other and with titin and cMyBP-C to dictate filament architecture and function. The packing of myosin tails in the filament backbone is also resolved. The structure suggests how cMyBP-C helps to generate the cardiac super-relaxed state3; how titin and cMyBP-C may contribute to length-dependent activation4; and how mutations in myosin and cMyBP-C might disturb interactions, causing disease5,6. The reconstruction resolves past uncertainties and integrates previous data on cardiac muscle structure and function. It provides a new paradigm for interpreting structural, physiological and clinical observations, and for the design of potential therapeutic drugs.


Sujet(s)
Myosines cardiaques , Cryomicroscopie électronique , Myocarde , Humains , Myosines cardiaques/composition chimique , Myosines cardiaques/métabolisme , Myosines cardiaques/ultrastructure , Protéines de transport/composition chimique , Protéines de transport/métabolisme , Protéines de transport/ultrastructure , Connectine/composition chimique , Connectine/métabolisme , Connectine/ultrastructure , Myocarde/composition chimique , Myocarde/ultrastructure
19.
Physiol Rep ; 11(22): e15788, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37985159

RÉSUMÉ

Titin-dependent stiffening of cardiomyocytes is a significant contributor to left ventricular (LV) diastolic dysfunction in heart failure with preserved LV ejection fraction (HFpEF). Small heat shock proteins (HSPs), such as HSPB5 and HSPB1, protect titin and administration of HSPB5 in vitro lowers cardiomyocyte stiffness in pressure-overload hypertrophy. In humans, oral treatment with geranylgeranylacetone (GGA) increases myocardial HSP expression, but the functional implications are unknown. Our objective was to investigate whether oral GGA treatment lowers cardiomyocyte stiffness and attenuates LV diastolic dysfunction in a rat model of the cardiometabolic syndrome. Twenty-one-week-old male lean (n = 10) and obese (n = 20) ZSF1 rats were studied, and obese rats were randomized to receive GGA (200 mg/kg/day) or vehicle by oral gavage for 4 weeks. Echocardiography and cardiac catheterization were performed before sacrifice at 25 weeks of age. Titin-based stiffness (Fpassive ) was determined by force measurements in relaxing solution with 100 nM [Ca2+ ] in permeabilized cardiomyocytes at sarcomere lengths (SL) ranging from 1.8 to 2.4 µm. In obese ZSF1 rats, GGA reduced isovolumic relaxation time of the LV without affecting blood pressure, EF or LV weight. In cardiomyocytes, GGA increased myofilament-bound HSPB5 and HSPB1 expression. Vehicle-treated obese rats exhibited higher cardiomyocyte stiffness at all SLs compared to lean rats, while GGA reduced stiffness at SL 2.0 µm. In obese ZSF1 rats, oral GGA treatment improves cardiomyocyte stiffness by increasing myofilament-bound HSPB1 and HSPB5. GGA could represent a potential novel therapy for the early stage of diastolic dysfunction in the cardiometabolic syndrome.


Sujet(s)
Défaillance cardiaque , Syndrome métabolique X , Dysfonction ventriculaire gauche , Humains , Rats , Mâle , Animaux , Myocytes cardiaques/métabolisme , Connectine/métabolisme , Syndrome métabolique X/traitement médicamenteux , Syndrome métabolique X/métabolisme , Débit systolique/physiologie , Obésité/traitement médicamenteux , Obésité/métabolisme
20.
Elife ; 122023 11 03.
Article de Anglais | MEDLINE | ID: mdl-37921850

RÉSUMÉ

Sarcomeres are the basic contractile units within cardiac myocytes, and the collective shortening of sarcomeres aligned along myofibrils generates the force driving the heartbeat. The alignment of the individual sarcomeres is important for proper force generation, and misaligned sarcomeres are associated with diseases, including cardiomyopathies and COVID-19. The actin bundling protein, α-actinin-2, localizes to the 'Z-Bodies" of sarcomere precursors and the 'Z-Lines' of sarcomeres, and has been used previously to assess sarcomere assembly and maintenance. Previous measurements of α-actinin-2 organization have been largely accomplished manually, which is time-consuming and has hampered research progress. Here, we introduce sarcApp, an image analysis tool that quantifies several components of the cardiac sarcomere and their alignment in muscle cells and tissue. We first developed sarcApp to utilize deep learning-based segmentation and real space quantification to measure α-actinin-2 structures and determine the organization of both precursors and sarcomeres/myofibrils. We then expanded sarcApp to analyze 'M-Lines' using the localization of myomesin and a protein that connects the Z-Lines to the M-Line (titin). sarcApp produces 33 distinct measurements per cell and 24 per myofibril that allow for precise quantification of changes in sarcomeres, myofibrils, and their precursors. We validated this system with perturbations to sarcomere assembly. We found perturbations that affected Z-Lines and M-Lines differently, suggesting that they may be regulated independently during sarcomere assembly.


Sujet(s)
Myocytes cardiaques , Sarcomères , Sarcomères/métabolisme , Myocytes cardiaques/métabolisme , Actinine/métabolisme , Myofibrilles/métabolisme , Connectine/métabolisme , Logiciel
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...