Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.383
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Nature ; 623(7988): 863-871, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37914933

RESUMO

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.


Assuntos
Miosinas Cardíacas , Miocárdio , Sarcômeros , Conectina/química , Conectina/metabolismo , Conectina/ultraestrutura , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Miocárdio/química , Miocárdio/citologia , Miocárdio/ultraestrutura , Sarcômeros/química , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/ultraestrutura
2.
Nature ; 623(7988): 853-862, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37914935

RESUMO

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.


Assuntos
Miosinas Cardíacas , Microscopia Crioeletrônica , Miocárdio , Humanos , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/ultraestrutura , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas de Transporte/ultraestrutura , Conectina/química , Conectina/metabolismo , Conectina/ultraestrutura , Miocárdio/química , Miocárdio/ultraestrutura
3.
Nature ; 619(7969): 410-419, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37196677

RESUMO

Voltage-gated ion channels (VGICs) comprise multiple structural units, the assembly of which is required for function1,2. Structural understanding of how VGIC subunits assemble and whether chaperone proteins are required is lacking. High-voltage-activated calcium channels (CaVs)3,4 are paradigmatic multisubunit VGICs whose function and trafficking are powerfully shaped by interactions between pore-forming CaV1 or CaV2 CaVα1 (ref. 3), and the auxiliary CaVß5 and CaVα2δ subunits6,7. Here we present cryo-electron microscopy structures of human brain and cardiac CaV1.2 bound with CaVß3 to a chaperone-the endoplasmic reticulum membrane protein complex (EMC)8,9-and of the assembled CaV1.2-CaVß3-CaVα2δ-1 channel. These structures provide a view of an EMC-client complex and define EMC sites-the transmembrane (TM) and cytoplasmic (Cyto) docks; interaction between these sites and the client channel causes partial extraction of a pore subunit and splays open the CaVα2δ-interaction site. The structures identify the CaVα2δ-binding site for gabapentinoid anti-pain and anti-anxiety drugs6, show that EMC and CaVα2δ interactions with the channel are mutually exclusive, and indicate that EMC-to-CaVα2δ hand-off involves a divalent ion-dependent step and CaV1.2 element ordering. Disruption of the EMC-CaV complex compromises CaV function, suggesting that the EMC functions as a channel holdase that facilitates channel assembly. Together, the structures reveal a CaV assembly intermediate and EMC client-binding sites that could have wide-ranging implications for the biogenesis of VGICs and other membrane proteins.


Assuntos
Canais de Cálcio Tipo L , Retículo Endoplasmático , Proteínas de Membrana , Humanos , Sítios de Ligação , Encéfalo , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/ultraestrutura , Microscopia Crioeletrônica , Retículo Endoplasmático/química , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Gabapentina/farmacologia , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas de Membrana/ultraestrutura , Miocárdio/química
4.
J Proteome Res ; 23(7): 2315-2322, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38913967

RESUMO

Native top-down mass spectrometry (nTDMS) allows characterization of protein structure and noncovalent interactions with simultaneous sequence mapping and proteoform characterization. The majority of nTDMS studies utilize purified recombinant proteins, with significant challenges hindering application to endogenous systems. To perform native top-down proteomics (nTDP), where endogenous proteins from complex biological systems are analyzed by nTDMS, it is essential to separate proteins under nondenaturing conditions. However, it remains difficult to achieve high resolution with MS-compatible online chromatography while preserving protein tertiary structure and noncovalent interactions. Herein, we report the use of online mixed-bed ion exchange chromatography (IEC) to enable separation of endogenous proteins from complex mixtures under nondenaturing conditions, preserving noncovalent interactions for nTDP analysis. We have successfully detected large proteins (>146 kDa) and identified endogenous metal-binding and oligomeric protein complexes in human heart tissue lysate. The use of a mixed-bed stationary phase allowed retention and elution of proteins over a wide range of isoelectric points without altering the sample or mobile phase pH. Overall, our method provides a simple online IEC-MS platform that can effectively separate proteins from complex mixtures under nondenaturing conditions and preserve higher-order structure for nTDP applications.


Assuntos
Proteômica , Cromatografia por Troca Iônica/métodos , Humanos , Proteômica/métodos , Miocárdio/química , Espectrometria de Massas/métodos , Misturas Complexas/química , Proteínas/química , Proteínas/análise , Proteínas/isolamento & purificação
5.
J Proteome Res ; 23(6): 1948-1959, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38717300

RESUMO

The availability of an increasingly large amount of public proteomics data sets presents an opportunity for performing combined analyses to generate comprehensive organism-wide protein expression maps across different organisms and biological conditions. Sus scrofa, a domestic pig, is a model organism relevant for food production and for human biomedical research. Here, we reanalyzed 14 public proteomics data sets from the PRIDE database coming from pig tissues to assess baseline (without any biological perturbation) protein abundance in 14 organs, encompassing a total of 20 healthy tissues from 128 samples. The analysis involved the quantification of protein abundance in 599 mass spectrometry runs. We compared protein expression patterns among different pig organs and examined the distribution of proteins across these organs. Then, we studied how protein abundances were compared across different data sets and studied the tissue specificity of the detected proteins. Of particular interest, we conducted a comparative analysis of protein expression between pig and human tissues, revealing a high degree of correlation in protein expression among orthologs, particularly in brain, kidney, heart, and liver samples. We have integrated the protein expression results into the Expression Atlas resource for easy access and visualization of the protein expression data individually or alongside gene expression data.


Assuntos
Rim , Proteômica , Animais , Proteômica/métodos , Humanos , Suínos , Rim/metabolismo , Rim/química , Especificidade de Órgãos , Fígado/metabolismo , Fígado/química , Bases de Dados de Proteínas , Encéfalo/metabolismo , Miocárdio/metabolismo , Miocárdio/química , Sus scrofa/metabolismo , Sus scrofa/genética , Proteoma/metabolismo , Proteoma/análise , Espectrometria de Massas
6.
J Proteome Res ; 23(8): 3161-3173, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-38456420

RESUMO

A computational analysis of mass spectrometry data was performed to uncover alternative splicing derived protein variants across chambers of the human heart. Evidence for 216 non-canonical isoforms was apparent in the atrium and the ventricle, including 52 isoforms not documented on SwissProt and recovered using an RNA sequencing derived database. Among non-canonical isoforms, 29 show signs of regulation based on statistically significant preferences in tissue usage, including a ventricular enriched protein isoform of tensin-1 (TNS1) and an atrium-enriched PDZ and LIM Domain 3 (PDLIM3) isoform 2 (PDLIM3-2/ALP-H). Examined variant regions that differ between alternative and canonical isoforms are highly enriched with intrinsically disordered regions. Moreover, over two-thirds of such regions are predicted to function in protein binding and RNA binding. The analysis here lends further credence to the notion that alternative splicing diversifies the proteome by rewiring intrinsically disordered regions, which are increasingly recognized to play important roles in the generation of biological function from protein sequences.


Assuntos
Processamento Alternativo , Proteínas Intrinsicamente Desordenadas , Isoformas de Proteínas , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Intrinsicamente Desordenadas/genética , Proteínas Intrinsicamente Desordenadas/metabolismo , Proteínas Intrinsicamente Desordenadas/química , Ventrículos do Coração/metabolismo , Proteoma/genética , Proteoma/metabolismo , Átrios do Coração/metabolismo , Miocárdio/metabolismo , Miocárdio/química , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Proteínas com Domínio LIM/química , Espectrometria de Massas , Tensinas/metabolismo , Tensinas/genética , Especificidade de Órgãos , Ligação Proteica
7.
Anal Chem ; 96(23): 9460-9467, 2024 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-38820243

RESUMO

Pathological cardiac hypertrophy is a complex process that often leads to heart failure. Label-free proteomics has emerged as an important platform to reveal protein variations and to elucidate the mechanisms of cardiac hypertrophy. Endomyocardial biopsy is a minimally invasive technique for sampling cardiac tissue, but it yields only limited amounts of an ethically permissible specimen. After regular pathological examination, the remaining trace samples pose significant challenges for effective protein extraction and mass spectrometry analysis. Herein, we developed trace cardiac tissue proteomics based on the anchor-nanoparticles (TCPA) method. We identified an average of 6666 protein groups using ∼50 µg of myocardial interventricular septum samples by TCPA. We then applied TCPA to acquire proteomics from patients' cardiac samples both diagnosed as hypertrophic hearts and myocarditis controls and identified significant alterations in pathways such as regulation of actin cytoskeleton, oxidative phosphorylation, and cGMP-PKG signaling pathway. Moreover, we found multiple lipid metabolic pathways to be dysregulated in transthyretin cardiac amyloidosis compared to other types of cardiac hypertrophy. TCPA offers a new technique for studying pathological cardiac hypertrophy and can serve as a platform toolbox for proteomic research in other cardiac diseases.


Assuntos
Miocárdio , Nanopartículas , Proteômica , Proteômica/métodos , Humanos , Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/química , Nanopartículas/química , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomegalia/diagnóstico , Amiloidose/metabolismo , Amiloidose/patologia , Neuropatias Amiloides Familiares
8.
PLoS Biol ; 19(4): e3001144, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33872299

RESUMO

Delineating human cardiac pathologies and their basic molecular mechanisms relies on research conducted in model organisms. Yet translating findings from preclinical models to humans present a significant challenge, in part due to differences in cardiac protein expression between humans and model organisms. Proteins immediately determine cellular function, yet their large-scale investigation in hearts has lagged behind those of genes and transcripts. Here, we set out to bridge this knowledge gap: By analyzing protein profiles in humans and commonly used model organisms across cardiac chambers, we determine their commonalities and regional differences. We analyzed cardiac tissue from each chamber of human, pig, horse, rat, mouse, and zebrafish in biological replicates. Using mass spectrometry-based proteomics workflows, we measured and evaluated the abundance of approximately 7,000 proteins in each species. The resulting knowledgebase of cardiac protein signatures is accessible through an online database: atlas.cardiacproteomics.com. Our combined analysis allows for quantitative evaluation of protein abundances across cardiac chambers, as well as comparisons of cardiac protein profiles across model organisms. Up to a quarter of proteins with differential abundances between atria and ventricles showed opposite chamber-specific enrichment between species; these included numerous proteins implicated in cardiac disease. The generated proteomics resource facilitates translational prospects of cardiac studies from model organisms to humans by comparisons of disease-linked protein networks across species.


Assuntos
Miocárdio/metabolismo , Proteoma/metabolismo , Animais , Coração/fisiologia , Ventrículos do Coração/química , Ventrículos do Coração/metabolismo , Cavalos , Humanos , Camundongos , Modelos Animais , Miocárdio/química , Especificidade de Órgãos , Processamento de Proteína Pós-Traducional , Proteoma/análise , Proteômica/métodos , Ratos , Especificidade da Espécie , Suínos , Peixe-Zebra
9.
Analyst ; 149(11): 3152-3160, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38630503

RESUMO

Cholesterol plays an important biological role in the body, and its disruption in homeostasis and synthesis has been implicated in several diseases. Mapping the locations of cholesterol is crucial for gaining a better understanding of these conditions. Silver deposition has proven to be an effective method for analyzing cholesterol using mass spectrometry imaging (MSI). We optimized and evaluated thermal evaporation as an alternative deposition technique to sputtering for silver deposition in MSI of cholesterol. A silver layer with a thickness of 6 nm provided an optimal combination of cholesterol signal intensity and mass resolution. The deposition of an ultrathin nanofilm of silver enabled high-resolution MSI with a pixel size of 10 µm. We used this optimized method to visualize the distribution of cholesterol in the senile plaques in the brains of APP/PS1 mice, a model that resembles Alzheimer's disease pathology. We found that cholesterol was evenly distributed across the frontal cortex tissue, with no evidence of plaque-like accumulation. Additionally, we investigated the presence and distribution of cholesterol in myocardial sections of a human heart affected by wild-type ATTR amyloidosis. We identified the presence of cholesterol in areas with amyloid deposition, but complete colocalization was not observed.


Assuntos
Colesterol , Prata , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Animais , Colesterol/análise , Colesterol/química , Prata/química , Humanos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Camundongos , Camundongos Transgênicos , Placa Amiloide , Encéfalo/metabolismo , Encéfalo/diagnóstico por imagem , Miocárdio/metabolismo , Miocárdio/química , Miocárdio/patologia , Amiloidose/metabolismo , Amiloidose/patologia , Volatilização , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Temperatura
10.
Anal Bioanal Chem ; 416(19): 4397-4407, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38861160

RESUMO

Plasmalogens are a special class of glycerophospholipids characterized by a vinyl ether bond (-C = C-O-) at the sn-1 position of the glycerol backbone. Altered plasmalogen profiles have been observed in neurodegenerative diseases and cancers. Profiling of plasmalogens requires specifying the vinyl ether bond and differentiating them from various types of isobars and isomers. Herein, by coupling C = C derivatization via offline Paternò-Büchi reaction with liquid chromatography-tandem mass spectrometry, we have developed a sensitive workflow for analysis of plasmalogens from biological samples. Using bovine heart lipid extract as a model system, we profiled more than 100 distinct structures of plasmenylethanolamines (PE-Ps) and plasmenylcholines (PC-Ps) at the C = C location level, far exceeding previous reports. Analysis of human glioma and normal brain tissue samples revealed elevated n-10 C = C isomers of PE-Ps in the glioma tissue samples. These findings suggest that the developed workflow holds potential in aiding the study of altered metabolism of plasmalogens in clinical samples.


Assuntos
Plasmalogênios , Espectrometria de Massas em Tandem , Plasmalogênios/análise , Plasmalogênios/química , Espectrometria de Massas em Tandem/métodos , Animais , Bovinos , Humanos , Glioma/metabolismo , Glioma/química , Cromatografia Líquida/métodos , Miocárdio/química , Miocárdio/metabolismo
11.
BMC Pregnancy Childbirth ; 24(1): 141, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38365664

RESUMO

OBJECTIVE: Whether or not the effects of anemia in the early phase, while the fetuses attempts to increase cardiac output to meet oxygen requirement in peripheral organs, is detrimental to the fetal developing vital organs is little-known. The objective of this is to compare prenatal cardiovascular changes and post-abortal cellular damages in the myocardium as a pumping organ and the brain as a perfused organ between anemic fetuses (using fetal Hb Bart's disease as a study model) in pre-hydropic phase and non-anemic fetuses. METHODS: Fetuses affected by Hb Bart's disease and non-anemic fetuses at 16-22 weeks were recruited to undergo comprehensive fetal echocardiography. Cord blood analysis was used to confirm the definite diagnosis of fetal Hb Bart's disease and normal fetuses. Fetal cardiac and brain tissues were collected shortly after pregnancy termination for the determination of oxidative stress and mitochondrial function, including mitochondrial ROS production and mitochondrial membrane changes. RESULTS: A total of 18 fetuses affected by Hb Bart's disease and 13 non-anemic fetuses were recruited. The clinical characteristics of both groups were comparable. The affected fetuses showed a significant increase in cardiac dimensions, cardiac function, cardiac output and brain circulation without deteriorating cardiac contractility and preload. However, in the affected fetuses, mitochondrial dysfunction was clearly demonstrated in brain tissues and in the myocardium, as indicated by a significant increase in the membrane potential change (p-value < 0.001), and a significant increase in ROS production in brain tissues, with a trend to increase in myocardium. The findings indicated cellular damage in spite of good clinical compensation. CONCLUSION: The new insight is that, in response to fetal anemia, fetal heart increases in size (dilatation) and function to increase cardiac output and blood flow velocity to provide adequate tissue perfusion, especially brain circulation. However, the myocardium and brain showed a significant increase in mitochondrial dysfunction, suggesting cellular damage secondary to anemic hypoxia. The compensatory increase in circulation could not completely prevent subtle brain and heart damage.


Assuntos
Anemia , Doenças Fetais , Hemoglobinas Anormais , Doenças Mitocondriais , Talassemia alfa , Feminino , Gravidez , Humanos , Segundo Trimestre da Gravidez , Espécies Reativas de Oxigênio , Hemoglobinas Anormais/análise , Doenças Fetais/diagnóstico , Coração Fetal/diagnóstico por imagem , Miocárdio/química , Edema , Débito Cardíaco
12.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33753506

RESUMO

Every heartbeat relies on cyclical interactions between myosin thick and actin thin filaments orchestrated by rising and falling Ca2+ levels. Thin filaments are comprised of two actin strands, each harboring equally separated troponin complexes, which bind Ca2+ to move tropomyosin cables away from the myosin binding sites and, thus, activate systolic contraction. Recently, structures of thin filaments obtained at low (pCa ∼9) or high (pCa ∼3) Ca2+ levels revealed the transition between the Ca2+-free and Ca2+-bound states. However, in working cardiac muscle, Ca2+ levels fluctuate at intermediate values between pCa ∼6 and pCa ∼7. The structure of the thin filament at physiological Ca2+ levels is unknown. We used cryoelectron microscopy and statistical analysis to reveal the structure of the cardiac thin filament at systolic pCa = 5.8. We show that the two strands of the thin filament consist of a mixture of regulatory units, which are composed of Ca2+-free, Ca2+-bound, or mixed (e.g., Ca2+ free on one side and Ca2+ bound on the other side) troponin complexes. We traced troponin complex conformations along and across individual thin filaments to directly determine the structural composition of the cardiac native thin filament at systolic Ca2+ levels. We demonstrate that the two thin filament strands are activated stochastically with short-range cooperativity evident only on one of the two strands. Our findings suggest a mechanism by which cardiac muscle is regulated by narrow range Ca2+ fluctuations.


Assuntos
Citoesqueleto de Actina/química , Actinas/química , Cálcio/metabolismo , Miocárdio/química , Miosinas/química , Sístole , Troponina/química , Animais , Cálcio/análise , Microscopia Crioeletrônica , Conformação Proteica , Suínos
13.
Nucleic Acids Res ; 49(1): 1-14, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33275144

RESUMO

Nucleic acid therapeutics (NATs) have proven useful in promoting the degradation of specific transcripts, modifying gene expression, and regulating mRNA splicing. In each situation, efficient delivery of nucleic acids to cells, tissues and intracellular compartments is crucial-both for optimizing efficacy and reducing side effects. Despite successes in NATs, our understanding of their cellular uptake and distribution in tissues is limited. Current methods have yielded insights into distribution of NATs within cells and tissues, but the sensitivity and resolution of these approaches are limited. Here, we show that nanoscale secondary ion mass spectrometry (NanoSIMS) imaging can be used to define the distribution of 5-bromo-2'-deoxythymidine (5-BrdT) modified antisense oligonucleotides (ASO) in cells and tissues with high sensitivity and spatial resolution. This approach makes it possible to define ASO uptake and distribution in different subcellular compartments and to quantify the impact of targeting ligands designed to promote ASO uptake by cells. Our studies showed that phosphorothioate ASOs are associated with filopodia and the inner nuclear membrane in cultured cells, and also revealed substantial cellular and subcellular heterogeneity of ASO uptake in mouse tissues. NanoSIMS imaging represents a significant advance in visualizing uptake and distribution of NATs; this approach will be useful in optimizing efficacy and delivery of NATs for treating human disease.


Assuntos
Oligonucleotídeos Antissenso/análise , Oligonucleotídeos Fosforotioatos/análise , Espectrometria de Massa de Íon Secundário/métodos , Células 3T3-L1 , Acetilgalactosamina/administração & dosagem , Acetilgalactosamina/análise , Animais , Receptor de Asialoglicoproteína/análise , Césio , Células HEK293 , Células HeLa , Humanos , Rim/química , Rim/ultraestrutura , Fígado/química , Fígado/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Miocárdio/química , Miocárdio/ultraestrutura , Oligonucleotídeos Antissenso/farmacocinética , Oligonucleotídeos Fosforotioatos/farmacocinética , Pseudópodes/química , Pseudópodes/ultraestrutura , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/biossíntese , RNA Longo não Codificante/genética , Frações Subcelulares/química , Enxofre/análise , Isótopos de Enxofre/análise , Distribuição Tecidual
14.
Proc Natl Acad Sci U S A ; 117(39): 24545-24556, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32929035

RESUMO

The relationship between oxidative stress and cardiac stiffness is thought to involve modifications to the giant muscle protein titin, which in turn can determine the progression of heart disease. In vitro studies have shown that S-glutathionylation and disulfide bonding of titin fragments could alter the elastic properties of titin; however, whether and where titin becomes oxidized in vivo is less certain. Here we demonstrate, using multiple models of oxidative stress in conjunction with mechanical loading, that immunoglobulin domains preferentially from the distal titin spring region become oxidized in vivo through the mechanism of unfolded domain oxidation (UnDOx). Via oxidation type-specific modification of titin, UnDOx modulates human cardiomyocyte passive force bidirectionally. UnDOx also enhances titin phosphorylation and, importantly, promotes nonconstitutive folding and aggregation of unfolded domains. We propose a mechanism whereby UnDOx enables the controlled homotypic interactions within the distal titin spring to stabilize this segment and regulate myocardial passive stiffness.


Assuntos
Miocárdio/química , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Proteínas Quinases/metabolismo , Animais , Elasticidade , Masculino , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Miócitos Cardíacos/química , Oxirredução , Fosforilação , Proteínas Quinases/química , Proteínas Quinases/genética
15.
J Mol Cell Cardiol ; 166: 137-151, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35219725

RESUMO

Ischemic and non-ischemic cardiomyopathies have distinct etiologies and underlying disease mechanisms, which require in-depth investigation for improved therapeutic interventions. The goal of this study was to use clinically obtained myocardium from healthy and heart failure patients, and characterize the changes in extracellular matrix (ECM) in ischemic and non-ischemic failing hearts, with and without mechanical unloading. Using tissue engineering methodologies, we also investigated how diseased human ECM, in the absence of systemic factors, can influence cardiomyocyte function. Heart tissues from heart failure patients with ischemic and non-ischemic cardiomyopathy were compared to explore differential disease phenotypes and reverse remodeling potential of left ventricular assisted device (LVAD) support at transcriptomic, proteomic and structural levels. The collected data demonstrated that the differential ECM compositions recapitulated the disease microenvironment and induced cardiomyocytes to undergo disease-like functional alterations. In addition, our study also revealed molecular profiles of non-ischemic and ischemic heart failure patients and explored the underlying mechanisms of etiology-specific impact on clinical outcome of LVAD support and tendency towards reverse remodeling.


Assuntos
Insuficiência Cardíaca , Coração Auxiliar , Matriz Extracelular , Coração Auxiliar/efeitos adversos , Humanos , Miocárdio/química , Proteômica
16.
Blood ; 136(21): 2469-2472, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-32604409

RESUMO

Recent reports indicate that suspended skeletal and cardiac myosin, such as might be released during injury, can act as procoagulants by providing membrane-like support for factors Xa and Va in the prothrombinase complex. Further, skeletal myosin provides membrane-like support for activated protein C. This raises the question of whether purified muscle myosins retain procoagulant phospholipid through purification. We found that lactadherin, a phosphatidyl-l-serine-binding protein, blocked >99% of prothrombinase activity supported by rabbit skeletal and by bovine cardiac myosin. Similarly, annexin A5 and phospholipase A2 blocked >95% of myosin-supported activity, confirming that contaminating phospholipid is required to support myosin-related prothrombinase activity. We asked whether contaminating phospholipid in myosin preparations may also contain tissue factor (TF). Skeletal myosin supported factor VIIa cleavage of factor X equivalent to contamination by ∼1:100 000 TF/myosin, whereas cardiac myosin had TF-like activity >10-fold higher. TF pathway inhibitor inhibited the TF-like activity similar to control TF. These results indicate that purified skeletal muscle and cardiac myosins support the prothrombinase complex indirectly through contaminating phospholipid and also support factor X activation through TF-like activity. Our findings suggest a previously unstudied affinity of skeletal and cardiac myosin for phospholipid membranes.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fator V/efeitos dos fármacos , Fator Xa/efeitos dos fármacos , Músculo Esquelético/química , Miocárdio/química , Miosinas/farmacologia , Fosfolipídeos/farmacologia , Animais , Antígenos de Superfície/farmacologia , Miosinas Cardíacas/isolamento & purificação , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/farmacologia , Bovinos , Contaminação de Medicamentos , Fator VIIa/metabolismo , Fator Xa/metabolismo , Humanos , Lipoproteínas/farmacologia , Proteínas do Leite/farmacologia , Miosinas/isolamento & purificação , Miosinas/metabolismo , Fosfolipases A2/farmacologia , Coelhos , Tromboplastina/farmacologia
17.
Proc Natl Acad Sci U S A ; 116(22): 10636-10645, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31068470

RESUMO

The rationale for genome-wide association study (GWAS) results is sequence variation in cis-regulatory elements (CREs) modulating a target gene's expression as the major cause of trait variation. To understand the complete molecular landscape of one of these GWAS loci, we performed in vitro reporter screens in cardiomyocyte cell lines for CREs overlapping nearly all common variants associated with any of five independent QT interval (QTi)-associated GWAS hits at the SCN5A-SCN10A locus. We identified 13 causal CRE variants using allelic reporter activity, cardiomyocyte nuclear extract-based binding assays, overlap with human cardiac tissue DNaseI hypersensitive regions, and predicted impact of sequence variants on DNaseI sensitivity. Our analyses identified at least one high-confidence causal CRE variant for each of the five sentinel hits that could collectively predict SCN5A cardiac gene expression and QTi association. Although all 13 variants could explain SCN5A gene expression, the highest statistical significance was obtained with seven variants (inclusive of the five above). Thus, multiple, causal, mutually associated CRE variants can underlie GWAS signals.


Assuntos
Regulação da Expressão Gênica/genética , Coração/fisiopatologia , Miocárdio/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Alelos , Animais , Linhagem Celular , Eletrocardiografia , Variação Genética/genética , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Miocárdio/química , Locos de Características Quantitativas/genética , Sequências Reguladoras de Ácido Nucleico/genética
18.
Proc Natl Acad Sci U S A ; 116(14): 6828-6835, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30877248

RESUMO

Cardiac muscle contraction is triggered by calcium binding to troponin. The consequent movement of tropomyosin permits myosin binding to actin, generating force. Cardiac myosin-binding protein C (cMyBP-C) plays a modulatory role in this activation process. One potential mechanism for the N-terminal domains of cMyBP-C to achieve this is by binding directly to the actin-thin filament at low calcium levels to enhance the movement of tropomyosin. To determine the molecular mechanisms by which cMyBP-C enhances myosin recruitment to the actin-thin filament, we directly visualized fluorescently labeled cMyBP-C N-terminal fragments and GFP-labeled myosin molecules binding to suspended actin-thin filaments in a fluorescence-based single-molecule microscopy assay. Binding of the C0C3 N-terminal cMyBP-C fragment to the thin filament enhanced myosin association at low calcium levels. However, at high calcium levels, C0C3 bound in clusters, blocking myosin binding. Dynamic imaging of thin filament-bound Cy3-C0C3 molecules demonstrated that these fragments diffuse along the thin filament before statically binding, suggesting a mechanism that involves a weak-binding mode to search for access to the thin filament and a tight-binding mode to sensitize the thin filament to calcium, thus enhancing myosin binding. Although shorter N-terminal fragments (Cy3-C0C1 and Cy3-C0C1f) bound to the thin filaments and displayed modes of motion on the thin filament similar to that of the Cy3-C0C3 fragment, the shorter fragments were unable to sensitize the thin filament. Therefore, the longer N-terminal fragment (C0C3) must possess the requisite domains needed to bind specifically to the thin filament in order for the cMyBP-C N terminus to modulate cardiac contractility.


Assuntos
Proteínas de Transporte/química , Simulação de Dinâmica Molecular , Miosinas/química , Tropomiosina/química , Animais , Proteínas de Transporte/metabolismo , Galinhas , Humanos , Contração Miocárdica , Miocárdio/química , Miocárdio/metabolismo , Miosinas/metabolismo , Ligação Proteica , Domínios Proteicos , Tropomiosina/metabolismo
19.
Proc Natl Acad Sci U S A ; 116(33): 16384-16393, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31358631

RESUMO

High-speed atomic force microscopy (HS-AFM) can be used to study dynamic processes with real-time imaging of molecules within 1- to 5-nm spatial resolution. In the current study, we evaluated the 3-state model of activation of cardiac thin filaments (cTFs) isolated as a complex and deposited on a mica-supported lipid bilayer. We studied this complex for dynamic conformational changes 1) at low and high [Ca2+] (pCa 9.0 and 4.5), and 2) upon myosin binding to the cTF in the nucleotide-free state or in the presence of ATP. HS-AFM was used to directly visualize the tropomyosin-troponin complex and Ca2+-induced tropomyosin movements accompanied by structural transitions of actin monomers within cTFs. Our data show that cTFs at relaxing or activating conditions are not ultimately in a blocked or activated state, respectively, but rather the combination of states with a prevalence that is dependent on the [Ca2+] and the presence of weakly or strongly bound myosin. The weakly and strongly bound myosin induce similar changes in the structure of cTFs as confirmed by the local dynamical displacement of individual tropomyosin strands in the center of a regulatory unit of cTF at the relaxed and activation conditions. The displacement of tropomyosin at the relaxed conditions had never been visualized directly and explains the ability of myosin binding to TF at the relaxed conditions. Based on the ratios of nonactivated and activated segments within cTFs, we proposed a mechanism of tropomyosin switching from different states that includes both weakly and strongly bound myosin.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Actinas/ultraestrutura , Subfragmentos de Miosina/ultraestrutura , Tropomiosina/ultraestrutura , Troponina/ultraestrutura , Citoesqueleto de Actina/química , Actinas/química , Animais , Cálcio/metabolismo , Bicamadas Lipídicas/química , Modelos Moleculares , Imagem Molecular , Contração Muscular/genética , Músculo Esquelético/química , Músculo Esquelético/ultraestrutura , Miocárdio/química , Miocárdio/ultraestrutura , Subfragmentos de Miosina/química , Miosinas/química , Ligação Proteica , Coelhos , Sarcômeros/química , Sarcômeros/ultraestrutura , Tropomiosina/química , Troponina/química
20.
J Am Soc Nephrol ; 32(11): 2912-2919, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34518280

RESUMO

BACKGROUND: Cerebral and myocardial hypoperfusion occur during hemodialysis in adults. Pediatric patients receiving chronic hemodialysis have fewer cardiovascular risk factors, yet cardiovascular morbidity remains prominent. METHODS: We conducted a prospective observational study of pediatric patients receiving chronic hemodialysis to investigate whether intermittent hemodialysis is associated with adverse end organ effects in the heart or with cerebral oxygenation (regional tissue oxyhemoglobin saturation [rSO2]). We assessed intradialytic cardiovascular function and rSO2 using noninvasive echocardiography to determine myocardial strain and continuous noninvasive near-infrared spectroscopy for rSO2. We measured changes in blood volume and measured central venous oxygen saturation (mCVO2) pre-, mid-, and post-hemodialysis. RESULTS: The study included 15 patients (median age, 12 years; median hemodialysis vintage, 13.2 [9-24] months). Patients were asymptomatic. The rSO2 did not change during hemodialysis, whereas mCVO2 decreased significantly, from 73% to 64.8%. Global longitudinal strain of the myocardium worsened significantly by mid-hemodialysis and persisted post-hemodialysis. The ejection fraction remained normal. Lower systolic BP and faster blood volume change were associated with worsening myocardial strain; only blood volume change was significant in multivariate analysis (ß-coefficient, -0.3; 95% confidence interval [CI], -0.38 to -0.21; P<0.001). Blood volume change was also associated with a significant decrease in mCVO2 (ß-coefficient, 0.42; 95% CI, 0.07 to 0.76; P=0.001). Access, age, hemodialysis vintage, and ultrafiltration volume were not associated with worsening strain. CONCLUSIONS: Unchanged rSO2 suggested that cerebral oxygenation was maintained during hemodialysis. However, despite maintained ejection fraction, intradialytic myocardial strain worsened in pediatric hemodialysis and was associated with blood volume change. The effect of hemodialysis on individual organ perfusion in pediatric versus adult patients receiving hemodialysis might differ.


Assuntos
Química Encefálica , Coração/fisiopatologia , Falência Renal Crônica/fisiopatologia , Oxigênio/sangue , Diálise Renal/efeitos adversos , Adolescente , Fatores Etários , Volume Sanguíneo , Circulação Cerebrovascular , Criança , Ecocardiografia , Feminino , Fatores de Risco de Doenças Cardíacas , Hemodinâmica , Humanos , Hipóxia Encefálica/etiologia , Falência Renal Crônica/sangue , Falência Renal Crônica/terapia , Masculino , Miocárdio/química , Oximetria , Volume Sistólico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA