Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Pathol ; 192(1): 160-177, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34710383

RESUMO

Sigma 1 receptor (Sigmar1) is a widely expressed, multitasking molecular chaperone protein that plays functional roles in several cellular processes. Mutations in the Sigmar1 gene are associated with several distal neuropathies with strong manifestation in skeletal muscle dysfunction with phenotypes like muscle wasting and atrophy. However, the physiological function of Sigmar1 in skeletal muscle remains unknown. Herein, the physiological role of Sigmar1 in skeletal muscle structure and function in gastrocnemius, quadriceps, soleus, extensor digitorum longus, and tibialis anterior muscles was determined. Quantification of myofiber cross-sectional area showed altered myofiber size distribution and changes in myofiber type in the skeletal muscle of the Sigmar1-/- mice. Interestingly, ultrastructural analysis by transmission electron microscopy showed the presence of abnormal mitochondria, and immunostaining showed derangements in dystrophin localization in skeletal muscles from Sigmar1-/- mice. In addition, myopathy in Sigmar1-/- mice was associated with an increased number of central nuclei, increased collagen deposition, and fibrosis. Functional studies also showed reduced endurance and exercise capacity in the Sigmar1-/- mice without any changes in voluntary locomotion, markers for muscle denervation, and muscle atrophy. Overall, this study shows, for the first time, a potential physiological function of Sigmar1 in maintaining healthy skeletal muscle structure and function.


Assuntos
Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Receptores sigma/deficiência , Animais , Colágeno/metabolismo , Distrofina/metabolismo , Fibrose , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/ultraestrutura , Condicionamento Físico Animal , Transporte Proteico , Receptores sigma/metabolismo , Receptor Sigma-1
2.
Mol Pharm ; 17(12): 4676-4690, 2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33151075

RESUMO

The molecular architecture of pH-responsive amphiphilic block copolymers, their self-assembly behavior to form nanoparticles (NPs), and doxorubicin (DOX)-loading technique govern the extent of DOX-induced cardiotoxicity. We observed that the choice of pH-sensitive tertiary amines, surface charge, and DOX-loading techniques within the self-assembled NPs strongly influence the release and stimulation of DOX-induced cardiotoxicity in primary cardiomyocytes. However, covalent conjugation of DOX to a pH-sensitive nanocarrier through a "conditionally unstable amide" linkage (PCPY-cDOX; PC = polycarbonate and PY = 2-pyrrolidine-1-yl-ethyl-amine) significantly reduced the cardiotoxicity of DOX in cardiomyocytes as compared to noncovalently encapsulated DOX NPs (PCPY-eDOX). When these formulations were tested for drug release in serum-containing media, the PCPY-cDOX systems showed prolonged control over drug release (for ∼72 h) at acidic pH compared to DOX-encapsulated nanocarriers, as expected. We found that DOX-encapsulated nanoformulations triggered cardiotoxicity in primary cardiomyocytes more acutely, while conjugated systems such as PCPY-cDOX prevented cardiotoxicity by disabling the nuclear entry of the drug. Using 2D and 3D (spheroid) cultures of an ER + breast cancer cell line (MCF-7) and a triple-negative breast cancer cell line (MDA-MB-231), we unravel that, similar to encapsulated systems (PCPY-eDOX-type) as reported earlier, the PCPY-cDOX system suppresses cellular proliferation in both cell lines and enhances trafficking through 3D spheroids of MDA-MB-231 cells. Collectively, our studies indicate that PCPY-cDOX is less cardiotoxic as compared to noncovalently encapsulated variants without compromising the chemotherapeutic properties of the drug. Thus, our studies suggest that the appropriate selection of the nanocarrier for DOX delivery may prove fruitful in shifting the balance between low cardiotoxicity and triggering the chemotherapeutic potency of DOX.


Assuntos
Cardiotoxicidade/prevenção & controle , Doxorrubicina/administração & dosagem , Portadores de Fármacos/química , Neoplasias/tratamento farmacológico , Polímeros/química , Animais , Animais Recém-Nascidos , Cardiotoxicidade/etiologia , Linhagem Celular Tumoral , Doxorrubicina/farmacocinética , Doxorrubicina/toxicidade , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Concentração de Íons de Hidrogênio , Miócitos Cardíacos , Nanopartículas/química , Neoplasias/patologia , Cimento de Policarboxilato , Cultura Primária de Células , Pirrolidinas/química , Ratos , Esferoides Celulares , Testes de Toxicidade Aguda
3.
Reprod Health ; 13(1): 129, 2016 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-27716260

RESUMO

BACKGROUND: Prior studies have shown that using uterotonics to augment or induce labor before arrival at comprehensive Emergency Obstetric and Neonatal Care (CEmONC) settings (henceforth, "outside uterotonics") may contribute to perinatal mortality in low- and middle-income countries. We estimate its effect on perinatal mortality in rural Bangladesh. METHODS: Using hospital records (23986 singleton term births, Jan 1, 2009-Dec 31, 2015) from rural Bangladesh, we use a logistic regression model to estimate the increased risk of perinatal death from uterotonics administered outside a CEmONC facility. RESULTS: Among term births (≥37 weeks gestation), the risk of perinatal death adjusted for key confounders is significantly increased among women reporting uterotonic use outside of CEmONC (OR = 3 · 0, 95 % CI = 2 · 4,3 · 7). This increased risk is particularly high for fresh stillbirths (OR = 4 · 0, 95 % CI = 3 · 0,5 · 3) and intrapartum-related causes of early neonatal deaths (birth asphyxia) (OR = 3 · 1, 95 % CI = 2 · 2,4 · 5). CONCLUSIONS: In this sample, outside uterotonic use was associated with substantially increased risk of fresh stillbirths, deaths due to birth asphyxia, and all perinatal deaths. In settings of high uterotonic use outside of controlled settings, substantial improvement in both stillbirth and early neonatal mortality may be made by reducing such use.


Assuntos
Ocitócicos/efeitos adversos , Mortalidade Perinatal , Bangladesh/epidemiologia , Estudos Transversais , Países em Desenvolvimento , Uso de Medicamentos/estatística & dados numéricos , Emergências , Feminino , Humanos , Complicações do Trabalho de Parto/induzido quimicamente , Complicações do Trabalho de Parto/epidemiologia , Ocitócicos/administração & dosagem , Gravidez , Resultado da Gravidez/epidemiologia , Medição de Risco/métodos , Saúde da População Rural/estatística & dados numéricos , Natimorto/epidemiologia
4.
Environ Res ; 143(Pt A): 123-30, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26476787

RESUMO

BACKGROUND: Arsenic (As) methylation capacity in epidemiologic studies is typically indicated by the proportions of inorganic As (%InAs), monomethylarsonic acid (%MMA), and dimethylarsinic acid (%DMA) in urine as a fraction of total urinary As. The relationship between renal function and indicators of As methylation capacity has not been thoroughly investigated. OBJECTIVES: Our two aims were to examine (1) associations between estimated glomerular filtration rate (eGFR) and %As metabolites in blood and urine, and (2) whether renal function modifies the relationship of blood %As metabolites with respective urinary %As metabolites. METHODS: In a cross-sectional study of 375 As-exposed Bangladeshi adults, we measured blood and urinary As metabolites, and calculated eGFR from plasma cystatin C. RESULTS: In covariate-adjusted linear models, a 1 ml/min/1.73 m(2) increase in eGFR was associated with a 0.39% increase in urinary %InAs (p<0.0001) and a mean decrease in urinary %DMA of 0.07 (p=0.0005). In the 292 participants with measurable blood As metabolites, the associations of eGFR with increased blood %InAs and decreased blood %DMA did not reach statistical significance. eGFR was not associated with urinary or blood %MMA in covariate-adjusted models. For a given increase in blood %InAs, the increase in urinary %InAs was smaller in those with reduced eGFR, compared to those with normal eGFR (p=0.06); this effect modification was not observed for %MMA or %DMA. CONCLUSIONS: Urinary excretion of InAs may be impaired in individuals with reduced renal function. Alternatively, increased As methylation capacity (as indicated by decreased urinary %InAs) may be detrimental to renal function.


Assuntos
Arsênio/toxicidade , Arsenicais , Ácido Cacodílico , Água Potável/química , Taxa de Filtração Glomerular/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Adulto , Idoso , Arsênio/sangue , Arsênio/urina , Arsenicais/sangue , Arsenicais/urina , Bangladesh , Ácido Cacodílico/sangue , Ácido Cacodílico/urina , Estudos Transversais , Feminino , Humanos , Modelos Lineares , Masculino , Metilação , Pessoa de Meia-Idade , Poluentes Químicos da Água/sangue , Poluentes Químicos da Água/urina
5.
PLoS Genet ; 8(2): e1002522, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22383894

RESUMO

Arsenic contamination of drinking water is a major public health issue in many countries, increasing risk for a wide array of diseases, including cancer. There is inter-individual variation in arsenic metabolism efficiency and susceptibility to arsenic toxicity; however, the basis of this variation is not well understood. Here, we have performed the first genome-wide association study (GWAS) of arsenic-related metabolism and toxicity phenotypes to improve our understanding of the mechanisms by which arsenic affects health. Using data on urinary arsenic metabolite concentrations and approximately 300,000 genome-wide single nucleotide polymorphisms (SNPs) for 1,313 arsenic-exposed Bangladeshi individuals, we identified genome-wide significant association signals (P<5×10(-8)) for percentages of both monomethylarsonic acid (MMA) and dimethylarsinic acid (DMA) near the AS3MT gene (arsenite methyltransferase; 10q24.32), with five genetic variants showing independent associations. In a follow-up analysis of 1,085 individuals with arsenic-induced premalignant skin lesions (the classical sign of arsenic toxicity) and 1,794 controls, we show that one of these five variants (rs9527) is also associated with skin lesion risk (P = 0.0005). Using a subset of individuals with prospectively measured arsenic (n = 769), we show that rs9527 interacts with arsenic to influence incident skin lesion risk (P = 0.01). Expression quantitative trait locus (eQTL) analyses of genome-wide expression data from 950 individual's lymphocyte RNA suggest that several of our lead SNPs represent cis-eQTLs for AS3MT (P = 10(-12)) and neighboring gene C10orf32 (P = 10(-44)), which are involved in C10orf32-AS3MT read-through transcription. This is the largest and most comprehensive genomic investigation of arsenic metabolism and toxicity to date, the only GWAS of any arsenic-related trait, and the first study to implicate 10q24.32 variants in both arsenic metabolism and arsenical skin lesion risk. The observed patterns of associations suggest that MMA% and DMA% have distinct genetic determinants and support the hypothesis that DMA is the less toxic of these two methylated arsenic species. These results have potential translational implications for the prevention and treatment of arsenic-associated toxicities worldwide.


Assuntos
Arsênio/metabolismo , Cromossomos Humanos Par 10/genética , Estudo de Associação Genômica Ampla , Polimorfismo de Nucleotídeo Único , Intoxicação por Arsênico/genética , Arsenicais/metabolismo , Bangladesh , Exposição Ambiental , Predisposição Genética para Doença , Humanos , Fenótipo , Poluentes Químicos da Água/toxicidade
6.
J Nutr ; 144(5): 690-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24598884

RESUMO

Chronic exposure to inorganic arsenic (InAs) through drinking water is a major problem worldwide. InAs undergoes hepatic methylation to form mono- and dimethyl arsenical species (MMA and DMA, respectively), facilitating arsenic elimination. Both reactions are catalyzed by arsenic (+3 oxidation state) methyltransferase (AS3MT) using S-adenosylmethionine (SAM) as the methyl donor, yielding the methylated product and S-adenosylhomocysteine (SAH), a potent product-inhibitor of AS3MT. SAM biosynthesis depends on folate- and cobalamin-dependent one-carbon metabolism. With the use of samples from 353 participants in the Folate and Oxidative Stress Study, our objective was to test the hypotheses that blood SAM and SAH concentrations are associated with arsenic methylation and that these associations differ by folate and cobalamin nutritional status. Blood SAM and SAH were measured by HPLC. Arsenic metabolites in blood and urine were measured by HPLC coupled to dynamic reaction cell inductively coupled plasma MS. In linear regression analyses, SAH was not associated with any of the arsenic metabolites. However, log(SAM) was negatively associated with log(% urinary InAs) (ß: -0.11; 95% CI: -0.19, -0.02; P = 0.01), and folate and cobalamin nutritional status significantly modified associations between SAM and percentage of blood MMA (%bMMA) and percentage of blood DMA (%bDMA) (P = 0.02 and P = 0.01, respectively). In folate- and cobalamin-deficient individuals, log(SAM) was positively associated with %bMMA (ß: 6.96; 95% CI: 1.86, 12.05; P < 0.01) and negatively associated with %bDMA (ß: -6.19; 95% CI: -12.71, 0.32; P = 0.06). These findings suggest that when exposure to InAs is high, and methyl groups are limiting, SAM is used primarily for MMA synthesis rather than for DMA synthesis, contributing additional evidence that nutritional status may explain some of the interindividual differences in arsenic metabolism and, consequently, susceptibility to arsenic toxicity.


Assuntos
Arsênio/sangue , Arsenicais/sangue , Exposição Ambiental , Ácido Fólico/sangue , S-Adenosilmetionina/sangue , Vitamina B 12/sangue , Adulto , Idoso , Arsênio/toxicidade , Arsênio/urina , Intoxicação por Arsênico/sangue , Intoxicação por Arsênico/urina , Arsenicais/urina , Bangladesh , Estudos Transversais , Água Potável , Feminino , Homocisteína/sangue , Humanos , Masculino , Metilação , Pessoa de Meia-Idade , Estresse Oxidativo , S-Adenosil-Homocisteína/sangue , S-Adenosilmetionina/urina
7.
JAMIA Open ; 7(1): ooae015, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38414534

RESUMO

Objectives: In the United States, end-stage kidney disease (ESKD) is responsible for high mortality and significant healthcare costs, with the number of cases sharply increasing in the past 2 decades. In this study, we aimed to reduce these impacts by developing an ESKD model for predicting its occurrence in a 2-year period. Materials and Methods: We developed a machine learning (ML) pipeline to test different models for the prediction of ESKD. The electronic health record was used to capture several kidney disease-related variables. Various imputation methods, feature selection, and sampling approaches were tested. We compared the performance of multiple ML models using area under the ROC curve (AUCROC), area under the Precision-Recall curve (PR-AUC), and Brier scores for discrimination, precision, and calibration, respectively. Explainability methods were applied to the final model. Results: Our best model was a gradient-boosting machine with feature selection and imputation methods as additional components. The model exhibited an AUCROC of 0.97, a PR-AUC of 0.33, and a Brier score of 0.002 on a holdout test set. A chart review analysis by expert physicians indicated clinical utility. Discussion and Conclusion: An ESKD prediction model can identify individuals at risk for ESKD and has been successfully deployed within our health system.

8.
JCI Insight ; 9(1)2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-38015634

RESUMO

Pulmonary fibrosis is a chronic and often fatal disease. The pathogenesis is characterized by aberrant repair of lung parenchyma, resulting in loss of physiological homeostasis, respiratory failure, and death. The immune response in pulmonary fibrosis is dysregulated. The gut microbiome is a key regulator of immunity. The role of the gut microbiome in regulating the pulmonary immunity in lung fibrosis is poorly understood. Here, we determine the impact of gut microbiota on pulmonary fibrosis in substrains of C57BL/6 mice derived from different vendors (C57BL/6J and C57BL/6NCrl). We used germ-free models, fecal microbiota transplantation, and cohousing to transmit gut microbiota. Metagenomic studies of feces established keystone species between substrains. Pulmonary fibrosis was microbiota dependent in C57BL/6 mice. Gut microbiota were distinct by ß diversity and α diversity. Mortality and lung fibrosis were attenuated in C57BL/6NCrl mice. Elevated CD4+IL-10+ T cells and lower IL-6 occurred in C57BL/6NCrl mice. Horizontal transmission of microbiota by cohousing attenuated mortality in C57BL/6J mice and promoted a transcriptionally altered pulmonary immunity. Temporal changes in lung and gut microbiota demonstrated that gut microbiota contributed largely to immunological phenotype. Key regulatory gut microbiota contributed to lung fibrosis, generating rationale for human studies.


Assuntos
Microbioma Gastrointestinal , Microbiota , Fibrose Pulmonar , Camundongos , Animais , Humanos , Microbioma Gastrointestinal/fisiologia , Camundongos Endogâmicos C57BL , Pulmão , Microbiota/fisiologia
9.
Redox Biol ; 68: 102949, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37922764

RESUMO

Hydropersulfide and hydropolysulfide metabolites are increasingly important reactive sulfur species (RSS) regulating numerous cellular redox dependent functions. Intracellular production of these species is known to occur through RSS interactions or through translational mechanisms involving cysteinyl t-RNA synthetases. However, regulation of these species under cell stress conditions, such as hypoxia, that are known to modulate RSS remain poorly understood. Here we define an important mechanism of increased persulfide and polysulfide production involving cystathionine gamma lyase (CSE) phosphorylation at serine 346 and threonine 355 in a substrate specific manner, under acute hypoxic conditions. Hypoxic phosphorylation of CSE occurs in an AMP kinase dependent manner increasing enzyme activity involving unique inter- and intramolecular interactions within the tetramer. Importantly, both cellular hypoxia and tissue ischemia result in AMP Kinase dependent CSE phosphorylation that regulates blood flow in ischemic tissues. Our findings reveal hypoxia molecular signaling pathways regulating CSE dependent persulfide and polysulfide production impacting tissue and cellular response to stress.


Assuntos
Sulfeto de Hidrogênio , Humanos , Sulfeto de Hidrogênio/metabolismo , Fosforilação , Adenilato Quinase/metabolismo , Cistationina gama-Liase/genética , Hipóxia
10.
Mitochondrion ; 62: 159-175, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34902622

RESUMO

Sigmar1 is a widely expressed molecular chaperone protein in mammalian cell systems. Accumulating research demonstrated the cardioprotective roles of pharmacologic Sigmar1 activation by ligands in preclinical rodent models of cardiac injury. Extensive biochemical and immuno-electron microscopic research demonstrated Sigmar1's sub-cellular localization largely depends on cell and organ types. Despite comprehensive studies, Sigmar1's direct molecular role in cardiomyocytes remains elusive. In the present study, we determined Sigmar1's subcellular localization, transmembrane topology, and function using complementary microscopy, biochemical, and functional assays in cardiomyocytes. Quantum dots in transmission electron microscopy showed Sigmar1 labeled quantum dots on the mitochondrial membranes, lysosomes, and sarcoplasmic reticulum-mitochondrial interface. Subcellular fractionation of heart cell lysates confirmed Sigmar1's localization in purified mitochondria fraction and lysosome fraction. Immunocytochemistry confirmed Sigmar1 colocalization with mitochondrial proteins in isolated adult mouse cardiomyocytes. Sigmar1's mitochondrial localization was further confirmed by Sigmar1 colocalization with Mito-Tracker in isolated mouse heart mitochondria. A series of biochemical experiments, including alkaline extraction and proteinase K treatment of purified heart mitochondria, demonstrated Sigmar1 as an integral mitochondrial membrane protein. Sigmar1's structural requirement for mitochondrial localization was determined by expressing FLAG-tagged Sigmar1 fragments in cells. Full-length Sigmar1 and Sigmar1's C terminal-deletion fragments were able to localize to the mitochondrial membrane, whereas N-terminal deletion fragment was unable to incorporate into the mitochondria. Finally, functional assays using extracellular flux analyzer and high-resolution respirometry showed Sigmar1 siRNA knockdown significantly altered mitochondrial respiration in cardiomyocytes. Overall, we found that Sigmar1 localizes to mitochondrial membranes and is indispensable for maintaining mitochondrial respiratory homeostasis in cardiomyocytes.


Assuntos
Mitocôndrias Cardíacas/fisiologia , Miócitos Cardíacos/metabolismo , Transporte Proteico/fisiologia , Receptores sigma/metabolismo , Animais , Metabolismo Energético/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Masculino , Camundongos , RNA Interferente Pequeno , Ratos , Receptores sigma/genética , Receptor Sigma-1
11.
Redox Biol ; 57: 102480, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36167027

RESUMO

Methamphetamine (METH) is an addictive illicit drug used worldwide that causes significant damage to blood vessels resulting in cardiovascular dysfunction. Recent studies highlight increased prevalence of cardiovascular disease (CVD) and associated complications including hypertension, vasospasm, left ventricular hypertrophy, and coronary artery disease in younger populations due to METH use. Here we report that METH administration in a mouse model of 'binge and crash' decreases cardiovascular function via cystathionine gamma lyase (CSE), hydrogen sulfide (H2S), nitric oxide (NO) (CSE/H2S/NO) dependent pathway. METH significantly reduced H2S and NO bioavailability in plasma and skeletal muscle tissues co-incident with a significant reduction in flow-mediated vasodilation (FMD) and blood flow velocity revealing endothelial dysfunction. METH administration also reduced cardiac ejection fraction (EF) and fractional shortening (FS) associated with increased tissue and perivascular fibrosis. Importantly, METH treatment selectively decreased CSE expression and sulfide bioavailability along with reduced eNOS phosphorylation and NO levels. Exogenous sulfide therapy or endothelial CSE transgenic overexpression corrected cardiovascular and associated pathological responses due to METH implicating a central molecular regulatory pathway for tissue pathology. These findings reveal that therapeutic intervention targeting CSE/H2S bioavailability may be useful in attenuating METH mediated cardiovascular disease.

12.
Front Physiol ; 11: 1054, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32982788

RESUMO

Mitochondria are the key to properly functioning energy generation in the metabolically demanding cardiomyocytes and thus essential to healthy heart contractility on a beat-to-beat basis. Mitochondria being the central organelle for cellular metabolism and signaling in the heart, its dysfunction leads to cardiovascular disease. The healthy mitochondrial functioning critical to maintaining cardiomyocyte viability and contractility is accomplished by adaptive changes in the dynamics, biogenesis, and degradation of the mitochondria to ensure cellular proteostasis. Recent compelling evidence suggests that the classical protein quality control system in cardiomyocytes is also under constant mitochondrial control, either directly or indirectly. Impairment of cytosolic protein quality control may affect the position of the mitochondria in relation to other organelles, as well as mitochondrial morphology and function, and could also activate mitochondrial proteostasis. Despite a growing interest in the mitochondrial quality control system, very little information is available about the molecular function of mitochondria in cardiac proteostasis. In this review, we bring together current understanding of the adaptations and role of the mitochondria in cardiac proteostasis and describe the adaptive/maladaptive changes observed in the mitochondrial network required to maintain proteomic integrity. We also highlight the key mitochondrial signaling pathways activated in response to proteotoxic stress as a cellular mechanism to protect the heart from proteotoxicity. A deeper understanding of the molecular mechanisms of mitochondrial adaptations and their role in cardiac proteostasis will help to develop future therapeutics to protect the heart from cardiovascular diseases.

13.
Redox Biol ; 36: 101660, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32750667

RESUMO

Mitochondria are highly dynamic organelles that constantly undergo fission and fusion events to adapt to changes in the cellular environment. Aberrant mitochondrial fission has been associated with several types of cardiovascular dysfunction; inhibition of pathologically aberrant mitochondrial fission has been shown to be cardioprotective. Pathological fission is mediated by the excessive activation of GTPase dynamin-related protein 1 (Drp1), making it an attractive therapeutic target in numerous cardiovascular diseases. Mitochondrial division inhibitor (mdivi-1) is widely used small molecule reported to inhibit Drp1-dependent fission, elongate mitochondria, and mitigate injury. The purpose of our study was to understand the pleiotropic effects of mdivi-1 on mitochondrial dynamics, mitochondrial respiration, electron transport activities, and macro-autophagy. In this study, we found that mdivi-1 treatment decreased Drp1 expression, proteolytically cleaved L-OPA1, and altered the expression of OXPHOS complex proteins, resulting in increased superoxide production. The altered expression of OXPHOS complex proteins may be directly associated with decreased Drp1 expression, as Drp1 siRNA knockdown in cardiomyocytes showed similar effects. Results from an autophagy flux assay showed that mdivi-1 induced impaired autophagy flux that could be restored by Atg7 overexpression, suggesting that mdivi-1 mediated inhibition of macro-autophagy in cardiomyocytes. Treatment with mdivi-1 resulted in increased expression of p62, which is required for Atg7 overexpression-induced rescue of mdivi-1-mediated impaired autophagy flux. In addition, mdivi-1-dependent proteolytic processing of L-OPA1 was associated with increased mitochondrial superoxide production and altered expression of mitochondrial serine/proteases. Overall, the novel pleiotropic effect of mdivi-1 in cardiomyocytes included proteolytically cleaved L-OPA1, altered expression of OXPHOS complex proteins, and increased superoxide production, which together resulted in defects in mitochondrial respiration and inhibition of macro-autophagy.


Assuntos
Dinâmica Mitocondrial , Miócitos Cardíacos , Autofagia , Dinaminas/genética , Proteínas Mitocondriais/genética , Quinazolinonas/farmacologia , Respiração
14.
J Am Heart Assoc ; 9(23): e017195, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33208022

RESUMO

Background The mutated α-B-Crystallin (CryABR120G) mouse model of desmin-related myopathy (DRM) shows an age-dependent onset of pathologic cardiac remodeling and progression of heart failure. CryABR120G expression in cardiomyocytes affects the mitochondrial spatial organization within the myofibrils, but the molecular perturbation within the mitochondria in the relation of the overall course of the proteotoxic disease remains unclear. Methods and Results CryABR120G mice show an accumulation of electron-dense aggregates and myofibrillar degeneration associated with the development of cardiac dysfunction. Though extensive studies demonstrated that these altered ultrastructural changes cause cardiac contractility impairment, the molecular mechanism of cardiomyocyte death remains elusive. Here, we explore early pathological processes within the mitochondria contributing to the contractile dysfunction and determine the pathogenic basis for the heart failure observed in the CryABR120G mice. In the present study, we report that the CryABR120G mice transgenic hearts undergo altered mitochondrial dynamics associated with increased level of dynamin-related protein 1 and decreased level of optic atrophy type 1 as well as mitofusin 1 over the disease process. In association with these changes, an altered level of the components of mitochondrial oxidative phosphorylation and pyruvate dehydrogenase complex regulatory proteins occurs before the manifestation of pathologic adverse remodeling in the CryABR120G hearts. Mitochondria isolated from CryABR120G transgenic hearts without visible pathology show decreased electron transport chain complex activities and mitochondrial respiration. Taken together, we demonstrated the involvement of mitochondria in the pathologic remodeling and progression of DRM-associated cellular dysfunction. Conclusions Mitochondrial dysfunction in the form of altered mitochondrial dynamics, oxidative phosphorylation and pyruvate dehydrogenase complex proteins level, abnormal electron transport chain complex activities, and mitochondrial respiration are evident on the CryABR120G hearts before the onset of detectable pathologies and development of cardiac contractile dysfunction.


Assuntos
Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Dinâmica Mitocondrial/fisiologia , Fosforilação Oxidativa , Animais , Cardiomiopatias/metabolismo , Desmina , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Cadeia B de alfa-Cristalina
15.
Commun Biol ; 3(1): 682, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33203971

RESUMO

Methamphetamine-associated cardiomyopathy is the leading cause of death linked with illicit drug use. Here we show that Sigmar1 is a therapeutic target for methamphetamine-associated cardiomyopathy and defined the molecular mechanisms using autopsy samples of human hearts, and a mouse model of "binge and crash" methamphetamine administration. Sigmar1 expression is significantly decreased in the hearts of human methamphetamine users and those of "binge and crash" methamphetamine-treated mice. The hearts of methamphetamine users also show signs of cardiomyopathy, including cellular injury, fibrosis, and enlargement of the heart. In addition, mice expose to "binge and crash" methamphetamine develop cardiac hypertrophy, fibrotic remodeling, and mitochondrial dysfunction leading to contractile dysfunction. Methamphetamine treatment inhibits Sigmar1, resulting in inactivation of the cAMP response element-binding protein (CREB), decreased expression of mitochondrial fission 1 protein (FIS1), and ultimately alteration of mitochondrial dynamics and function. Therefore, Sigmar1 is a viable therapeutic agent for protection against methamphetamine-associated cardiomyopathy.


Assuntos
Cardiomiopatias/induzido quimicamente , Metanfetamina/toxicidade , Mitocôndrias/efeitos dos fármacos , Receptores sigma/metabolismo , Animais , Cardiomiopatias/prevenção & controle , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Esquema de Medicação , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Humanos , Metanfetamina/administração & dosagem , Camundongos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Receptores sigma/genética , Receptor Sigma-1
16.
Sci Rep ; 9(1): 2002, 2019 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-30765730

RESUMO

Doxorubicin (Dox) is a highly effective anticancer drug but cause acute ventricular dysfunction, and also induce late-onset cardiomyopathy and heart failure. Despite extensive studies, the pathogenic sequelae leading to the progression of Dox-associated cardiomyopathy remains unknown. We assessed temporal changes in autophagy, mitochondrial dynamics, and bioenergetics in mouse models of acute and chronic Dox-cardiomyopathy. Time course study of acute Dox-treatment showed accumulation of LC3B II in heart lysates. Autophagy flux assays confirmed that the Dox-induced accumulation of autophagosomes occurs due to blockage of the lysosomal degradation process. Dox-induced autophagosomes and autolysosome accumulation were confirmed in vivo by using GFP-LC3 and mRFP-GFP-LC3 transgenic (Tg) mice. Mitochondria isolated from acute Dox-treated hearts showed significant suppression of oxygen consumption rate (OCR). Chronic Dox-cardiotoxicity also exhibited time-dependent accumulation of LC3B II levels and increased accumulation of green puncta in GFP-LC3 Tg hearts. Mitochondria isolated from chronic Dox-treated hearts also showed significant suppression of mitochondrial OCR. The in vivo impairment of autophagic degradation process and mitochondrial dysfunction data were confirmed in vitro using cultured neonatal cardiomyocytes. Both acute and chronic Dox-associated cardiomyopathy involves a multifocal disease process resulting from autophagosomes and autolysosomes accumulation, altered expression of mitochondrial dynamics and oxidative phosphorylation regulatory proteins, and mitochondrial respiratory dysfunction.


Assuntos
Autofagia/efeitos dos fármacos , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/patologia , Respiração Celular/efeitos dos fármacos , Doxorrubicina/efeitos adversos , Mitocôndrias/efeitos dos fármacos , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Feminino , Masculino , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fosforilação Oxidativa/efeitos dos fármacos
17.
J Am Heart Assoc ; 7(14)2018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29987122

RESUMO

BACKGROUND: Desmin filament proteins interlink the contractile myofibrillar apparatus with mitochondria, nuclei and the sarcolemma. Mutations in the human desmin gene cause cardiac disease, remodeling, and heart failure but the pathophysiological mechanisms remain unknown. METHODS AND RESULTS: Cardiomyocyte-specific overexpression of mutated desmin (a 7 amino acid deletion R172-E178, D7-Des Tg) causes accumulations of electron-dense aggregates and myofibrillar degeneration associated with cardiac dysfunction. Though extensive studies demonstrated that these altered ultrastructural changes cause impairment of cardiac contractility, the molecular mechanism of cardiomyocyte death remains elusive. In the present study, we report that the D7-Des Tg mouse hearts undergo aberrant mitochondrial fission associated with increased expression of mitochondrial fission regulatory proteins. Mitochondria isolated from D7-Des Tg hearts showed decreased mitochondrial respiration and increased apoptotic cell death. Overexpression of mutant desmin by adenoviral infection in cultured cardiomyocytes led to increased mitochondrial fission, inhibition of mitochondrial respiration, and activation of cellular toxicity. Inhibition of mitochondrial fission by mitochondrial division inhibitor mdivi-1 significantly improved mitochondrial respiration and inhibited cellular toxicity associated with D7-Des overexpression in cardiomyocytes. CONCLUSIONS: Aberrant mitochondrial fission results in mitochondrial respiratory defects and apoptotic cell death in D7-Des Tg hearts. Inhibition of aberrant mitochondrial fission using mitochondrial division inhibitor significantly preserved mitochondrial function and decreased apoptotic cell death. Taken together, our study shows that maladaptive aberrant mitochondrial fission causes desminopathy-associated cellular dysfunction.


Assuntos
Cardiomiopatias/genética , DNA/genética , Desmina/genética , Mitocôndrias Cardíacas/metabolismo , Mutação , Miócitos Cardíacos/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Células Cultivadas , Análise Mutacional de DNA , Desmina/metabolismo , Modelos Animais de Doenças , Imuno-Histoquímica , Mitocôndrias Cardíacas/patologia , Miócitos Cardíacos/patologia , Ratos Sprague-Dawley
18.
J Am Heart Assoc ; 7(20): e009775, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30371279

RESUMO

Background The Sigma 1 receptor (Sigmar1) functions as an interorganelle signaling molecule and elicits cytoprotective functions. The presence of Sigmar1 in the heart was first reported on the basis of a ligand-binding assay, and all studies to date have been limited to pharmacological approaches using less-selective ligands for Sigmar1. However, the physiological function of cardiac Sigmar1 remains unknown. We investigated the physiological function of Sigmar1 in regulating cardiac hemodynamics using the Sigmar1 knockout mouse (Sigmar1-/-). Methods and Results Sigmar1-/- hearts at 3 to 4 months of age showed significantly increased contractility as assessed by left ventricular catheterization with stimulation by increasing doses of a ß1-adrenoceptor agonist. Noninvasive echocardiographic measurements were also used to measure cardiac function over time, and the data showed the development of cardiac contractile dysfunction in Sigmar1 -/- hearts as the animals aged. Histochemistry demonstrated significant cardiac fibrosis, collagen deposition, and increased periostin in the Sigmar1 -/- hearts compared with wild-type hearts. Ultrastructural analysis of Sigmar1-/- cardiomyocytes revealed an irregularly shaped, highly fused mitochondrial network with abnormal cristae. Mitochondrial size was larger in Sigmar1-/- hearts, resulting in decreased numbers of mitochondria per microscopic field. In addition, Sigmar1-/- hearts showed altered expression of mitochondrial dynamics regulatory proteins. Real-time oxygen consumption rates in isolated mitochondria showed reduced respiratory function in Sigmar1-/- hearts compared with wild-type hearts. Conclusions We demonstrate a potential function of Sigmar1 in regulating normal mitochondrial organization and size in the heart. Sigmar1 loss of function led to mitochondrial dysfunction, abnormal mitochondrial architecture, and adverse cardiac remodeling, culminating in cardiac contractile dysfunction.


Assuntos
Cardiopatias/fisiopatologia , Mitocôndrias Cardíacas/fisiologia , Dinâmica Mitocondrial/fisiologia , Receptores sigma/metabolismo , Trifosfato de Adenosina/metabolismo , Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Animais , Biomarcadores/metabolismo , Respiração Celular/fisiologia , Dobutamina/farmacocinética , Ecocardiografia , Transporte de Elétrons/fisiologia , Metabolismo Energético/fisiologia , Feminino , Fibrose/fisiopatologia , Hemodinâmica/fisiologia , Masculino , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Mitocôndrias Cardíacas/ultraestrutura , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/fisiopatologia , Contração Miocárdica/fisiologia , Miocárdio/patologia , Miócitos Cardíacos/fisiologia , Miócitos Cardíacos/ultraestrutura , Consumo de Oxigênio/fisiologia , Receptor Sigma-1
19.
Am J Clin Nutr ; 86(4): 1202-9, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17921403

RESUMO

BACKGROUND: Chronic arsenic exposure currently affects >100 million persons worldwide. Methylation of ingested inorganic arsenic (InAs) to monomethylarsonic (MMAs) and dimethylarsinic (DMAs) acids relies on folate-dependent one-carbon metabolism and facilitates urinary arsenic elimination. OBJECTIVE: We hypothesized that folic acid supplementation to arsenic-exposed Bangladeshi adults would increase arsenic methylation and thereby lower total blood arsenic. DESIGN: In this randomized, double-blind, placebo-controlled trial, we evaluated blood concentrations of total arsenic, InAs, MMAs, and DMAs in 130 participants with low plasma folate (<9 nmol/L) before and after 12 wk of supplementation with folic acid (400 microg/d) or placebo. RESULTS: MMAs in blood was reduced by a mean +/- SE of 22.24 +/- 2.86% in the folic acid supplementation group and by 1.24 +/- 3.59% in the placebe group (P < 0.0001). There was no change in DMAs in blood; DMAs is rapidly excreted in urine as evidenced by an increase in urinary DMAs (P = 0.0099). Total blood arsenic was reduced by 13.62% in the folic acid supplementation group and by 2.49% in the placebo group (P = 0.0199). CONCLUSIONS: Folic acid supplementation to participants with low plasma concentrations of folate lowered blood arsenic concentrations, primarily by decreasing blood MMAs and increasing urinary DMAs. Therapeutic strategies to facilitate arsenic methylation, particularly in populations with folate deficiency or hyperhomocysteinemia or both, may lower blood arsenic concentrations and thereby contribute to the prevention of arsenic-induced illnesses.


Assuntos
Arsênio/metabolismo , Suplementos Nutricionais , Ácido Fólico/farmacologia , Metilação/efeitos dos fármacos , Complexo Vitamínico B/farmacologia , Poluentes Químicos da Água/metabolismo , Arsênio/sangue , Arsênio/urina , Arsenicais/metabolismo , Arsenicais/urina , Bangladesh/epidemiologia , Método Duplo-Cego , Exposição Ambiental/efeitos adversos , Feminino , Ácido Fólico/administração & dosagem , Ácido Fólico/sangue , Homocisteína/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Complexo Vitamínico B/administração & dosagem , Complexo Vitamínico B/sangue , Poluentes Químicos da Água/sangue , Poluentes Químicos da Água/urina
20.
Disaster Med Public Health Prep ; 11(1): 21-24, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27181426

RESUMO

BACKGROUND: Rana Plaza building collapse is the worst industrial disaster of Bangladesh so far. The 9-storied structure collapsed suddenly on April 24, 2013, with more than 4000 people inside. Bangladesh Armed Forces played a key role in the massive rescue operations. METHODS: We conducted a cross-sectional study with 423 victims who were treated at a Combined Military Hospital to review the pattern of injuries and management provided. RESULTS: Middle-aged (35±12.75 years) females (68.32%) were the majority of the victims. Among the injured, 42.35% had soft tissue injury, 22.55% had abrasions, 18.79% had fractures, 3.75% had facial injuries, and 2.5% each had head and abdominal injuries. We treated the injured with various surgical approaches, such as soft tissue debridement (38.84%), fasciotomy (18.79%), amputation (3.75%), and other procedures. We had to refer 8.27% of the patients to different advanced centers. The mortality rate was 5.91%, including 1 volunteer rescuer. CONCLUSION: Pattern of injuries and modalities of management needed in an industrial disaster is a valuable experience which can be utilized in preparing to face disasters in the future and beyond. Death of a voluntary rescuer once again warrants the necessity of using a helmet and safety gear during any rescue operation. (Disaster Med Public Health Preparedness. 2017;11:21-24).


Assuntos
Serviços Médicos de Emergência/métodos , Incidentes com Feridos em Massa/estatística & dados numéricos , Trabalho de Resgate/normas , Ferimentos e Lesões/epidemiologia , Adolescente , Adulto , Bangladesh/epidemiologia , Estudos Transversais , Serviços Médicos de Emergência/estatística & dados numéricos , Feminino , Hospitais Militares/organização & administração , Hospitais Militares/estatística & dados numéricos , Humanos , Masculino , Pessoa de Meia-Idade , Militares/estatística & dados numéricos , Trabalho de Resgate/métodos , Recursos Humanos , Ferimentos e Lesões/mortalidade
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa