Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 88, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38349408

RESUMEN

Atrial fibrillation (AF) is the most prevalent sustained cardiac arrhythmia, and recent epidemiological studies suggested type 2 diabetes mellitus (T2DM) is an independent risk factor for the development of AF. Zinc finger and BTB (broad-complex, tram-track and bric-a-brac) domain containing 16 (Zbtb16) serve as transcriptional factors to regulate many biological processes. However, the potential effects of Zbtb16 in AF under T2DM condition remain unclear. Here, we reported that db/db mice displayed higher AF vulnerability and Zbtb16 was identified as the most significantly enriched gene by RNA sequencing (RNA-seq) analysis in atrium. In addition, thioredoxin interacting protein (Txnip) was distinguished as the key downstream gene of Zbtb16 by Cleavage Under Targets and Tagmentation (CUT&Tag) assay. Mechanistically, increased Txnip combined with thioredoxin 2 (Trx2) in mitochondrion induced excess reactive oxygen species (ROS) release, calcium/calmodulin-dependent protein kinase II (CaMKII) overactivation, and spontaneous Ca2+ waves (SCWs) occurrence, which could be inhibited through atrial-specific knockdown (KD) of Zbtb16 or Txnip by adeno-associated virus 9 (AAV9) or Mito-TEMPO treatment. High glucose (HG)-treated HL-1 cells were used to mimic the setting of diabetic in vitro. Zbtb16-Txnip-Trx2 signaling-induced excess ROS release and CaMKII activation were also verified in HL-1 cells under HG condition. Furthermore, atrial-specific Zbtb16 or Txnip-KD reduced incidence and duration of AF in db/db mice. Altogether, we demonstrated that interrupting Zbtb16-Txnip-Trx2 signaling in atrium could decrease AF susceptibility via reducing ROS release and CaMKII activation in the setting of T2DM.


Asunto(s)
Fibrilación Atrial , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Animales , Ratones , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Portadoras/genética , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Especies Reactivas de Oxígeno , Tiorredoxinas/genética
2.
Cytotherapy ; 26(3): 311-316, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38219142

RESUMEN

BACKGROUND: Our previous single-center, randomized, double-blinded, placebo-controlled phase 2 study evaluated the safety and effectiveness of human umbilical cord mesenchymal stromal cell (UC-MSC) transfusion for treating patients with type 2 diabetes mellitus (T2DM). Indeed, this potential treatment strategy was able to reduce insulin use by half in a considerable number of patients. However, many other patients' responses to UC-MSC transfusion were insignificant. The selection of patients who might benefit from UC-MSC treatment is crucial from a clinical standpoint. METHODS: In this post hoc analysis, 37 patients who received UC-MSC transfusions were divided into two groups based on whether their glycated hemoglobin (hemoglobin A1c, or HbA1c) level was less than 7% after receiving UC-MSC treatment. The baseline differences between the two groups were summarized, and potential factors influencing efficacy of UC-MSCs for T2DM were analyzed by univariate and multivariate logistic regression. The correlations between the relevant hormone levels and the treatment effect were further analyzed. RESULTS: At the 9-week follow-up, 59.5% of patients achieved their targeted HbA1c level. Male patients with lower baseline HbA1c and greater C-peptide area under the curve (AUCC-pep) values responded favorably to UC-MSC transfusion, according to multivariate analysis. The effectiveness of UC-MSCs transfusion was predicted by AUCC-pep (cutoff value: 14.22 ng/h/mL). Further investigation revealed that AUCC-pep was increased in male patients with greater baseline testosterone levels. CONCLUSIONS: Male patients with T2DM with greater AUCC-pep may be more likely to respond clinically to UC-MSC therapy, and further large-scale multi-ethnic clinical studies should be performed to confirm the conclusion.


Asunto(s)
Diabetes Mellitus Tipo 2 , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Humanos , Masculino , Diabetes Mellitus Tipo 2/terapia , Diabetes Mellitus Tipo 2/metabolismo , Hemoglobina Glucada , Cordón Umbilical , Resultado del Tratamiento , Células Madre Mesenquimatosas/fisiología
3.
Langmuir ; 40(11): 6026-6034, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38451161

RESUMEN

The photocatalytic transformation of carbon dioxide (CO2) into valuable chemicals is a challenging process that requires effective and selective catalysts. However, most polymer-based photocatalysts with electron donor-acceptor (D-A) structures are synthesized with a fixed D-A ratio by using expensive monomers. Herein, we report a simple strategy to prepare polyarene oxides (PAOs) with quinone structural units via oxidation treatment of polyarene (PA). The resultant PAOs show tunable D-A structures and electronic band positions depending on the degree of oxidation, which can catalyze the photoreduction of CO2 with water under visible light irradiation, generating CO as the sole carbonaceous product without H2 generation. Especially, the PAO with an oxygen content of 17.6% afforded the highest CO production rate of 161.9 µmol g-1 h-1. It is verified that the redox transformation between quinone and phenolic hydroxyl in PAOs achieves CO2 photoreduction coupled with water oxidation. This study provides a facile way to access conjugated polymers with a tunable D-A structure and demonstrates that the resultant PAOs are promising photocatalysts for CO2 reduction.

4.
J Mol Cell Cardiol ; 182: 57-72, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37482037

RESUMEN

Hypertension-induced tunica media thickening (TMT) is the most important fundamental for the subsequent complications like stroke and cardiovascular diseases. Pathogenically, TMT originates from both vascular smooth muscle cells (VSMCs) hypertrophy due to synthesizing more amount of intracellular contractile proteins and excess secretion of extracellular matrix. However, what key molecules are involved in the pathogenesis of TMT is unknown. We hypothesize that formin homology 2 domain-containing protein 1 (FHOD1), an amply expressed mediator for assembly of thin actin filament in VSMCs, is a key regulator for the pathogenesis of TMT. In this study, we found that FHOD1 expression and its phosphorylation/activation were both upregulated in the arteries of three kinds of hypertensive rats. Ang-II induced actin filament formation and hypertrophy through activation and upregulation of FHOD1 in VSMCs. Active FHOD1-mediated actin filament assembly and secretions of collagen-1α/collagen-3α played crucial roles in Ang-II-induced VSMCs hypertrophy in vitro and hypertensive TMT in vivo. Proteomics demonstrated that activated FL-FHOD1 or its C-terminal diaphanous-autoregulatory domain significantly upregulated RNF213 (ring finger protein 213), a 591-kDa cytosolic E3 ubiquitin ligase with its loss-of-functional mutations being a susceptibility gene for Moyamoya disease which has prominent tunica media thinning in both intracranial and systemic arteries. Mechanistically, activated FHOD1 upregulated its downstream effector RNF213 independently of its classical pathway of decreasing G-actin/F-actin ratio, transcription, and translation, but dependently on its C-terminus-mediated stabilization of RNF213 protein. FHOD1-RNF213 signaling dramatically promoted collagen-1α/collagen-3α syntheses in VSMCs. Our results discovered a novel signaling axis of FHOD1-RNF213-collagen-1α/collagen-3α and its key role in the pathogenesis of hypertensive TMT.


Asunto(s)
Actinas , Hipertensión , Animales , Ratas , Actinas/metabolismo , Hipertensión/etiología , Hipertrofia , Transducción de Señal/fisiología , Factores de Transcripción , Túnica Media/metabolismo
5.
Int J Mol Sci ; 24(7)2023 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-37047522

RESUMEN

Heart failure is a serious and life-threatening disease worldwide. Cadherin-11 (Cad-11) is highly expressed in the heart and closely associated with inflammation. There is currently limited understanding on how Cad-11 contributes to cardiac remodeling and its underline molecular mechanism. We found an increased expression of Cad-11 in biopsy heart samples from heart failure patients, suggesting a link between Cad-11 and heart failure. To determine the role of Cad-11 in cardiac remodeling, Cad-11-deficient mice were used in a well-established mouse transverse aortic constriction (TAC) model. Loss of Cad11 greatly improved pressure overload-induced LV structural and electrical remodeling. IL (interleukin)-6 production was increased following TAC in WT mice and this increase was inhibited in cadherin-11-/- mice. We further tested the effect of IL-6 on myocyte hypertrophy and fibrosis in a primary culture system. The addition of hCad-11-Fc to cultured cardiac fibroblasts increased IL-6 production and fibroblast cell activation, whereas neutralizing IL-6 with an IL-6 antibody resulted in alleviating the fibroblast activation induced by hCad-11-Fc. On the other hand, cardiomyocytes were promoted to cardiomyocyte hypertrophy when cultured in condition media collected from cardiac fibroblasts stimulated by hCad-11-Fc.Similarly, neutralizing IL-6 prevented cardiomyocyte hypertrophy. Finally, we found that MAPKs and CaMKII-STAT3 pathways were activated in both hCad-11-Fc stimulated fibroblasts and cardiomyocytes treated with hCad-11-Fc stimulated fibroblast condition medium. IL-6 neutralization inhibited such MAPK and CaMKII-STAT3 signaling activation. These data demonstrate that Cad-11 functions in pressure overload-induced ventricular remodeling through inducing IL-6 secretion from cardiac fibroblasts to modulate the pathophysiology of neighboring cardiomyocytes.


Asunto(s)
Insuficiencia Cardíaca , Miocitos Cardíacos , Ratones , Animales , Miocitos Cardíacos/metabolismo , Interleucina-6/metabolismo , Remodelación Ventricular , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Insuficiencia Cardíaca/metabolismo , Fibroblastos/metabolismo , Hipertrofia/metabolismo , Ratones Endogámicos C57BL , Fibrosis , Cardiomegalia/metabolismo
6.
BMC Oral Health ; 23(1): 411, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37344840

RESUMEN

OBJECTIVE: To analyze the abundance of infiltrating tumor immune cells in patients with oral squamous cell carcinoma (OSCC) and to search for potential targets that can predict patient prognosis. METHODS: A total of 400 samples from 210 patients with OSCC were collected using The Cancer Genome Atlas (TCGA) database. CIBERSORTx was used to evaluate the infiltration abundance of tumor immune cells. Potential target genes were searched to predict patient prognosis through case grouping, differential analysis, and enrichment analysis. Surgical excisional tissue sections of patients with oral squamous cell carcinoma admitted to the Department of Oral and Maxillofacial Surgery, Second Affiliated Hospital of Shantou University Medical College, from 2015 to 2018 were collected and followed up. RESULTS: The CIBERSORTx deconvolution algorithm was used to analyze the infiltration abundance of immune cells in the samples. Cases with a high infiltration abundance of naive and memory B lymphocytes improved the prognosis of OSCC patients. The prognosis of patients with low CD79A expression was significantly better than that of patients with high CD79A expression. CONCLUSION: CD79A can predict the infiltration abundance of B lymphocytes in the tumor microenvironment of patients with OSCC. CD79A is a potential target for predicting the prognosis of patients with OSCC. This study provides novel ideas for the treatment of OSCC and for predicting patient prognosis.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello , Carcinoma de Células Escamosas/patología , Neoplasias de la Boca/patología , Pronóstico , Microambiente Tumoral , Antígenos CD79
7.
BMC Oral Health ; 22(1): 213, 2022 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-35643546

RESUMEN

BACKGROUND: To assess the contributing risk factors for the progression of, and the postoperative poor prognosis associated with, osteoradionecrosis of jaw (ORNJ) following non-nasopharyngeal cancer treatment in head and neck. METHODS: A retrospective study of 124 non-nasopharyngeal carcinoma patients in head and neck treated at one institution between 2001 and 2020 was conducted. A cumulative meta-analysis was conducted according to PRISMA protocol and the electronic search was performed on the following search engines: PubMed, Embase, and Web of Science. After assessing surgery with jaw lesions as a risk factor for the occurrence of ORNJ, 124 cases were categorized into two groups according to the "BS" classification, after which jaw lesions, chemotherapy, flap reconstruction and onset time of ORNJ were analyzed through the chi-square test and t-test to demonstrate the potential association between them and the progression of ORNJ. Postoperative outcomes of wound healing, occlusal disorders, and nerve injury were statistically analyzed. RESULTS: With the statistically significant results of the meta-analysis (odds ratio = 3.07, 95% CI: 1.84-5.13, p < 0.0001), the chi-square test and t-test were used to validate our hypotheses and identified that surgery with jaw lesions could aggravate the progression and accelerate the appearance of ORNJ. Patients who underwent chemotherapy tended to suffer from severe-to-advanced osteonecrosis but did not shorten the onset time of ORNJ. Flap reconstruction presented obvious advantages in wound healing (p < 0.001) and disordered occlusion (p < 0.005). The mean onset time of ORNJ in non-nasopharyngeal cancer patients (4.5 years) was less than that in patients with nasopharyngeal cancer (NPC) (6.8 years). CONCLUSIONS: Iatrogenic jaw lesions are evaluated as a significant risk factor in the occurrence and progression of ORNJ in non-nasopharyngeal carcinoma patients who tend to have more severe and earlier osteonecrosis after radiotherapy than NPC patients. Flap reconstruction is a better choice for protecting the remaining bone tissue and reducing postoperative complications of ORNJ.


Asunto(s)
Neoplasias Nasofaríngeas , Osteonecrosis , Osteorradionecrosis , Humanos , Carcinoma Nasofaríngeo , Osteonecrosis/complicaciones , Osteorradionecrosis/etiología , Complicaciones Posoperatorias , Estudios Retrospectivos
8.
BMC Oral Health ; 22(1): 322, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35915482

RESUMEN

BACKGROUND: We established a MSBOS for flap reconstruction in oral and maxillofacial cancer patients. METHOD: We enrolled 2080 cases of oral and maxillofacial flap reconstruction from January 1, 2010 to December 31, 2021. Patient data were collected, including age, sex, BMI, preoperative Hb levels, ASA grade, T stage, flap type, tumor location, and bone flap. Scoring criteria were established based on a multivariate model of independent risk variables and their odds ratios. Two flap-type groups were divided into low-risk, intermediate-risk and high-risk groups by the scoring criteria, and analyzed using univariate and multivariate logistic regression. Perioperative transfusion analysis identified independent risk factors at various Hb levels. The cumulative percentage of patients requiring perioperative blood transfusion for each surgical procedure was calculated to establish the MSBOS. RESULTS: (1) Regression analysis showed that BMI, tumor T staging, ASA grade, preoperative Hb level (male: Hb < 130 g/L, female: Hb < 120 g/L), and bone flap were independent risk factors for perioperative blood transfusion. (2) Regression analysis showed that independent risk factors for perioperative transfusion included the following: BMI, tumor T3-T4 stage, ASA III, IV grade, and free flap/pediculated flap/bone flap in patients with different Hb levels; T3-T4 stage, ASA grade III-IV in mildly anemic patients; and ASA grade III-IV in moderately anemic patients. (3) A MSBOS was established for flap reconstruction in head and neck cancer patients. CONCLUSION: A MSBOS for head and neck cancer procedures was reduced by approximately 30% perioperative blood preparation while ensuring that clinical blood use standards were met. It help optimize blood inventory, and save blood resources.


Asunto(s)
Transfusión Sanguínea , Neoplasias de Cabeza y Cuello , Procedimientos de Cirugía Plástica , Colgajos Quirúrgicos , Femenino , Neoplasias de Cabeza y Cuello/cirugía , Humanos , Masculino , Procedimientos de Cirugía Plástica/métodos , Estudios Retrospectivos , Factores de Riesgo
9.
J Cell Physiol ; 236(8): 5725-5741, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33452701

RESUMEN

Atrial fibrillation (AF) is the most common cardiac arrhythmia nowadays. The occurrence of AF is closely associated with obesity. Cadherin-11 (Cad-11), as a member of the cadherin family, can make a contribution to diet-induced obesity and it will be informative to know whether Cad-11 exerts its effects on atrial remodeling and AF vulnerability in a diet-induced obesity model. In this study, we demonstrated that the expression of Cad-11 was significantly upregulated in the left atrium of AF patients with obesity and mice following 16 weeks of high-fat diet (HFD) feeding. Further confirmed that Cad-11 could regulate the activity of atrial fibroblasts by participating in inducing proinflammatory cytokines production. At animal levels, we found that although there was a lack of statistical difference in body weight, Cad-11-/- mice could markedly improve impaired glucose tolerance and hyperlipidemia. Adverse atrial structural remodeling, including atrial enlargement, inflammation, and fibrosis provoked by HFD feeding were mitigated in Cad-11-/- mice. Mechanistically, Cad-11 activated mitogen-activated protein kinases and nuclear factor-κB for interleukin-6 production in atrial fibroblasts that may contribute to the atrial fibrosis process in obesity-related AF, suggesting Cad-11 might be a new therapeutic target for obesity-related AF.


Asunto(s)
Fibrilación Atrial/metabolismo , Remodelación Atrial/genética , Cadherinas/deficiencia , Dieta Alta en Grasa , Inflamación/metabolismo , Animales , Remodelación Atrial/fisiología , Cardiomiopatías/patología , Fibrosis/genética , Fibrosis/metabolismo , Atrios Cardíacos/fisiopatología , Humanos , Inflamación/patología , Ratones
10.
J Mol Cell Cardiol ; 135: 119-133, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31408621

RESUMEN

Angiotensin II (Ang-II)-induced fibroblast differentiation plays an important role in the development of atrial fibrosis and atrial fibrillation (AF). Here, we show that the expression of the histone methyltransferase enhancer of zeste homolog 2 (EZH2) is increased in atrial muscle and atrial fibroblasts in patients with AF, accompanied by significant atrial fibrosis and atrial fibroblast differentiation. In addition, EZH2 is induced in murine models of atrial fibrosis. Furthermore, either pharmacological GSK126 inhibition or molecular silencing of EZH2 can inhibit the differentiation of atrial fibroblasts and the ability to produce ECM induced by Ang-II. Simultaneously, inhibition of EZH2 can block the Ang-II-induced migration of atrial fibroblasts. We found that EZH2 promotes fibroblast differentiation mainly through the Smad signaling pathway and can form a transcription complex with Smad2 to bind to the promoter region of the ACTA2 gene. Finally, our in vivo experiments demonstrated that the EZH2 inhibitor GSK126 significantly inhibited Ang-II-induced atrial enlargement and fibrosis and reduced AF vulnerability. Our results demonstrate that targeting EZH2 or EZH2-regulated genes might present therapeutic potential in AF.


Asunto(s)
Fibrilación Atrial , Proteína Potenciadora del Homólogo Zeste 2 , Fibroblastos , Regulación de la Expresión Génica/efectos de los fármacos , Indoles/farmacología , Piridonas/farmacología , Angiotensina II/efectos adversos , Angiotensina II/farmacología , Animales , Fibrilación Atrial/inducido químicamente , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Modelos Animales de Enfermedad , Perros , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad
11.
J Mol Cell Cardiol ; 127: 83-96, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30529267

RESUMEN

Cardiac fibrosis (CF), a process characterized by potentiated proliferation of cardiac fibroblasts and excessive secretion and deposition of extracellular matrix (ECM) from the cells, contributes strongly to the pathogenesis of a series of cardiovascular (CV) diseases, including AMI, heart failure and atrial fibrillation. Endothelial-mesenchymal transition (EndMT), one of the sources of transformed cardiac fibroblasts, has been reported as a key factor involved in CF. However, the molecular basis of EndMT has not been thoroughly elucidated to date. At the posttranscriptional level, of the three epigenetic regulators, writer and eraser are reported to be involved in EndMT, but the role of reader in the process is still unknown. In this study, we aimed to explore the role of Bromodomain-containing protein 4 (BRD4), an acetyl-lysine reader protein, in EndMT-induced CF and related mechanisms. We found that BRD4 was upregulated in endothelial cells (ECs) in the pressure-overload mouse heart and that its functional inhibitor JQ1 potently attenuated the TAC-induced CF and preserved cardiac function. In umbilical vein endothelial cells (HUVECs) and mouse aortic endothelial cells (MAECs), bothJQ1 and shRNA-mediated silencing of BRD4 blocked TGF-ß-induced EC migration, EndMT and ECM synthesis and preserved the EC sprouting behavior, possibly through the downregulation of a group of transcription factors specific for EndMT (Snail, Twist and Slug), the Smads pathway and TGF-ß receptor I. In the absence of TGF-ß stimulation, ectopic expression of BRD4 alone could facilitate EndMT, accelerate migration and increase the synthesis of ECM. In vivo, JQ1 also attenuated TAC-induced EndMT and CF, which was consistent with JQ1's intracellular mechanisms of action. Our results showed that BRD4 plays a critical role in EndMT-induced CF and that targeting BRD4 might be a novel therapeutic option for CF.


Asunto(s)
Aorta/patología , Proteínas de Ciclo Celular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Mesodermo/metabolismo , Miocardio/patología , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/efectos adversos , Animales , Biomarcadores/metabolismo , Movimiento Celular/efectos de los fármacos , Constricción , Regulación hacia Abajo/efectos de los fármacos , Proteínas de la Matriz Extracelular/biosíntesis , Fibroblastos/metabolismo , Fibrosis , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo
12.
J Mol Cell Cardiol ; 105: 1-11, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28214509

RESUMEN

Cardiac fibrosis (CF), a repairing process following myocardial infarction (MI), is characterized by abnormal proliferation of cardiac fibroblasts and excessive deposition of extracellular matrix (ECM) resulting in inevitable resultant heart failure. TGF-ß (transforming growth factor-ß)/ALK5 (Activin receptor-like kinase 5)/Smad2/3/4 pathways have been reported to be involved in the process. Recent studies have implicated both activin and its specific downstream component ALK4 in stimulating fibrosis in non-cardiac organs. We recently reported that ALK4 is upregulated in the pressure-overloaded heart and its partial inhibition attenuated the pressure overload-induced CF and cardiac dysfunction. However, the role of ALK4 in the pathogenesis of MI-induced CF, which is usually more severe than that induced by pressure-overload, remains unknown. Here we report: 1) In a wild-type mouse model of MI, ALK4 upregulation was restricted in the fibroblasts of the infarct border zone; 2) In contrast, ALK4+/- mice with a haplodeficiency of ALK4 gene, showed a significantly attenuated CF in the border zone, with a smaller scar size, a preserved cardiac function and an improved survival rate post-MI; 3) Similarly to pressure-overloaded heart, these beneficial effects might be through a partial inactivation of the Smad3/4 pathway but not MAPK cascades; 4) The apoptotic rate of the cardiomyocytes were indistinguishable in the border zone of the wild-type control and ALK4+/- mice; 5) Cardiac fibroblasts isolated from ALK4+/- mice showed reduced migration, proliferation and ECM synthesis in response to hypoxia. These results indicate that partial inhibition of ALK4 may reduce MI-induced CF, suggesting ALK4 as a novel target for inhibition of unfavorable CF and for preservation of LV systolic function induced by not only pressure-overload but also MI.


Asunto(s)
Receptores de Activinas Tipo I/genética , Haploinsuficiencia , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Receptores de Activinas Tipo I/deficiencia , Receptores de Activinas Tipo I/metabolismo , Animales , Movimiento Celular/genética , Proliferación Celular , Modelos Animales de Enfermedad , Ecocardiografía , Matriz Extracelular , Fibrosis , Regulación de la Expresión Génica , Genotipo , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Mortalidad , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miofibroblastos/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Proteína Smad4/metabolismo , Función Ventricular
13.
Basic Res Cardiol ; 112(4): 47, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28639003

RESUMEN

Atrial fibrosis, the hallmark of structural remodeling associated with atrial fibrillation (AF), is characterized by abnormal proliferation of atrial fibroblasts and excessive deposition of extracellular matrix. Transforming growth factor-ß1 (TGF-ß1)/activin receptor-like kinase 5 (ALK5)/Smad2/3/4 pathway has been reported to be involved in the process. Recent studies have implicated both activin A and its specific downstream component activin receptor-like kinase 4 (ALK4) in stimulating fibrosis in non-cardiac organs. We recently reported that ALK4 haplodeficiency attenuated the pressure overload- and myocardial infarction-induced ventricular fibrosis. However, the role of activin A/ALK4 in the pathogenesis of atrial fibrosis and vulnerability to AF remains unknown. Our study provided experimental and clinical evidence for the involvement of activin A and ALK4 in the pathophysiology of atrial fibrosis and AF. Patients with AF had higher activin A and ALK4 expression in atriums as compared to individuals devoid of AF. After angiotensin-II (Ang-II) stimulation which mimicked atrial fibrosis progression, ALK4-deficient mice showed lower expression of ALK4 in atriums, reduced activation of atrial fibroblasts, blunted atrial enlargement and atrial fibrosis, and further reduced AF vulnerability upon right atrial electrophysiological studies as compared to wild-type littermates. Moreover, we found that apart from the well-known TGF-ß1/ALK5 pathway, the activation of activin A/ALK4/smad2/3 pathway played an important role in the pathogenesis of Ang-II-mediated atrial fibrosis and inducibility of AF, suggesting that targeting ALK4 might be a potential therapy for atrial fibrosis and AF.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Activinas/metabolismo , Fibrilación Atrial/metabolismo , Atrios Cardíacos/patología , Adulto , Anciano , Angiotensina II/toxicidad , Animales , Fibrilación Atrial/patología , Femenino , Fibrosis , Atrios Cardíacos/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad
15.
Brain Inj ; 31(13-14): 1863-1870, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28945462

RESUMEN

PRIMARY OBJECTIVE: This qualitative study aimed to gain a better understanding of how medical and social services in the UK currently support patients with Traumatic Brain Injury (TBI) in the community. Furthermore, we explored patients' wishes and expectations of a newly established TBI clinic. METHODS AND PROCEDURES: We conducted semi-structured interviews with 10 patients with mild-to-severe TBI. The interview schedule was designed to cover contacts with health services, information provided, post-discharge support, current social circumstances, expectations from the newly established brain injury service and participants' desires for any new service. Transcripts were analysed using a thematic analysis. MAIN RESULTS: Participants highlighted the importance of the human component of their care and of fostering trusting relationships. This validates patients' experience and helps them to regain confidence. Follow-up and education are important for patients and relatives through all stages of care, regardless of the severity of the injury. Patients strive for meaningful lives and need to be supported to engage in activities. They need hands-on support, particularly with the UK's bureaucratic welfare system. CONCLUSIONS: There is much room for improvement in the TBI community care in the UK. Our findings support the development of a holistic service that can address the multifactorial problems which the patients with TBI and their families face.


Asunto(s)
Lesiones Traumáticas del Encéfalo/psicología , Lesiones Traumáticas del Encéfalo/rehabilitación , Personal de Salud/psicología , Necesidades y Demandas de Servicios de Salud , Adolescente , Adulto , Anciano , Actitud del Personal de Salud , Femenino , Humanos , Relaciones Interpersonales , Masculino , Persona de Mediana Edad , Investigación Cualitativa , Características de la Residencia , Apoyo Social , Reino Unido , Adulto Joven
16.
Int J Mol Sci ; 18(4)2017 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-28358314

RESUMEN

BACKGROUND: Hydrogen peroxide (H2O2)-induced oxidative stress has been demonstrated to induce afterdepolarizations and triggered activities in isolated myocytes, but the underlying mechanisms remain not fully understood. We aimed to explore whether protein kinase C (PKC) activation plays an important role in oxidative stress-induced afterdepolarizations. METHODS: Action potentials and ion currents of isolated rabbit cardiomyocytes were recorded using the patch clamp technique. H2O2 (1 mM) was perfused to induce oxidative stress and the specific classical PKC inhibitor, Gö 6983 (1 µM), was applied to test the involvement of PKC. RESULTS: H2O2 perfusion prolonged the action potential duration and induced afterdepolarizations. Pretreatment with Gö 6983 prevented the emergence of H2O2-induced afterdepolarizations. Additional application of Gö 6983 with H2O2 effectively suppressed H2O2-induced afterdepolarizations. H2O2 increased the late sodium current (INa,L) (n = 7, p < 0.01) and the L-type calcium current (ICa,L) (n = 5, p < 0.01), which were significantly reversed by Gö 6983 (p < 0.01). H2O2 also increased the transient outward potassium current (Ito) (n = 6, p < 0.05). However, Gö 6983 showed little effect on H2O2-induced enhancement of Ito. CONCLUSIONS: H2O2 induced afterdepolarizations via the activation of PKC and the enhancement of ICa,L and INa,L. These results provide evidence of a link between oxidative stress, PKC activation and afterdepolarizations.


Asunto(s)
Potenciales de la Membrana , Miocitos Cardíacos/fisiología , Estrés Oxidativo , Proteína Quinasa C/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Miocitos Cardíacos/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Conejos
17.
Basic Res Cardiol ; 111(2): 19, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26891723

RESUMEN

Dual-specificity phosphatase 14 (Dusp14), an important negative modulator of mitogen-activated protein kinase (MAPK) signaling pathways, has been implicated in inflammatory immune response, cancers, cell differentiation and proliferation. The role of Dusp14 in chronic pressure overload-induced cardiac hypertrophy has not been explored. Here we have shown that Dusp14-/- knockout mice and cardiac-specific Dusp14 transgenic mice were generated and subjected to aortic banding (AB) for 4 weeks. Our results demonstrated that genetic loss of Dusp14 significantly aggravated cardiac hypertrophy, fibrosis, ventricular dilation and dysfunction, whereas transgenic cardiac-specific Dusp14 overexpression significantly attenuated AB-induced cardiac dysfunction and remodeling. In vitro, adenoviral overexpression of constitutive Dusp14 blocked angiotensin II-induced hypertrophic growth of cardiomyocytes, while Dusp14 knockdown led to opposite effects. Mechanistically, excessive phosphorylation of TAK1, P38MAPK and JNK1/2 was evidenced in Dusp14-/- knockout mice post-AB and inactivation of TAK1-P38MAPK and -JNK1/2 signaling using TAK1 inhibitor 5Z-7-ox shares similar antihypertrophic effect as Dusp14 overexpression. Moreover, we show that Dusp14 directly interacted with TAK1. Results from present experiments indicate that Dusp14 protects the heart from AB-induced cardiac hypertrophy and dysfunction possibly through inactivation of TAK1-P38MAPK/-JNK1/2 signaling pathway. Future studies are warranted to test the feasibility of overexpressing Dusp14 as a therapeutic strategy to attenuate cardiac hypertrophy and failure.


Asunto(s)
Cardiomegalia/enzimología , Fosfatasas de Especificidad Dual/metabolismo , Insuficiencia Cardíaca/enzimología , Sistema de Señalización de MAP Quinasas , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Angiotensina II , Animales , Secuencia de Bases , Estudios de Casos y Controles , Células Cultivadas , Fosfatasas de Especificidad Dual/genética , Células HEK293 , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Miocitos Cardíacos/fisiología , Ratas
18.
Europace ; 18(12): 1886-1896, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26541708

RESUMEN

AIMS: Myocardial infarction (MI) induces neural remodelling of the left stellate ganglion (LSG), which may contribute to ischaemia-induced arrhythmias. The neural chemorepellent Semaphorin 3a (Sema3a) has been identified as a negative regulator of sympathetic innervation in the LSG and heart. We previously reported that overexpression of Sema3a in the border zone could reduce the arrhythmogenic effects of cardiac sympathetic hyperinnervation post-MI. This study investigated whether Sema3a overexpression within the LSG confers an antiarrhythmic effect after MI through decreasing extra- and intra-cardiac neural remodelling. METHODS AND RESULTS: Sprague-Dawley rats were subjected to MI, and randomly allocated to intra-LSG microinjection of either phosphate-buffered saline (PBS), adenovirus encoding green fluorescent protein (AdGFP), or adenovirus encoding Sema3a (AdSema3a). Sham-operated rats served as controls. Two weeks after infarction, MI-induced nerve sprouting and sympathetic hyperinnervation in the LSG and myocardium were significantly attenuated by intra-LSG injection with AdSema3a, as assessed by immunohistochemistry and western blot analysis of growth-associated protein 43 and tyrosine hydroxylase. This was also confirmed by sympathetic nerve function changes assessed by cardiac norepinephrine content. Additionally, intra-LSG injection with AdSema3a alleviated MI-induced accumulation of dephosphorylated connexin 43 in the infarct border zone. Furthermore, Sema3a overexpression in the LSG reduced the incidence of inducible ventricular tachyarrhythmia by programmed electrical stimulation post-MI, and arrhythmia scores were significantly lower in the AdSema3a group than in the PBS and AdGFP groups. CONCLUSION: Semaphorin 3a overexpression in the LSG ameliorates the inducibility of ventricular arrhythmias after MI, mainly through attenuation of neural remodelling within the cardiac-neuraxis.


Asunto(s)
Infarto del Miocardio/complicaciones , Semaforina-3A/uso terapéutico , Ganglio Estrellado/metabolismo , Taquicardia Ventricular/terapia , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Electrocardiografía , Terapia Genética , Corazón/inervación , Masculino , Miocardio/metabolismo , Norepinefrina , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Semaforina-3A/genética , Ganglio Estrellado/efectos de los fármacos , Transfección
19.
J Cell Mol Med ; 19(3): 642-50, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25656605

RESUMEN

BACKGROUND: Phospholemman (PLM) is an important phosphorylation substrate for protein kinases A and C in the heart. Until now, the association between PLM phosphorylation status and L-type calcium channels (LTCCs) gating has not been fully understood. We investigated the kinetics of LTCCs in HEK 293T cells expressing phosphomimetic or nonphosphorylatable PLM mutants. METHODS: The LTCCs gating was measured in HEK 293T cells transfected with LTCC and wild-type (WT) PLM, phosphomimetic or nonphosphorylatable PLM mutants: 6263AA, 6869AA, AAAA, 6263DD, 6869DD or DDDD. RESULTS: WT PLM significantly slowed LTCCs activation and deactivation while enhanced voltage-dependent inactivation (VDI). PLM mutants 6869DD and DDDD significantly increased the peak of the currents. 6263DD accelerated channel activation, while 6263AA slowed it more than WT PLM. 6869DD significantly enhanced PLM-induced increase of VDI. AAAA slowed the channel activation more than 6263AA, and DDDD accelerated the channel VDI more than 6869DD. CONCLUSIONS: Our results demonstrate that phosphomimetic PLM could stimulate LTCCs and alter their dynamics, while PLM nonphosphorylatable mutant produced the opposite effects.


Asunto(s)
Canales de Calcio Tipo L/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/genética , Línea Celular , Células HEK293 , Humanos , Mutación/genética , Técnicas de Placa-Clamp
20.
Acta Pharmacol Sin ; 36(2): 229-40, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25544362

RESUMEN

AIM: Small GTPase Rac1 is a member of the Ras superfamily, which plays important roles in regulation of cytoskeleton reorganization, cell growth, proliferation, migration, etc. The aim of this study was to determine how a constitutively active Rac1b regulated cell proliferation and to investigate the effects of the Rac1b inhibitor sanguinarine. METHODS: Three HEK293T cell lines stably overexpressing GFP, Rac1-GFP or Rac1b-GFP were constructed by lentiviral infection. The cells were treated with sanguinarine (1 µmol/L) or its analogue berberine (1 µmol/L) for 4 d. Cell proliferation was evaluated by counting cell numbers and with a BrdU incorporation assay. The levels of cleaved PARP-89 (an apoptosis marker) and cyclin-D1 (a proliferative index) were measured using Western blotting. RESULTS: In 10% serum-containing media, overexpressing either Rac1 or Rac1b did not significantly change the cell proliferation. In the serum-starved media, however, the survival rate of Rac1b cells was significantly increased, whereas that of Rac1 cells was moderately increased. The level of cleaved PARP-89 was significantly increased in serum-starved Rac1 cells, but markedly reduced in serum-starved Rac1b cells. The level of cyclin-D1 was significantly increased in both serum-starved Rac1 and Rac1b cells. Treatment with sanguinarine, but not berberine, inhibited the proliferation of Rac1b cells, which was accompanied by significantly increased the level of PARP-89, and decreased both the level of cyclin-D1 and the percentage of BrdU positive cells. CONCLUSION: Rac1b enhances the cell proliferation under a growth-limiting condition via both anti-apoptotic and pro-proliferative mechanisms. Sanguinarine, as the specific inhibitor of Rac1b, is a potential therapeutic agent for malignant tumors with up-regulated Rac1b.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzofenantridinas/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Isoquinolinas/farmacología , Proteína de Unión al GTP rac1/metabolismo , Línea Celular , Células HEK293 , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA