Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Heliyon ; 9(6): e17434, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37426799

RESUMEN

Aims: Type 1 diabetes mellitus (T1DM) is associated with increased risk of cardiovascular disease (CVD) and mortality. The underlying mechanisms for T1DM-induced heart disease still remains unclear. In this study, we aimed to investigate the effects of cardiac non-neuronal cholinergic system (cNNCS) activation on T1DM-induced cardiac remodelling. Methods: T1DM was induced in C57Bl6 mice using low-dose streptozotocin. Western blot analysis was used to measure the expression of cNNCS components at different time points (4, 8, 12, and 16 weeks after T1DM induction). To assess the potential benefits of cNNCS activation, T1DM was induced in mice with cardiomyocyte-specific overexpression of choline acetyltransferase (ChAT), the enzyme required for acetylcholine (Ac) synthesis. We evaluated the effects of ChAT overexpression on cNNCS components, vascular and cardiac remodelling, and cardiac function. Key findings: Western blot analysis revealed dysregulation of cNNCS components in hearts of T1DM mice. Intracardiac ACh levels were also reduced in T1DM. Activation of ChAT significantly increased intracardiac ACh levels and prevented diabetes-induced dysregulation of cNNCS components. This was associated with preserved microvessel density, reduced apoptosis and fibrosis, and improved cardiac function. Significance: Our study suggests that cNNCS dysregulation may contribute to T1DM-induced cardiac remodelling, and that increasing ACh levels may be a potential therapeutic strategy to prevent or delay T1DM-induced heart disease.

3.
Cardiovasc Diabetol ; 20(1): 50, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33618724

RESUMEN

BACKGROUND: Acetylcholine (ACh) plays a crucial role in the function of the heart. Recent evidence suggests that cardiomyocytes possess a non-neuronal cholinergic system (NNCS) that comprises of choline acetyltransferase (ChAT), choline transporter 1 (CHT1), vesicular acetylcholine transporter (VAChT), acetylcholinesterase (AChE) and type-2 muscarinic ACh receptors (M2AChR) to synthesize, release, degrade ACh as well as for ACh to transduce a signal. NNCS is linked to cardiac cell survival, angiogenesis and glucose metabolism. Impairment of these functions are hallmarks of diabetic heart disease (DHD). The role of the NNCS in DHD is unknown. The aim of this study was to examine the effect of diabetes on cardiac NNCS and determine if activation of cardiac NNCS is beneficial to the diabetic heart. METHODS: Ventricular samples from type-2 diabetic humans and db/db mice were used to measure the expression pattern of NNCS components (ChAT, CHT1, VAChT, AChE and M2AChR) and glucose transporter-4 (GLUT-4) by western blot analysis. To determine the function of the cardiac NNCS in the diabetic heart, a db/db mouse model with cardiac-specific overexpression of ChAT gene was generated (db/db-ChAT-tg). Animals were followed up serially and samples collected at different time points for molecular and histological analysis of cardiac NNCS components and prosurvival and proangiogenic signaling pathways. RESULTS: Immunoblot analysis revealed alterations in the components of cardiac NNCS and GLUT-4 in the type-2 diabetic human and db/db mouse hearts. Interestingly, the dysregulation of cardiac NNCS was followed by the downregulation of GLUT-4 in the db/db mouse heart. Db/db-ChAT-tg mice exhibited preserved cardiac and vascular function in comparison to db/db mice. The improved function was associated with increased cardiac ACh and glucose content, sustained angiogenesis and reduced fibrosis. These beneficial effects were associated with upregulation of the PI3K/Akt/HIF1α signaling pathway, and increased expression of its downstream targets-GLUT-4 and VEGF-A. CONCLUSION: We provide the first evidence for dysregulation of the cardiac NNCS in DHD. Increased cardiac ACh is beneficial and a potential new therapeutic strategy to prevent or delay the development of DHD.


Asunto(s)
Acetilcolina/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Cardiomiopatías Diabéticas/prevención & control , Glucosa/metabolismo , Ventrículos Cardíacos/metabolismo , Acetilcolinesterasa/metabolismo , Anciano , Animales , Estudios de Casos y Controles , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Femenino , Proteínas Ligadas a GPI/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Masculino , Proteínas de Transporte de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor Muscarínico M2/metabolismo , Simportadores/metabolismo , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
4.
J Clin Endocrinol Metab ; 104(4): 1239-1248, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30383229

RESUMEN

CONTEXT: Obesity is a global epidemic and an independent risk factor for several diseases. miRNAs are gaining interest as early molecular regulators of various pathological processes. OBJECTIVE: To examine the miRNA signatures in women who are obese and determine the response of miRNAs to acute weight loss. METHODS: Plasma samples were collected from women who are obese (n = 80) before and after acute weight loss (mean, 7.2%). Plasma samples from age-matched lean volunteers (n = 80) were used as controls. Total RNA was extracted from the plasma samples and subjected to NanoString analysis of 822 miRNAs. The expression level of candidate miRNAs was validated in all participants using quantitative real-time PCR analysis. RESULTS: NanoString analysis identified substantial dysregulation of 21 miRNAs in women who are obese that were associated with impaired glucose tolerance, senescence, cardiac hypertrophy, angiogenesis, inflammation, and cell death. Acute weight loss reversed the expression pattern of 18 of these miRNAs toward those seen in the lean control group. Furthermore, real-time PCR validation of all the samples for 13 miRNAs with at least twofold upregulation or downregulation confirmed substantial dysregulation of all the chosen miRNAs in women who are obese at baseline. After acute weight loss, the levels of seven miRNAs in women who are obese and who are lean were comparable, with no statistically significant evidence for differences between the two groups. CONCLUSIONS: Our study has provided evidence that the circulating miRNAs associated with various disorders are dysregulated in women who are obese. We also found that seven of these miRNAs showed levels comparable to those in lean controls after acute weight loss in women who are obese.


Asunto(s)
MicroARN Circulante/sangre , Obesidad/terapia , Pérdida de Peso , Programas de Reducción de Peso , Adulto , Anciano , Biomarcadores/sangre , MicroARN Circulante/aislamiento & purificación , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Humanos , Persona de Mediana Edad , Nueva Zelanda , Obesidad/sangre , Obesidad/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Resultado del Tratamiento , Regulación hacia Arriba
5.
Cell Death Differ ; 25(7): 1336-1349, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29302057

RESUMEN

Increased apoptosis and premature cellular ageing of the diabetic heart underpin the development of diabetic heart disease. The molecular mechanisms underlying these pathologies are still unclear. Here we determined the role of pro-senescence microRNA (miR)-34a in accelerating the ageing of the diabetic heart. RT-PCR analysis showed a significant increase in the level of circulating miR-34a from early stages in asymptomatic type-2 diabetic individuals compared to non-diabetic controls. We also observed significant upregulation of miR-34a in the type-2 human diabetic heart suggesting circulating miR-34a may be cardiac in origin. Moreover, western blot analysis identified marked downregulation of the pro-survival protein sirtuin 1 (SIRT1), a direct target of miR-34a. Analysis of cultured human adult cardiomyocytes exposed to high glucose and cardiac progenitor cells (CPCs) isolated from the diabetic heart confirmed significant upregulation of miR-34a and downregulation of SIRT1, associated with a marked increase in pro-apoptotic caspase-3/7 activity. Although therapeutic inhibition of miR-34a activity restored SIRT1 expression in both cardiomyocytes and CPCs, p53 expression was further upregulated in cardiomyocytes but conversely downregulated in CPCs. In spite of increased p53, miR-34a inhibition significantly reduced high glucose induced apoptotic cell death in cardiomyocytes. However, this effect was not observed in CPCs, which in fact showed reduced proliferation following miR-34a inhibition. Taken together, our results demonstrate upregulation of miR-34a in the diabetic heart and in the circulation from an early stage of the disease. However, inhibition of miR-34a activity has differential effects depending on the cell type, thereby warranting the need to eliminate off-target effects when introducing miR-based therapy.


Asunto(s)
Senescencia Celular , Diabetes Mellitus Tipo 2/metabolismo , MicroARNs/biosíntesis , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre/metabolismo , Anciano , Anciano de 80 o más Años , Apoptosis , Diabetes Mellitus Tipo 2/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miocardio/patología , Miocitos Cardíacos/patología , Sirtuina 1/biosíntesis , Células Madre/patología
6.
J Orthop Sports Phys Ther ; 47(9): 617-627, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28704625

RESUMEN

Study Design Controlled laboratory study. Background Spinal manipulation (SM) can trigger a cascade of responses involving multiple systems, including the sympathetic nervous system and the endocrine system, specifically, the hypothalamic-pituitary axis. However, no manual therapy study has investigated the neuroendocrine response to SM (ie, sympathetic nervous system-hypothalamic-pituitary axis) in the same trial. Objective To determine short-term changes in sympathetic nervous system activity, heart rate variability, and endocrine activity (cortisol, testosterone, and testosterone-cortisol [T/C] ratio) following a thoracic SM. Methods Twenty-four healthy men aged between 18 and 45 years were randomized into 2 groups: thoracic SM (n = 12) and sham (n = 12). Outcome measures were salivary cortisol (micrograms per deciliter), salivary testosterone (picograms per milliliter), T/C ratio, heart rate variability, and changes in oxyhemoglobin concentration of the right calf muscle (micromoles per liter). Measurements were done before and at 5 minutes, 30 minutes, and approximately 6 hours after intervention. Results A statistically significant group-by-time interaction was noted for T/C ratio (P<.05) and salivary cortisol (P<.01) concentrations. Significant between-group differences were noted for salivary cortisol concentration at 5 minutes (mean difference, 0.35; 95% confidence interval: 0.12, 0.6; interaction: P<.01) and for T/C ratio at 6 hours postintervention (mean difference, -0.09; 95% confidence interval: -0.16, -0.04; P = .02). However, SM did not differentially alter oxyhemoglobin, testosterone, or heart rate variability relative to responses in the sham group. Conclusion Thoracic SM resulted in an immediate decrease in salivary cortisol concentration and reduced T/C ratio 6 hours after intervention. A pattern of immediate sympathetic excitation was also observed in the SM group. J Orthop Sports Phys Ther 2017;47(9):617-627. Epub 13 Jul 2017. doi:10.2519/jospt.2017.7348.


Asunto(s)
Sistema Hipotálamo-Hipofisario/fisiología , Manipulación Espinal/métodos , Sistema Nervioso Simpático/fisiología , Adulto , Frecuencia Cardíaca/fisiología , Humanos , Hidrocortisona/metabolismo , Masculino , Oxihemoglobinas/metabolismo , Saliva/metabolismo , Testosterona/metabolismo , Vértebras Torácicas
7.
Clin Sci (Lond) ; 131(9): 847-863, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28289072

RESUMEN

Aim: Myocardial fibrosis is a well-established cause of increased myocardial stiffness and subsequent diastolic dysfunction in the diabetic heart. The molecular regulators that drive the process of fibrotic events in the diabetic heart are still unknown. We determined the role of the microRNA (miR)-15 family in fibrotic remodelling of the diabetic heart.Methods and results: Right atrial appendage (RAA) and left ventricular (LV) biopsy tissues collected from diabetic and non-diabetic (ND) patients undergoing coronary artery bypass graft surgery showed significant down-regulation of miR-15a and -15b. This was associated with marked up-regulation of pro-fibrotic transforming growth factor-ß receptor-1 (TGFßR1) and connective tissue growth factor (CTGF), direct targets for miR-15a/b and pro-senescence p53 protein. Interestingly, down-regulation of miR-15a/b preceded the development of diastolic dysfunction and fibrosis in Type 2 diabetic mouse heart. Therapeutic restoration of miR-15a and -15b in HL-1 cardiomyocytes reduced the activation of pro-fibrotic TGFßR1 and CTGF, and the pro-senescence p53 protein expression, confirming a causal regulation of these fibrotic and senescence mediators by miR-15a/b. Moreover, conditioned medium (CM) collected from cardiomyocytes treated with miR-15a/b markedly diminished the differentiation of diabetic human cardiac fibroblasts.Conclusion: Our results provide first evidence that early down-regulation of miR-15a/b activates fibrotic signalling in diabetic heart, and hence could be a potential target for the treatment/prevention of diabetes-induced fibrotic remodelling of the heart.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Regulación hacia Abajo , MicroARNs/genética , Miocardio/metabolismo , Animales , Western Blotting , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Fibrosis/genética , Fibrosis/metabolismo , Glucosa/farmacología , Humanos , Ratones , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miofibroblastos/citología , Miofibroblastos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Cardiovasc Res ; 113(1): 90-101, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28065883

RESUMEN

AIM: Microangiopathy due to endothelial dysfunction is a major contributing factor to the development of diabetes-induced cardiovascular disease (CVD). Dysregulation of endothelial-specific microRNAs (miRs) is correlated with impaired angiogenesis and cell survival. We investigated the profile of two angiomiRs, miR-126, and miR-132, in the plasma of type 2 diabetic individuals without any known history of CVD as well as in the cardiac tissues collected from diabetics undergoing cardiac surgery. METHODS AND RESULTS: The presence of diabetes alone significantly decreased both angiomiRs in the plasma and the myocardium. The down-regulation of angiomiRs was also associated with reduced capillaries and arterioles and increased endothelial cell apoptosis, the hallmark of microangiopathy. Importantly, a time course study in a type 2 diabetic mouse model confirmed that the down-regulation of angiomiRs preceded endothelial apoptosis as well as alterations in the density of the microvasculature. Finally, therapeutic overexpression of both angiomiRs in diabetic aortic rings and human umbilical vein endothelial cells exposed to high glucose (HG) abrogated the deleterious effects of diabetes and HG on cell survival and proliferation and restored their angiogenic potential. CONCLUSIONS: These novel findings demonstrate that the down-regulation of angiomiRs is a major underlying mechanism for the development of microangiopathy in diabetic hearts. Therefore, therapeutic restoration of angiomiRs could become a potential approach to combat the cardiovascular complications of diabetes.


Asunto(s)
Enfermedad de la Arteria Coronaria/metabolismo , Vasos Coronarios/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Angiopatías Diabéticas/metabolismo , MicroARNs/metabolismo , Animales , Apoptosis , Enfermedad de la Arteria Coronaria/etiología , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/patología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Angiopatías Diabéticas/etiología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Ratones Endogámicos C57BL , MicroARNs/sangre , MicroARNs/genética , Miocardio/metabolismo , Neovascularización Fisiológica , Transducción de Señal , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección
10.
Int J Cardiol ; 202: 13-20, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26386349

RESUMEN

BACKGROUND: Diabetes promotes progressive loss of cardiac cells, which are replaced by a fibrotic matrix, resulting in the loss of cardiac function. In the current study we sought to identify if excessive autophagy plays a major role in inducing this progressive loss. METHODS AND RESULTS: Immunofluorescence and western blotting analysis of the right atrial appendages collected from diabetic and non-diabetic patients undergoing coronary artery bypass graft surgery showed a marked increase in the level of autophagy in the diabetic heart, as evidenced by increased expression of autophagy marker LC3B-II and its mediator Beclin-1 and decreased expression of p62, which incorporates into autophagosomes to be efficiently degraded. Moreover, a marked activation of pro-apoptotic caspase-3 was observed. Electron microscopy showed increased autophagosomes in the diabetic heart. In vivo measurement of autophagic flux by choloroquine injection resulted in further enhancement of LC3B-II in the diabetic myocardium, confirming increased autophagic activity in the type-2 diabetic heart. Importantly, in-vitro genetic depletion of beclin-1 in high glucose treated adult rat cardiomyocytes markedly inhibited the level of autophagy and subsequent apoptotic cell death. CONCLUSIONS: These findings demonstrate the pathological role of autophagy in the type-2 diabetic heart, opening up a potentially novel therapeutic avenue for the treatment of diabetic heart disease.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Diabetes Mellitus Tipo 2/genética , Cardiomiopatías Diabéticas/genética , Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Miocardio/metabolismo , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/biosíntesis , Autofagia/genética , Beclina-1 , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Femenino , Humanos , Etiquetado Corte-Fin in Situ , Masculino , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Obesos , Microscopía Electrónica , Miocardio/ultraestructura , ARN/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Zucker , Transducción de Señal/genética
11.
Endocrinology ; 157(2): 432-45, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26672806

RESUMEN

Current therapeutic strategies for the treatment of critical limb ischemia (CLI) have only limited success. Recent in vitro evidence in the literature, using cell lines, proposes that the peptide hormone ghrelin may have angiogenic properties. In this study, we aim to investigate if ghrelin could promote postischemic angiogenesis in a mouse model of CLI and, further, identify the mechanistic pathway(s) that underpin ghrelin's proangiogenic properties. CLI was induced in male CD1 mice by femoral artery ligation. Animals were then randomized to receive either vehicle or acylated ghrelin (150 µg/kg sc) for 14 consecutive days. Subsequently, synchrotron radiation microangiography was used to assess hindlimb perfusion. Subsequent tissue samples were collected for molecular and histological analysis. Ghrelin treatment markedly improved limb perfusion by promoting the generation of new capillaries and arterioles (internal diameter less than 50 µm) within the ischemic hindlimb that were both structurally and functionally normal; evident by robust endothelium-dependent vasodilatory responses to acetylcholine. Molecular analysis revealed that ghrelin's angiogenic properties were linked to activation of prosurvival Akt/vascular endothelial growth factor/Bcl-2 signaling cascade, thus reducing the apoptotic cell death and subsequent fibrosis. Further, ghrelin treatment activated proangiogenic (miR-126 and miR-132) and antifibrotic (miR-30a) microRNAs (miRs) while inhibiting antiangiogenic (miR-92a and miR-206) miRs. Importantly, in vitro knockdown of key proangiogenic miRs (miR-126 and miR-132) inhibited the angiogenic potential of ghrelin. These results therefore suggest that clinical use of ghrelin for the early treatment of CLI may be a promising and potent inducer of reparative vascularization through modulation of key molecular factors.


Asunto(s)
Ghrelina/uso terapéutico , Miembro Posterior/irrigación sanguínea , Isquemia/tratamiento farmacológico , MicroARNs/genética , Neovascularización Fisiológica/efectos de los fármacos , Flujo Sanguíneo Regional/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Ghrelina/administración & dosificación , Ghrelina/farmacología , Miembro Posterior/patología , Humanos , Masculino , Ratones , Ratones Endogámicos , MicroARNs/metabolismo , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Neovascularización Fisiológica/genética , Flujo Sanguíneo Regional/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Activación Transcripcional/efectos de los fármacos , Vasodilatación/efectos de los fármacos
12.
Data Brief ; 5: 269-75, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26543890

RESUMEN

This data article contains full list of autophagy related genes that are altered in diabetic heart. This article also shows data from in vitro cultured cardiomyocytes that are exposed the high glucose treatment to simulate hyperglycemic state in vitro. The interpretation of these data and further extensive insights into the regulation of SG biogenesis by AMPK can be found in "Type-2 diabetes increases autophagy in the human heart through promotion of Beclin-1 mediated pathway" (Munasinghe et al., in press) [1].

13.
Cardiovasc Res ; 97(1): 55-65, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22997160

RESUMEN

AIMS: Diabetes impinges upon mechanisms of cardiovascular repair. However, the biochemical adaptation of cardiac stem cells to sustained hyperglycaemia remains largely unknown. Here, we investigate the molecular targets of high glucose-induced damage in cardiac progenitor cells (CPCs) from murine and human hearts and attempt safeguarding CPC viability and function through reactivation of the pentose phosphate pathway. METHODS AND RESULTS: Type-1 diabetes was induced by streptozotocin. CPC abundance was determined by flow cytometry. Proliferating CPCs were identified in situ by immunostaining for the proliferation marker Ki67. Diabetic hearts showed marked reduction in CPC abundance and proliferation when compared with controls. Moreover, Sca-1(pos) CPCs isolated from hearts of diabetic mice displayed reduced activity of key enzymes of the pentose phosphate pathway, glucose-6-phosphate dehydrogenase (G6PD), and transketolase, increased levels of superoxide and advanced glucose end-products (AGE), and inhibition of the Akt/Pim-1/Bcl-2 signalling pathway. Similarly, culture of murine CPCs or human CD105(pos) progenitor cells in high glucose inhibits the pentose phosphate and pro-survival signalling pathways, leading to the activation of apoptosis. In vivo and in vitro supplementation with benfotiamine reactivates the pentose phosphate pathway and rescues CPC availability and function. This benefit is abrogated by either G6PD silencing by small interfering RNA (siRNA) or Akt inhibition by dominant-negative Akt. CONCLUSION: We provide new evidence of the negative impact of diabetes and high glucose on mechanisms controlling CPC redox state and survival. Boosting the pentose phosphate pathway might represent a novel mechanistic target for protection of CPC integrity.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Vía de Pentosa Fosfato/efectos de los fármacos , Células Madre/efectos de los fármacos , Tiamina/análogos & derivados , Animales , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Endoglina , Citometría de Flujo , Glucosafosfato Deshidrogenasa/genética , Glucosafosfato Deshidrogenasa/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Interferencia de ARN , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo , Células Madre/patología , Superóxidos/metabolismo , Tiamina/farmacología , Transfección , Transcetolasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA