Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443158

RESUMEN

The sinus node (SAN) is the primary pacemaker of the human heart, and abnormalities in its structure or function cause sick sinus syndrome, the most common reason for electronic pacemaker implantation. Here we report that transcription factor GATA6, whose mutations in humans are linked to arrhythmia, is highly expressed in the SAN and its haploinsufficiency in mice results in hypoplastic SANs and rhythm abnormalities. Cell-specific deletion reveals a requirement for GATA6 in various SAN lineages. Mechanistically, GATA6 directly activates key regulators of the SAN genetic program in conduction and nonconduction cells, such as TBX3 and EDN1, respectively. The data identify GATA6 as an important regulator of the SAN and provide a molecular basis for understanding the conduction abnormalities associated with GATA6 mutations in humans. They also suggest that GATA6 may be a potential modifier of the cardiac pacemaker.


Asunto(s)
Factor de Transcripción GATA6/metabolismo , Frecuencia Cardíaca/fisiología , Nodo Sinoatrial/embriología , Animales , Arritmias Cardíacas/fisiopatología , Diferenciación Celular/genética , Factor de Transcripción GATA6/genética , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis , Nodo Sinoatrial/fisiología , Proteínas de Dominio T Box/genética
2.
Pflugers Arch ; 471(11-12): 1551-1564, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31713764

RESUMEN

Hydrogen sulfide (H2S) is endogenously produced in pancreatic ß cells and its level is elevated in diabetes. Here, we report that H2S affects insulin secretion via two mechanisms that converge on cytosolic free Ca2+ ([Ca2+]i), a key mediator of insulin exocytosis. Cellular calcium imaging, using Fura-2 or Fluo-4, showed that exposure of INS-1E cells to H2S (30-100 µM) reduced both [Ca2+]i levels (by 21.7 ± 2.3%) and oscillation frequency (p < 0.01, n = 4). Consistent with a role of plasma membrane KATP channels (plasma-KATP), the effects of H2S on [Ca2+]i were blocked by gliclazide (a blocker of plasma-KATP channels), but were mimicked by diazoxide (an activator of plasma-KATP channels). Surprisingly, when Ca2+ entry via plasma membrane was inhibited using Ca2+-free external solutions, H2S increased [Ca2+]i by 39.7 ± 3.6% suggesting Ca2+ release from intracellular stores. H2S-induced [Ca2+]i increases were abolished by either FCCP (which depletes Ca2+ stored in mitochondria) or cyclosporine A (an inhibitor of mitochondrial permeability transition pore, mPTP) suggesting that H2S induces Ca2+ release from mitochondria. Measurement of mitochondrial membrane potential (MMP) suggested that H2S causes MMP depolarization, which was blocked by cyclosporine A. Finally, insulin measurements by ELISA indicated that H2S decreased insulin release from INS-1E cells, but after plasma membrane Ca2+ entry was blocked by nifedipine, H2S-induced mitochondrial Ca2+ release is able to increase insulin release. Together, our results indicate that H2S has dual effects on insulin release suggesting that, with different metabolic conditions, H2S may differentially modulate the insulin release from pancreatic ß cells and play a role in ß cell dysfunction.


Asunto(s)
Calcio/metabolismo , Citosol/metabolismo , Homeostasis/fisiología , Sulfuro de Hidrógeno/metabolismo , Secreción de Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Canales KATP/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Señalización del Calcio/fisiología , Línea Celular Tumoral , Membrana Celular/metabolismo , Exocitosis/fisiología , Insulina/metabolismo , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Ratas
3.
Eur J Neurosci ; 47(4): 305-313, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29363836

RESUMEN

Opioid-induced hyperalgesia (OIH) and allodynia is a well-known phenomenon and refers to the pain sensitization in patients after prolonged opioid exposure. OIH limits the use of opioids in pain control, but the underlying mechanisms are not fully clear. This study investigated the role of mitochondrial Ca2+ uniporter (MCU) in remifentanil (a commonly used opioid analgesic)-induced allodynia. Using a rat model of OIH, we found that incision- and remifentanil-induced mechanical allodynia were remarkably attenuated by pretreatment with Ru360, a specific MCU antagonist, suggesting a critical role of MCU in both incision- and opioid-induced allodynia. In addition, imaging studies with Rhod-2 (a mitochondrial Ca2+ dye) in spinal tissues demonstrated increased mitochondrial Ca2+ level in response to incision and remifentanil infusion, which was attenuated by Ru360. Western blot and immunohistochemistry showed that pNR [phosphorylated N-methyl-D-aspartate (NMDA) receptor] and pERK (phosphorylated extracellular signal-regulated kinase) are increased during both incision-induced hyperalgesia and remifentanil-induced hyperalgesia, and again the increases in pNR and pERK were remarkably attenuated by Ru360. Together, our data demonstrate that MCU plays a critical role in remifentanil-induced postoperative mechanical allodynia, with NMDA receptor and ERK as possible downstream effectors. Our findings provide novel mechanisms for remifentanil-induced mechanical allodynia and encourage future studies to examine the mitochondrial Ca2+ uniporter as a potential therapeutic target for prevention of OIH.


Asunto(s)
Calcio/metabolismo , Hiperalgesia/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Remifentanilo/farmacología , Analgésicos Opioides/farmacología , Animales , Masculino , Mitocondrias/metabolismo , Fosforilación , Piperidinas/farmacología , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
4.
J Neurosci Res ; 91(6): 786-98, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23553889

RESUMEN

Nerve cell injury associated with apoptosis plays an important role in the development of diabetic peripheral neuropathy (DPN). However, it remains unclear whether preexisting or potential neurocyte damage induced by hyperglycemia increases sensitivity to local anesthetics. SH-SY5Y cells were pretreated with a high concentration of glucose in vitro, to imitate DPN prior to administration of bupivacaine or placebo. Cell viability and apoptosis were investigated with a CCK-8 assay and flow cytometry, respectively. In addition, mitochondrial membrane potential, reactive oxygen species (ROS), mitochondrially generated ROS, and activity of mitochondrial complexes I and III were studied to explore the molecular mechanism of bupivacaine-induced mitochondrial injury. Grp78 and caspase-12 expression were measured by qRT-PCR and Western blot, representing endoplasmic reticulum (ER) stress. Cell structure was also assessed via transmission electron microscopy. Incubation with bupivacaine decreased the activity of mitochondrial complexes I and III; increased ROS production at cell and mitochondrial levels, mitochondrial potential depolarization, and Grp78 and caspase-12 expression at both transcription and translation levels; and affected cell structure, which could be enhanced by glucose pretreatment. These findings indicate that mitochondrial dysfunction and ER stress related to ROS are involved in bupivacaine-induced apoptosis and may be enhanced by glucose administration.


Asunto(s)
Anestésicos Locales/toxicidad , Bupivacaína/toxicidad , Estrés del Retículo Endoplásmico/fisiología , Hiperglucemia/complicaciones , Neuronas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Western Blotting , Supervivencia Celular/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Citometría de Flujo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Physiol Rep ; 11(10): e15696, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37226398

RESUMEN

The voltage-gated Nav 1.5 channels mediate the fast Na+ current (INa ) in cardiomyocytes initiating action potentials and cardiac contraction. Downregulation of INa , as occurs in Brugada syndrome (BrS), causes ventricular arrhythmias. The present study investigated whether the Wnt/ß-catenin signaling regulates Nav 1.5 in human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). In healthy male and female iPSC-CMs, activation of Wnt/ß-catenin signaling by CHIR-99021 reduced (p < 0.01) both Nav 1.5 protein and SCN5A mRNA. In iPSC-CMs from a BrS patient, both Nav 1.5 protein and peak INa were reduced compared to those in healthy iPSC-CMs. Treatment of BrS iPSC-CMs with Wnt-C59, a small-molecule Wnt inhibitor, led to a 2.1-fold increase in Nav 1.5 protein (p = 0.0005) but surprisingly did not affect SCN5A mRNA (p = 0.146). Similarly, inhibition of Wnt signaling using shRNA-mediated ß-catenin knockdown in BrS iPSC-CMs led to a 4.0-fold increase in Nav 1.5, which was associated with a 4.9-fold increase in peak INa but only a 2.1-fold increase in SCN5A mRNA. The upregulation of Nav 1.5 by ß-catenin knockdown was verified in iPSC-CMs from a second BrS patient. This study demonstrated that Wnt/ß-catenin signaling inhibits Nav 1.5 expression in both male and female human iPSC-CMs, and inhibition of Wnt/ß-catenin signaling upregulates Nav 1.5 in BrS iPSC-CMs through both transcriptional and posttranscriptional mechanisms.


Asunto(s)
Síndrome de Brugada , Células Madre Pluripotentes Inducidas , Canal de Sodio Activado por Voltaje NAV1.5 , Vía de Señalización Wnt , Femenino , Humanos , Masculino , beta Catenina , Miocitos Cardíacos , Canal de Sodio Activado por Voltaje NAV1.5/genética
6.
J Vis Exp ; (185)2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35969083

RESUMEN

Heart disease is the leading cause of morbidity and mortality worldwide. Due to their low cost, ease of handling, and abundance of transgenic strains, rodents have become essential models for cardiovascular research. However, spontaneous lethal cardiac arrhythmias that often cause mortality in heart disease patients are rare in rodent models of heart disease. This is primarily due to the species differences in cardiac electrical properties between human and rodents and poses a challenge to the study of cardiac arrhythmias using rodents. This protocol describes an approach to enable efficient transgene expression in mouse and rat ventricular myocardium using echocardiography-guided intramuscular injections of recombinant virus (adenovirus and adeno-associated virus). This work also outlines a method to enable reliable assessment of cardiac susceptibility to arrhythmias using isolated, Langendorff-perfused mouse and rat hearts with both adrenergic and programmed electrical stimulations. These techniques are critical for studying heart rhythm disorders associated with adverse cardiac remodeling after injuries, such as myocardial infarction.


Asunto(s)
Arritmias Cardíacas , Transgenes , Animales , Animales Modificados Genéticamente , Arritmias Cardíacas/fisiopatología , Modelos Animales de Enfermedad , Corazón , Humanos , Ratones , Miocardio/metabolismo , Ratas
7.
Sci Rep ; 11(1): 17722, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34489488

RESUMEN

Wnt/ß-catenin signaling is activated in the heart after myocardial infarction (MI). This study aims to investigate if ß-catenin deletion affects post-MI ion channel gene alterations and ventricular tachycardias (VT). MI was induced by permanent ligation of left anterior descending artery in wild-type (WT) and cardiomyocyte-specific ß-catenin knockout (KO) mice. KO mice showed reduced susceptibility to VT (18% vs. 77% in WT) at 8 weeks after MI, associated with reduced scar size and attenuated chamber dilation. qPCR analyses of both myocardial tissues and purified cardiomyocytes demonstrated upregulation of Wnt pathway genes in border and infarct regions after MI, including Wnt ligands (such as Wnt4) and receptors (such as Fzd1 and Fzd2). At 1 week after MI, cardiac sodium channel gene (Scn5a) transcript was reduced in WT but not in KO hearts, consistent with previous studies showing Scn5a inhibition by Wnt/ß-catenin signaling. At 8 weeks after MI when Wnt genes have declined, Scn5a returned to near sham levels and K+ channel gene downregulations were not different between WT and KO mice. This study demonstrated that VT susceptibility in the chronic phase after MI is reduced in mice with cardiomyocyte-specific ß-catenin deletion primarily through attenuated structural remodeling, but not ion channel gene alterations.


Asunto(s)
Arritmias Cardíacas/metabolismo , Corazón/fisiopatología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , beta Catenina/metabolismo , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Remodelación Ventricular/fisiología , beta Catenina/genética
8.
Can J Cardiol ; 36(4): 564-576, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32046907

RESUMEN

BACKGROUND: Myocardial infarction and heart failure are associated with reduced voltage-gated Na+ current (INa) that promotes arrhythmias and sudden deaths. We have previously shown that the Wnt/ß-catenin signalling (Wnt signalling), which is active in heart disease, reduces cardiac INa, suggesting that Wnt signalling may be a potential therapeutic target. However, because Wnt signalling is required for the homeostasis of many noncardiac tissues, administration of Wnt inhibitors to heart patients would cause significant side effects. The present study aims to elucidate the molecular mechanisms of cardiac INa inhibition by Wnt, which would identify cardiac-specific therapeutic targets. METHODS: Wnt signalling was activated in neonatal rat ventricular myocytes by Wnt3a protein. Adenovirus expressing Wnt3a was injected into the adult rat ventricle. CRISPR/Cas9 and chromatin immunoprecipitation were used for mechanistic studies. RESULTS: Wnt signalling activation in neonatal rat ventricular myocytes reduced Nav1.5 protein and Scn5a mRNA, but increased Tbx3, a known suppressor of Scn5a. Chromatin immunoprecipitation showed that Wnt signalling inhibits Scn5a expression through downstream mediator (TCF4) binding to both Tbx3 and Scn5a promoters. Overexpression or knockdown of Tbx3 directly modified Nav1.5 and INa, whereas CRISPR/Cas9-induced mutations at TCF4 binding sites within the Scn5a promoter attenuated Wnt inhibition of Scn5a and Nav1.5. In adult rat hearts, adenovirus expressing Wnt3a reduced Nav1.5, increased QRS duration in electrocardiogram, and increased the susceptibility to ventricular tachycardia. CONCLUSIONS: Wnt signalling inhibits the Na+ channel by direct and indirect (via Tbx3) suppression of Scn5a transcription. Strategies to block TCF4 binding to the Tbx3 and Scn5a promoters would represent novel strategies for cardiac-specific inhibition of the Wnt pathway to rescue INa and prevent sudden cardiac deaths.


Asunto(s)
Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canales de Sodio/fisiología , Vía de Señalización Wnt , Animales , Regulación de la Expresión Génica , Ventrículos Cardíacos/citología , Ratas
9.
Biomaterials ; 247: 120010, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32259654

RESUMEN

While encapsulation of cells within protective nanoporous gel cocoons increases cell retention and pro-survival integrin signaling, the influence of cocoon size and intra-capsular cell-cell interactions on therapeutic repair are unknown. Here, we employ a microfluidic platform to dissect the impact of cocoon size and intracapsular cell number on the regenerative potential of transplanted heart explant-derived cells. Deterministic increases in cocoon size boosted the proportion of multicellular aggregates within cocoons, reduced vascular clearance of transplanted cells and enhanced stimulation of endogenous repair. The latter being attributable to cell-cell stimulation of cytokine and extracellular vesicle production while also broadening of the miRNA cargo within extracellular vesicles. Thus, by tuning cocoon size and cell occupancy, the paracrine signature and retention of transplanted cells can be enhanced to promote paracrine stimulation of endogenous tissue repair.


Asunto(s)
Vesículas Extracelulares , Infarto del Miocardio , Corazón , Humanos , Microfluídica , Miocardio , Comunicación Paracrina
10.
Stem Cells Int ; 2017: 4528941, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28303153

RESUMEN

The advent of cellular reprogramming technology has revolutionized biomedical research. De novo human cardiac myocytes can now be obtained from direct reprogramming of somatic cells (such as fibroblasts), from induced pluripotent stem cells (iPSCs, which are reprogrammed from somatic cells), and from human embryonic stem cells (hESCs). Such de novo human cardiac myocytes hold great promise for in vitro disease modeling and drug screening and in vivo cell therapy of heart disease. Here, we review the technique advancements for generating de novo human cardiac myocytes. We also discuss several challenges for the use of such cells in research and regenerative medicine, such as the immature phenotype and heterogeneity of de novo cardiac myocytes obtained with existing protocols. We focus on the recent advancements in addressing such challenges.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA