Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Nature ; 572(7769): E16-E17, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31363231

RESUMO

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Nature ; 556(7700): 239-243, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29618819

RESUMO

Cardiac tissues generated from human induced pluripotent stem cells (iPSCs) can serve as platforms for patient-specific studies of physiology and disease1-6. However, the predictive power of these models is presently limited by the immature state of the cells1, 2, 5, 6. Here we show that this fundamental limitation can be overcome if cardiac tissues are formed from early-stage iPSC-derived cardiomyocytes soon after the initiation of spontaneous contractions and are subjected to physical conditioning with increasing intensity over time. After only four weeks of culture, for all iPSC lines studied, such tissues displayed adult-like gene expression profiles, remarkably organized ultrastructure, physiological sarcomere length (2.2 µm) and density of mitochondria (30%), the presence of transverse tubules, oxidative metabolism, a positive force-frequency relationship and functional calcium handling. Electromechanical properties developed more slowly and did not achieve the stage of maturity seen in adult human myocardium. Tissue maturity was necessary for achieving physiological responses to isoproterenol and recapitulating pathological hypertrophy, supporting the utility of this tissue model for studies of cardiac development and disease.


Assuntos
Diferenciação Celular , Coração/crescimento & desenvolvimento , Células-Tronco Pluripotentes Induzidas/citologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Técnicas de Cultura de Tecidos , Adulto , Cálcio/metabolismo , Diferenciação Celular/genética , Metabolismo Energético/efeitos dos fármacos , Coração/efeitos dos fármacos , Humanos , Isoproterenol/farmacologia , Mitocôndrias/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Sarcômeros/metabolismo , Transcriptoma
3.
Circ J ; 85(5): 657-666, 2021 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-33716265

RESUMO

BACKGROUND: Although adipose-derived stem cell (ADSC) sheets improve the cardiac function after myocardial infarction (MI), underlying mechanisms remain to be elucidated. The aim of this study was to determine the fate of transplanted ADSC sheets and candidate angiogenic factors released from ADSCs for their cardiac protective actions.Methods and Results:MI was induced by ligation of the left anterior descending coronary artery. Sheets of transgenic (Tg)-ADSCs expressing green fluorescence protein (GFP) and luciferase or wild-type (WT)-ADSCs were transplanted 1 week after MI. Both WT- and Tg-ADSC sheets improved cardiac functions evaluated by echocardiography at 3 and 5 weeks after MI. Histological examination at 5 weeks after MI demonstrated that either sheet suppressed fibrosis and increased vasculogenesis. Luciferase signals from Tg-ADSC sheets were detected at 1 and 2 weeks, but not at 4 weeks, after transplantation. RNA sequencing of PKH (yellow-orange fluorescent dye with long aliphatic tails)-labeled Tg-ADSCs identified mRNAs of 4 molecules related to angiogenesis, including those of Esm1 and Stc1 that increased under hypoxia. Administration of Esm1 or Stc1 promoted tube formation by human umbilical vein endothelial cells. CONCLUSIONS: ADSC sheets improved cardiac contractile functions after MI by suppressing cardiac fibrosis and enhancing neovascularization. Transplanted ADSCs existed for >2 weeks on MI hearts and produced the angiogenic factors Esm1 and Stc1, which may improve cardiac functions after MI.


Assuntos
Tecido Adiposo , Insuficiência Cardíaca , Infarto do Miocárdio , Indutores da Angiogênese , Animais , Insuficiência Cardíaca/terapia , Células Endoteliais da Veia Umbilical Humana , Humanos , Infarto do Miocárdio/terapia , Ratos , Transplante de Células-Tronco
4.
J Mol Cell Cardiol ; 115: 158-169, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29355491

RESUMO

The human ether-a-go-go-related gene (hERG) encodes the α subunit of a rapidly activating delayed-rectifier potassium (IKr) channel. Mutations of the hERG cause long QT syndrome type 2 (LQT2). Acetylation of lysine residues occurs in a subset of non-histone proteins and this modification is controlled by both histone acetyltransferases and deacetylases (HDACs). The aim of this study was to clarify effects of HDAC(s) on wild-type (WT) and mutant hERG proteins. WThERG and two trafficking-defective mutants (G601S and R752W) were transiently expressed in HEK293 cells, which were treated with a pan-HDAC inhibitor Trichostatin A (TSA) or an isoform-selective HDAC6 inhibitor Tubastatin A (TBA). Both TSA and TBA increased protein levels of WThERG and induced expression of mature forms of the two mutants. Immunoprecipitation showed an interaction between HDAC6 and immature forms of hERG. Coexpression of HDAC6 decreased acetylation and, reciprocally, increased ubiquitination of hERG, resulting in its decreased expression. siRNA against HDAC6, as well as TBA, exerted opposite effects. Immunochemistry revealed that HDAC6 knockdown increased expression of the WThERG and two mutants both in the endoplasmic reticulum and on the cell surface. Electrophysiology showed that HDAC6 knockdown or TBA treatment increased the hERG channel current corresponding to the rapidly activating delayed-rectifier potassium current (IKr) in HEK293 cells stably expressing the WT or mutants. Three lysine residues (K116, K495 and K757) of hERG were predicted to be acetylated. Substitution of these lysine residues with arginine eliminated HDAC6 effects. In HL-1 mouse cardiomyocytes, TBA enhanced endogenous ERG expression, increased IKr, and shortened action potential duration. These results indicate that hERG is a substrate of HDAC6. HDAC6 inhibition induced acetylation of hERG which counteracted ubiquitination leading its stabilization. HDAC6 inhibition may be a novel therapeutic option for LQT2.


Assuntos
Canal de Potássio ERG1/metabolismo , Desacetilase 6 de Histona/metabolismo , Proteínas Mutantes/metabolismo , Acetilação/efeitos dos fármacos , Animais , Canal de Potássio ERG1/química , Células HEK293 , Inibidores de Histona Desacetilases/farmacologia , Humanos , Lisina/metabolismo , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos
5.
Biochem Biophys Res Commun ; 495(1): 1278-1284, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29175323

RESUMO

Human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) are a promising source for cell transplantation into the damaged heart, which has limited regenerative ability. Many methods have been developed to obtain large amounts of functional CMs from hPSCs for therapeutic applications. However, during the differentiation process, a mixed population of various cardiac cells, including ventricular, atrial, and pacemaker cells, is generated, which hampers the proper functional analysis and evaluation of cell properties. Here, we established NKX2-5eGFP/w and MLC2vmCherry/w hPSC double knock-ins that allow for labeling, tracing, purification, and analysis of the development of ventricular cells from early to late stages. As with the endogenous transcriptional activities of these genes, MLC2v-mCherry expression following NKX2-5-eGFP expression was observed under previously established culture conditions, which mimic the in vivo cardiac developmental process. Patch-clamp and microelectrode array electrophysiological analyses showed that the NKX2-5 and MLC2v double-positive cells possess ventricular-like properties. The results demonstrate that the NKX2-5eGFP/w and MLC2vmCherry/w hPSCs provide a powerful model system to capture region-specific cardiac differentiation from early to late stages. Our study would facilitate subtype-specific cardiac development and functional analysis using the hPSC-derived sources.


Assuntos
Técnicas de Cultura Celular por Lotes/métodos , Miosinas Cardíacas/metabolismo , Rastreamento de Células/métodos , Ventrículos do Coração/citologia , Proteína Homeobox Nkx-2.5/metabolismo , Miócitos Cardíacos/citologia , Cadeias Leves de Miosina/metabolismo , Células-Tronco Pluripotentes/citologia , Miosinas Cardíacas/genética , Diferenciação Celular/fisiologia , Separação Celular/métodos , Células Cultivadas , Técnicas de Introdução de Genes , Genes Reporter/genética , Ventrículos do Coração/metabolismo , Proteína Homeobox Nkx-2.5/genética , Humanos , Miócitos Cardíacos/metabolismo , Cadeias Leves de Miosina/genética , Células-Tronco Pluripotentes/metabolismo , Engenharia Tecidual/métodos
6.
Circ J ; 79(12): 2659-68, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26477273

RESUMO

BACKGROUND: Hyperuricemia induces endothelial dysfunction, oxidative stress and inflammation, increasing cardiovascular morbidities. It also raises the incidence of atrial fibrillation; however, underlying mechanisms are unknown. METHODS AND RESULTS: The effects of urate on expression of Kv1.5 in cultured mouse atrial myocytes (HL-1 cells) using reverse transcriptase-PCR, immunoblots, flow cytometry and patch-clamp experiments were studied. Treatment with urate at 7 mg/dl for 24 h increased the Kv1.5 protein level, enhanced ultra-rapid delayed-rectifier K(+)channel currents and shortened action potential duration in HL-1 cells. HL-1 cells expressed the influx uric acid transporter (UAT), URATv1, and the efflux UATs, ABCG2 and MRP4. An inhibitor against URATv1, benzbromarone, abolished the urate effects, whereas an inhibitor against ABCG2, KO143, augmented them. Flow cytometry showed that urate induced an increase in reactive oxygen species, which was abolished by the antioxidant, N-acetylcysteine (NAC), and the NADPH-oxidase inhibitor, apocynin. Both NAC and apocynin abolished the enhancing effects of urate on Kv1.5 expression. A urate-induced increase in the Kv1.5 proteins was accompanied by phosphorylation of extracellular signal-regulated kinase (ERK), and was abolished by an ERK inhibitor, PD98059. NAC abolished phosphorylation of ERK by urate. CONCLUSIONS: Intracellular urate taken up by UATs enhanced Kv1.5 protein expression and function in HL-1 atrial myocytes, which could be attributable to ERK phosphorylation and oxidative stress derived from nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Hiperuricemia/metabolismo , Canal de Potássio Kv1.5/biossíntese , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Ácido Úrico/farmacologia , Animais , Linhagem Celular , Átrios do Coração/metabolismo , Átrios do Coração/patologia , Hiperuricemia/patologia , Canal de Potássio Kv1.5/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Miócitos Cardíacos/patologia , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos
7.
Pacing Clin Electrophysiol ; 37(7): 853-63, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24499369

RESUMO

BACKGROUND: A KCNE1 polymorphism, D85N, causes long QT syndrome (LQTS) with a decrease in the slowly activating delayed-rectifier K(+) channel current (IKs ). We examined impacts of D85N polymorphism on KCNE1 protein stability and functions, and tested the ability of various drugs to modify them. METHODS: KCNE1-D85N or the wild-type protein was coexpressed in COS7 cells with KCNQ1 to form K(+) channels. Expression, degradation, and intracellular localization of KCNE1 proteins, as well as the currents conferred by KCNQ1/KCNE1 complexes, were determined using immunoblots, immunofluorescence, and patch-clamp techniques. RESULTS: The protein level of KCNE1-D85N was lower than that of the wild-type, in spite of the comparable levels of their mRNA. KCNE1-D85N was highly ubiquitinated and rapidly degraded as compared to the wild-type; a proteasome inhibitor, MG132, inhibited its degradation and increased its steady-state level. Both KCNE1-D85N and the wild-type proteins were co-immunoprecipitated with KCNQ1. Immunofluorescent signals of KCNE1-D85N accumulated in the endoplasmic reticulum and Golgi apparatus, with reduced levels on the cell membrane. Patch-clamp experiments demonstrated that the membrane current corresponding to IKs was much smaller in cells expressing KCNE1-D85N than in those expressing the wild-type. Verapamil (0.5-10 µM) increased the protein level of KCNE1-D85N, decreased its ubiquitination, slowed its degradation, and enhanced KCNQ1/KCNE1-D85N channel currents. Pretreatment with amiodarone abolished these effects of verapamil. CONCLUSION: KCNE1-D85N is less stable than the wild-type protein, and is rapidly degraded through the ubiquitin-proteasome system. Verapamil may be of a therapeutic value in LQTS patients via preventing degradation of KCNE1-D85N.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/genética , Polimorfismo Genético , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Verapamil/farmacologia , Verapamil/uso terapêutico , Células Cultivadas , Humanos
8.
J Radiat Res ; 65(2): 205-214, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38330507

RESUMO

Recent in vitro experiments showed that combined treatment with MHY1485, a low-molecular-weight compound, and X-ray irradiation significantly increased apoptosis and senescence in tumor cells, which was associated with oxidative stress, endoplasmic reticulum (ER) stress and p21 stabilization, compared to radiation treatment alone. However, evidence for MHY1485 treatment-mediated suppression of tumor growth in animals is still lacking. Furthermore, it has been shown that ER stress enhances immunogenic cell death (ICD) in tumor cells, as it can exert a favorable influence on the anti-cancer immune system. In the present study, we examined whether co-treatment of MHY1485 and X-ray irradiation induces ICD and in vivo tumor growth suppression using the CT26 and Lewis lung carcinoma murine tumor cell lines. We found that MHY1485 + X-ray treatment promotes ICD more effectively than X-ray treatment alone. MHY1485 suppresses tumor growth in vivo under co-treatment with X-rays and increases INF-γ, tumor necrosis factor, interleukin-2 and interleukin-12 levels in the spleen as well as the presence of CD8+ cells in the tumor. The results suggest that MHY1485 treatment leads to the conversion of irradiated tumors into effective vaccines. Thus, MHY1485 is a promising lead compound for use in combination with radiotherapy.


Assuntos
Carcinoma Pulmonar de Lewis , Morte Celular Imunogênica , Morfolinas , Triazinas , Animais , Camundongos , Carcinoma Pulmonar de Lewis/radioterapia , Carcinoma Pulmonar de Lewis/patologia , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral
9.
FEBS Open Bio ; 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39223831

RESUMO

Establishing a highly efficient photoactivatable Cre recombinase PA-Cre3.0 can allow spatiotemporal control of Cre recombinase activity. This technique may help to elucidate cell lineages, as well as facilitate gene and cell function analysis during development. This study examined the blue light-mediated optical regulation of Cre-loxP recombination using PA-Cre3.0 transgenic early mouse pre-implantation embryos. We found that inducing PA-Cre3.0 expression in the heterozygous state did not show detectable recombination activation with blue light. Conversely, in homozygous embryos, DNA recombination by PA-Cre3.0 was successfully induced by blue light and resulted in the activation of the red fluorescent protein reporter gene, while almost no leaks of Cre recombination activity were detected in embryos without light illumination. Thus, we characterize the conditions under which the PA-Cre3.0 system functions efficiently in early mouse embryos. These results are expected to provide a new optogenetic tool for certain biological studies, such as developmental process analysis and lineage tracing in early mouse embryos.

10.
Biochem Biophys Res Commun ; 431(2): 225-31, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23313480

RESUMO

P19 embryonal carcinoma (EC) cells are pluripotent stem cells and have numerous morphological and biochemical properties in common with embryonic stem (ES) cells. However, P19 cells differentiate very ineffectively as embryoid bodies (EBs) without the specific chemical inducers whereas ES cells exhibit spontaneous differentiation to the three germ layers. Recently the heterochromatin protein 1 (HP1) family protein HP1γ, which is an epigenetic modulator that binds histone H3 methylated at lysine 9, is shown to be associated with the progression from pluripotent to differentiated status in ES cells. Therefore, to study the role of HP1γ in the differentiation capacity of P19 cells, we have established a HP1γ-overexpressing P19 cell line (HPlγ-P19). Similar to the parental P19 cells, undifferentiated HP1γ-P19 cells continued to express pluripotency marker genes. However, HP1γ-P19 cells exhibited significant morphological differentiation including beating cardiomyocytes, as well as Tuj1-positive neuronal cells and Sox17-positive endodermal cells after EB formation under a normal culture condition. Moreover, real-time RT-qPCR analysis revealed that HP1γ-P19 EB cells expressed various differentiation marker genes. Thus, HP1γ-P19 cells could give rise to all three germ layers in EBs without any drug treatment. Therefore, HP1γ affects the spontaneous differentiation potential of P19 cells, and might play major roles in the decision of cell fates in pluripotent stem cells.


Assuntos
Diferenciação Celular/fisiologia , Proteínas Cromossômicas não Histona/biossíntese , Células-Tronco de Carcinoma Embrionário/citologia , Células Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Técnicas de Cultura de Células , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/genética , Expressão Gênica , Humanos
11.
Biochem Biophys Res Commun ; 434(1): 131-6, 2013 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-23542464

RESUMO

Remote reperfusion lung injury following skeletal muscle ischemia and reperfusion accounts for high morbidity and mortality. AMP deaminase (AMPD), a key enzyme for nucleotide cycle, has been implicated in the regulation of this phenomenon. However, the function of Ampd2 and Ampd3 subtype has not been elucidated in remote reperfusion rodent lung injury. We utilized AMPD3 and AMPD2-deficient mice. The two types of AMPD-deficient mice and wild-type (WT) littermates were subjected to ischemia-reperfusion injury. After 3h bilateral hind-limb ischemia and reperfusion, AMPD3 mRNA, AMPD activity and inosine monophosphate (IMP) increased significantly in WT and AMPD2-deficient mice lungs, while they did not show significant alterations in AMPD3-deficient mice lungs. Genetic inactivation of Ampd3 resulted in markedly accelerated myeloperoxidase (MPO) activity along with exaggerated neutrophils infiltration and hemorrhage in the lungs compared to WT and AMPD2-deficient mice, furthermore, IMP treatment significantly attenuated MPO activity and neutrophils infiltration in WT and the two types of AMPD-deficient mice lungs after 3h reperfusion. These findings demonstrate for the first time in AMP-deficient mice models that AMPD3 plays a critical role in remote reperfusion lung injury via generation of IMP and validate the potential to use IMP into the clinical arena to attenuate remote ischemia-reperfusion lung injury.


Assuntos
AMP Desaminase/fisiologia , Lesão Pulmonar/enzimologia , Traumatismo por Reperfusão/enzimologia , AMP Desaminase/deficiência , AMP Desaminase/genética , Animais , Modelos Animais de Doenças , Inosina Monofosfato/administração & dosagem , Inosina Monofosfato/biossíntese , Lesão Pulmonar/genética , Lesão Pulmonar/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
12.
Circ J ; 76(12): 2875-83, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22972200

RESUMO

BACKGROUND: The prion protein (PrP) has been reported to serve as a surface maker for isolation of cardiomyogenic progenitors from murine embryonic stem (ES) cells. Although PrP-positive cells exhibited automaticity, their electrophysiological characteristics remain unresolved. The aim of the present study was therefore to investigate the electrophysiological properties of PrP-positive cells in comparison with those of HCN4p-or Nkx2.5-positive cells. METHODS AND RESULTS: Differentiation of AB1, HCN5p-EGFP and hcgp7 ES cells into cardiac progenitors was induced by embryoid body (EB) formation. EBs were dissociated and cells expressing PrP, HCN4-EGFP and/or Nkx2.5-GFP were collected via flow cytometry. Sorted cells were subjected to reverse transcriptase-polymerase chain reaction, immunostaining and patch-clamp experiments. PrP-positive cells expressed mRNA of undifferentiation markers, first and second heart field markers, and cardiac-specific genes and ion channels, indicating their commitment to cardiomyogenic progenitors. PrP-positive cells with automaticity showed positive and negative chronotropic responses to isoproterenol and carbamylcholine, respectively. Hyperpolarization-activated cation current (I(f)) was barely detectable, whereas Na(+) and L-type Ca(2+) channel currents were frequently observed. Their spontaneous activity was slowed by inhibition of sarcoplasmic reticulum Ca(2+) uptake and release but not by blocking I(f). The maximum diastolic potential of their spontaneous firings was more depolarized than that of Nkx2.5-GFP-positive cells. CONCLUSIONS: PrP-positive cells contained cardiac progenitors that separated from the lineage of sinoatrial node cells. PrP can be used as a marker to enrich nascent cardiac progenitors.


Assuntos
Potenciais de Ação , Células-Tronco Embrionárias/metabolismo , Miócitos Cardíacos/metabolismo , Príons/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Separação Celular/métodos , Técnicas de Cocultura , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Camundongos , Camundongos da Linhagem 129 , Contração Miocárdica , Técnicas de Patch-Clamp , Periodicidade , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
13.
J Radiat Res ; 63(1): 30-35, 2022 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-34718686

RESUMO

Ionizing radiation exposure affects the redox state in vivo. Recently, whole-blood antioxidant capacity (WBAC) has been reported to decrease in a dose-dependent manner after acute total body irradiation (TBI). However, changes in WBAC after localized and chronic irradiations have not been reported. This study analyzed changes to WBAC in mice after either localized irradiation (irradiation of the left hind leg only) or chronic TBI using the i-STrap method. Leg-localized irradiation exerted limited effects on WBAC, while WBAC decreased in a dose rate-dependent manner after TBI. Further, the WBAC reached the minimum value in a shorter period at a smaller dose rate. Our results suggest that changes in WBAC do not directly reflect absorbed dose, but may reflect radiation-induced biological damage at the systemic level. This study will contribute to the understanding of radiation-induced injuries and diseases, and will facilitate the establishment of biomarkers for radiation exposure.


Assuntos
Doença Enxerto-Hospedeiro , Lesões por Radiação , Animais , Antioxidantes , Biomarcadores , Camundongos , Proteínas de Ligação a RNA , Irradiação Corporal Total
14.
Regen Ther ; 21: 239-249, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36092505

RESUMO

Introduction: Dysfunction of the sinoatrial node (SAN) cells causes arrhythmias, and many patients require artificial cardiac pacemaker implantation. However, the mechanism of impaired SAN automaticity remains unknown, and the generation of human SAN cells in vitro may provide a platform for understanding the pathogenesis of SAN dysfunction. The short stature homeobox 2 (SHOX2) and hyperpolarization-activated cyclic nucleotide-gated cation channel 4 (HCN4) genes are specifically expressed in SAN cells and are important for SAN development and automaticity. In this study, we aimed to purify and characterize human SAN-like cells in vitro, using HCN4 and SHOX2 as SAN markers. Methods: We developed an HCN4-EGFP/SHOX2-mCherry dual reporter cell line derived from human induced pluripotent stem cells (hiPSCs), and HCN4 and SHOX2 gene expressions were visualized using the fluorescent proteins EGFP and mCherry, respectively. The dual reporter cell line was established using an HCN4-EGFP bacterial artificial chromosome-based semi-knock-in system and a CRISPR-Cas9-dependent knock-in system with a SHOX2-mCherry targeting vector. Flow cytometry, RT-PCR, and whole-cell patch-clamp analyses were performed to identify SAN-like cells. Results: Flow cytometry analysis and cell sorting isolated HCN4-EGFP single-positive (HCN4+/SHOX2-) and HCN4-EGFP/SHOX2-mCherry double-positive (HCN4+/SHOX2+) cells. RT-PCR analyses showed that SAN-related genes were enriched within the HCN4+/SHOX2+ cells. Further, electrophysiological analyses showed that approximately 70% of the HCN4+/SHOX2+ cells exhibited SAN-like electrophysiological characteristics, as defined by the action potential parameters of the maximum upstroke velocity and action potential duration. Conclusions: The HCN4-EGFP/SHOX2-mCherry dual reporter hiPSC system developed in this study enabled the enrichment of SAN-like cells within a mixed HCN4+/SHOX2+ population of differentiating cardiac cells. This novel cell line is useful for the further enrichment of human SAN-like cells. It may contribute to regenerative medicine, for example, biological pacemakers, as well as testing for cardiotoxic and chronotropic actions of novel drug candidates.

15.
Nat Cardiovasc Res ; 1(2): 142-156, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36051854

RESUMO

Some missense gain-of-function mutations in CACNA1C gene, encoding calcium channel CaV1.2, cause a life-threatening form of long QT syndrome named Timothy syndrome, with currently no clinically-effective therapeutics. Here we report that pharmacological targeting of sigma non-opioid intracellular receptor 1 (SIGMAR1) can restore electrophysiological function in iPSC-derived cardiomyocytes generated from patients with Timothy syndrome and two common forms of long QT syndrome, type 1 (LQTS1) and 2 (LQTS2), caused by missense trafficking mutations in potassium channels. Electrophysiological recordings demonstrate that an FDA-approved cough suppressant, dextromethorphan, can be used as an agonist of SIGMAR1, to shorten the prolonged action potential in Timothy syndrome cardiomyocytes and human cellular models of LQTS1 and LQTS2. When tested in vivo, dextromethorphan also normalized the prolonged QT intervals in Timothy syndrome model mice. Overall, our study demonstrates that SIGMAR1 is a potential therapeutic target for Timothy syndrome and possibly other inherited arrhythmias such as LQTS1 and LQTS2.

16.
J Radiat Res ; 62(5): 782-792, 2021 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-34265852

RESUMO

The mammalian target of rapamycin (mTOR) is a sensor of nutrient status and plays an important role in cell growth and metabolism. Although inhibition of mTOR signaling promotes tumor cell death and several mTOR inhibitors have been used clinically, recent reports have shown that co-treatment with MHY1485, an mTOR activator, enhances the anti-cancer effects of anti-PD-1 antibody and 5-fluorouracil. However, it remains unclear whether MHY1485 treatment alters the effects of radiation on tumor cells. In this study, the radiosensitizing effects of MHY1485 were investigated using murine CT26 and LLC cell lines. We examined mTOR signaling, tumor cell growth, colony formation, apoptosis, senescence, oxidative stress, p21 accumulation and endoplasmic reticulum (ER) stress levels in cells treated with MHY1485 and radiation, either alone or together. We found that MHY1485 treatment inhibited growth and colony formation in both cell lines under irradiation and no-irradiation conditions, results that were not fully consistent with MHY1485's known role in activating mTOR signaling. Furthermore, we found that combined treatment with MHY1485 and radiation significantly increased apoptosis and senescence in tumor cells in association with oxidative stress, ER stress and p21 stabilization, compared to radiation treatment alone. Our results suggested that MHY1485 enhances the radiosensitivity of tumor cells by a mechanism that may differ from MHY1485's role in mTOR activation.


Assuntos
Apoptose/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Morfolinas/farmacologia , Proteínas de Neoplasias/agonistas , Serina-Treonina Quinases TOR/efeitos dos fármacos , Triazinas/farmacologia , Animais , Apoptose/efeitos da radiação , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Senescência Celular/efeitos da radiação , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos da radiação , Genes p53 , Genes ras , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos da radiação , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/efeitos da radiação , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Ensaio Tumoral de Célula-Tronco
17.
Cell Rep Methods ; 1(7): 100092, 2021 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-35475001

RESUMO

Lactate metabolism has been shown to have increasingly important implications in cellular functions as well as in the development and pathophysiology of disease. The various roles as a signaling molecule and metabolite have led to interest in establishing a new method to detect lactate changes in live cells. Here we report our development of a genetically encoded metabolic indicator specifically for probing lactate (GEM-IL) based on superfolder fluorescent proteins and mutagenesis. With improvements in its design, specificity, and sensitivity, GEM-IL allows new applications compared with the previous lactate indicators, Laconic and Green Lindoblum. We demonstrate the functionality of GEM-IL to detect differences in lactate changes in human oncogenic neural progenitor cells and mouse primary ventricular myocytes. The development and application of GEM-IL show promise for enhancing our understanding of lactate dynamics and roles.


Assuntos
Ácido Láctico , Células-Tronco Neurais , Humanos , Animais , Camundongos , Ácido Láctico/metabolismo , Células-Tronco Neurais/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais
18.
Pacing Clin Electrophysiol ; 33(3): 290-303, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19895411

RESUMO

BACKGROUND: Development of biological pacemaker is a potential treatment for bradyarrhythmias. Pacemaker cells could be extracted from differentiated embryonic stem (ES) cells based on their specific cell marker hyperpolarization-activated cyclic nucleotide-gated (HCN)4. The goal of this study was to develop a method of identification, isolation, and characterization of pacemaking cells derived from differentiated ES cells with GFP driven by HCN4 promoter. METHODS AND RESULTS: Polymerase chain reaction (PCR) screening and southern blot analysis revealed that HCN4p-EGFP trans-gene was stably integrated into the chromosome of mouse AB1 ES cells. RT-PCR and immunostaining results showed similar expression of the specific cardiac pacemaker markers of the HCN4p-EGFP ES cells and its parental AB1 ES cell lines. Although HCN4p-EGFP trans-gene may have slight effect on the general mesodermal differentiation, it had no effect on the pluripotency of ES cells, on the transcription of cardiac specific factors and cardiac contractile proteins, and on the capability of ES cells to differentiate into pacemaker cells. Electrophysiological study indicated that HCN4p-GFP-positive cells revealed the spontaneous action potential, which was slowed by the treatment with 2 mM Cs(+), and expressed the hyperpolarization-activeted cation current I(f) encoded by HCN4 gene. CONCLUSION: By the approach of using stable transfectant of HCN4p-EGFP gene, the identification, isolation, and characterization of ES cell-derived pacemaking cells could be carried out.


Assuntos
Relógios Biológicos/fisiologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Células-Tronco Embrionárias/citologia , Sistema de Condução Cardíaco/citologia , Animais , Relógios Biológicos/genética , Southern Blotting , Bradicardia/genética , Bradicardia/terapia , Diferenciação Celular , Linhagem Celular , Separação Celular , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Primers do DNA , Técnicas Eletrofisiológicas Cardíacas , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Camundongos , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
19.
Nat Commun ; 11(1): 2141, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32358538

RESUMO

Optogenetic genome engineering tools enable spatiotemporal control of gene expression and provide new insight into biological function. Here, we report the new version of genetically encoded photoactivatable (PA) Cre recombinase, PA-Cre 3.0. To improve PA-Cre technology, we compare light-dimerization tools and optimize for mammalian expression using a CAG promoter, Magnets, and 2A self-cleaving peptide. To prevent background recombination caused by the high sequence similarity in the dimerization domains, we modify the codons for mouse gene targeting and viral production. Overall, these modifications significantly reduce dark leak activity and improve blue-light induction developing our new version, PA-Cre 3.0. As a resource, we have generated and validated AAV-PA-Cre 3.0 as well as two mouse lines that can conditionally express PA-Cre 3.0. Together these new tools will facilitate further biological and biomedical research.


Assuntos
Integrases/metabolismo , Recombinação Genética/genética , Animais , Códon/genética , Engenharia Genética/métodos , Integrases/genética , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Optogenética , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/efeitos da radiação , Recombinação Genética/efeitos da radiação
20.
Dev Growth Differ ; 51(2): 81-93, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19207180

RESUMO

Embryonic carcinoma (EC) cells, which are malignant stem cells of teratocarcinoma, have numerous morphological and biochemical properties in common with pluripotent stem cells such as embryonic stem (ES) cells. However, three EC cell lines (F9, P19 and PCC3) show different developmental potential and self-renewal capacity from those of ES cells. All three EC cell lines maintain self-renewal capacity in serum containing medium without Leukemia Inhibitory factor (LIF) or feeder layer, and show limited differentiation capacity into restricted lineage and cell types. To reveal the underlying mechanism of these characteristics, we took the approach of characterizing extrinsic factors derived from EC cells on the self-renewal capacity and pluripotency of mouse ES cells. Here we demonstrate that EC cell lines F9 and P19 produce factor(s) maintaining the undifferentiated state of mouse ES cells via an unidentified signal pathway, while P19 and PCC3 cells produce self-renewal factors of ES cells other than LIF that were able to activate the STAT3 signal; however, inhibition of STAT3 activation with Janus kinase inhibitor shows only partial impairment on the maintenance of the undifferentiated state of ES cells. Thus, these factors present in EC cells-derived conditioned medium may be responsible for the self-renewal capacity of EC and ES cells independently of LIF signaling.


Assuntos
Fatores Biológicos/farmacologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Transdução de Sinais , Animais , Fatores Biológicos/metabolismo , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Meios de Cultura/farmacologia , Meios de Cultivo Condicionados/farmacologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Reporter , Camadas Germinativas/citologia , Janus Quinases/antagonistas & inibidores , Janus Quinases/fisiologia , Fator Inibidor de Leucemia/fisiologia , Camundongos , Células-Tronco Pluripotentes/citologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa