Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 439
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 150(2): 111-127, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38726666

RESUMEN

BACKGROUND: G protein-coupled receptors play a critical role in atrial fibrillation (AF). Spexin is a novel ligand of galanin receptors (GALRs). In this study, we investigated the regulation of spexin and GALRs on AF and the underlying mechanisms. METHODS: Global spexin knockout (SPX-KO) and cardiomyocyte-specific GALRs knockout (GALR-cKO) mice underwent burst pacing electrical stimulation. Optical mapping was used to determine atrial conduction velocity and action potential duration. Atrial myocyte action potential duration and inward rectifying K+ current (IK1) were recorded using whole-cell patch clamps. Isolated cardiomyocytes were stained with Fluo-3/AM dye, and intracellular Ca2+ handling was examined by CCD camera. A mouse model of AF was established by Ang-II (angiotensin II) infusion. RESULTS: Spexin plasma levels in patients with AF were lower than those in subjects without AF, and knockout of spexin increased AF susceptibility in mice. In the atrium of SPX-KO mice, potassium inwardly rectifying channel subfamily J member 2 (KCNJ2) and sarcolipin (SLN) were upregulated; meanwhile, IK1 current was increased and Ca2+ handling was impaired in isolated atrial myocytes of SPX-KO mice. GALR2-cKO mice, but not GALR1-cKO and GALR3-cKO mice, had a higher incidence of AF, which was associated with higher IK1 current and intracellular Ca2+ overload. The phosphorylation level of CREB (cyclic AMP responsive element binding protein 1) was upregulated in atrial tissues of SPX-KO and GALR2-cKO mice. Chromatin immunoprecipitation confirmed the recruitment of p-CREB to the proximal promoter regions of KCNJ2 and SLN. Finally, spexin treatment suppressed CREB signaling, decreased IK1 current and decreased intracellular Ca2+ overload, which thus reduced the inducibility of AF in Ang-II-infused mice. CONCLUSIONS: Spexin reduces atrial fibrillation susceptibility by inhibiting CREB phosphorylation and thus downregulating KCNJ2 and SLN transcription by GALR2 receptor. The spexin/GALR2/CREB signaling pathway represents a novel therapeutic avenue in the development of agents against atrial fibrillation.


Asunto(s)
Fibrilación Atrial , Ratones Noqueados , Miocitos Cardíacos , Hormonas Peptídicas , Receptor de Galanina Tipo 2 , Animales , Fibrilación Atrial/metabolismo , Hormonas Peptídicas/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Receptor de Galanina Tipo 2/metabolismo , Receptor de Galanina Tipo 2/genética , Humanos , Potenciales de Acción/efectos de los fármacos , Masculino , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Femenino , Transducción de Señal
2.
Circ Res ; 132(2): 208-222, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36656967

RESUMEN

OBJECTIVE: ASPP1 (apoptosis stimulating of p53 protein 1) is critical in regulating cell apoptosis as a cofactor of p53 to promote its transcriptional activity in the nucleus. However, whether cytoplasmic ASPP1 affects p53 nuclear trafficking and its role in cardiac diseases remains unknown. This study aims to explore the mechanism by which ASPP1 modulates p53 nuclear trafficking and the subsequent contribution to cardiac ischemia/reperfusion (I/R) injury. METHODS AND RESULTS: The immunofluorescent staining showed that under normal condition ASPP1 and p53 colocalized in the cytoplasm of neonatal mouse ventricular cardiomyocytes, while they were both upregulated and translocated to the nuclei upon hypoxia/reoxygenation treatment. The nuclear translocation of ASPP1 and p53 was interdependent, as knockdown of either ASPP1 or p53 attenuated nuclear translocation of the other one. Inhibition of importin-ß1 resulted in the cytoplasmic sequestration of both p53 and ASPP1 in neonatal mouse ventricular cardiomyocytes with hypoxia/reoxygenation stimulation. Overexpression of ASPP1 potentiated, whereas knockdown of ASPP1 inhibited the expression of Bax (Bcl2-associated X), PUMA (p53 upregulated modulator of apoptosis), and Noxa, direct apoptosis-associated targets of p53. ASPP1 was also increased in the I/R myocardium. Cardiomyocyte-specific transgenic overexpression of ASPP1 aggravated I/R injury as indicated by increased infarct size and impaired cardiac function. Conversely, knockout of ASPP1 mitigated cardiac I/R injury. The same qualitative data were observed in neonatal mouse ventricular cardiomyocytes exposed to hypoxia/reoxygenation injury. Furthermore, inhibition of p53 significantly blunted the proapoptotic activity and detrimental effects of ASPP1 both in vitro and in vivo. CONCLUSIONS: Binding of ASPP1 to p53 triggers their nuclear cotranslocation via importin-ß1 that eventually exacerbates cardiac I/R injury. The findings imply that interfering the expression of ASPP1 or the interaction between ASPP1 and p53 to block their nuclear trafficking represents an important therapeutic strategy for cardiac I/R injury.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Daño por Reperfusión , Proteína p53 Supresora de Tumor , Animales , Ratones , Apoptosis/fisiología , Hipoxia/metabolismo , Isquemia/metabolismo , Carioferinas , Miocitos Cardíacos/metabolismo , Daño por Reperfusión/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Adaptadoras Transductoras de Señales/genética
3.
J Cell Mol Med ; 28(3): e18094, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38214430

RESUMEN

Lung cancer is a leading cause of cancer-related deaths worldwide. Recent studies have identified pyroptosis, a type of programmed cell death, as a critical process in the development and progression of lung cancer. In this study, we investigated the effect of EEBR, a new compound synthesized by our team, on pyroptosis in non-small cell lung cancer cells (NSCLC) and the underlying molecular mechanisms. Our results demonstrated that EEBR significantly reduced the proliferation and metastasis of NSCLC cells in vitro. Moreover, EEBR-induced pyroptosis in NSCLC cells, as evidenced by cell membrane rupture, the release of cytokines such as interleukin-18 and interleukin-1 beta and the promotion of Gasdermin D cleavage in a Caspase-1-dependent manner. Furthermore, EEBR promoted the nuclear translocation of NF-κB and upregulated the protein level of NLRP3. Subsequent studies revealed that EEBR-induced pyroptosis was suppressed by the inhibition of NF-κB. Finally, EEBR effectively suppressed the growth of lung cancer xenograft tumours by promoting NSCLC pyroptosis in animal models. Taken together, our findings suggest that EEBR induces Caspase-1-dependent pyroptosis through the NF-κB/NLRP3 signalling cascade in NSCLC, highlighting its potential as a candidate drug for NSCLC treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Humanos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Piroptosis , Caspasa 1/metabolismo , Inflamasomas/metabolismo
4.
J Transl Med ; 21(1): 52, 2023 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-36707890

RESUMEN

BACKGROUND/AIMS: Arsenic trioxide (ATO) is the first-line therapeutic drug for acute promyelocytic leukemia. However, the cardiotoxicity of ATO limits its clinical application. This study aims to explore the long noncoding RNA (lncRNA) involved molecular mechanism in ATO-induced cardiotoxicity and to identify available prevention strategies. METHODS: ATO was administered to mice or primary cultured mouse cardiomyocytes. Small interfering RNA targeting lncRNA Kcnq1ot1 (si-Kcnq1ot1) was used to knockdown lncRNA Kcnq1ot1. MiR-34a-5p mimic and antisense morpholino oligonucleotide targeting miR-34a-5p (AMO-34a-5p) were used to upregulate and downregulate the expression of miR-34a-5p, respectively. TUNEL staining was conducted to detect cell DNA damage. Flow cytometry assay was used to detect cell apoptosis. Western blot was conducted to detect Bcl-2, Bax and Sirt1 protein expression. Real-time PCR was used to detect lncRNA Kcnq1ot1, miR-34a-5p, and Sirt1 mRNA expression. Dual-luciferase reporter assay was performed to validate the predicted binding site. RESULTS: ATO induced apoptosis in cardiomyocytes both in vivo and in vitro. Simultaneously, the expression of lncRNA Kcnq1ot1 and Sirt1 was downregulated, and miR-34a-5p was upregulated. MiR-34a-5p has binding sites with lncRNA Kcnq1ot1 and Sirt1. Knockdown of lncRNA Kcnq1ot1 induced apoptosis of cardiomyocytes, with increased miR-34a-5p and decreased Sirt1 expression. Inhibition of miR-34a-5p attenuated si-Kcnq1ot1-induced apoptosis in cardiomyocytes. Therefore, the lncRNA Kcnq1ot1/miR-34a-5p/Sirt1 signaling pathway is involved in ATO-induced cardiotoxicity. Propranolol alleviated ATO-induced apoptosis in cardiomyocytes both in vivo and in vitro, which was related to the lncRNA Kcnq1ot1/miR-34a-5p/Sirt1 signaling pathway. CONCLUSION: The lncRNA Kcnq1ot1/miR-34a-5p/Sirt1 pathway is involved in ATO-induced cardiotoxicity. Propranolol can attenuate ATO-induced cardiotoxicity at least partially through the lncRNA Kcnq1ot1/miR-34a-5p/Sirt1 pathway. Combined administration with propranolol may be a new strategy for alleviating the cardiotoxicity of ATO.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Ratones , Animales , Trióxido de Arsénico , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cardiotoxicidad , Sirtuina 1/genética , Propranolol , Apoptosis/genética
5.
J Magn Reson Imaging ; 57(4): 1212-1221, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35856797

RESUMEN

BACKGROUND: Angelman syndrome (AS) is a genetic disorder that affects neurodevelopment. The investigation of changes in the brain white matter network, which would contribute to a better understanding of the pathogenesis of AS brain, was lacking. PURPOSE: To investigate both local and global alterations of white matter in patients with AS. STUDY TYPE: Prospective. SUBJECTS: A total of 29 AS patients (6.6 ± 1.4 years, 15 [52%] females) and 19 age-matched healthy controls (HC) (7.0 ± 1.5 years, 10 [53%] females). FIELD STRENGTH/SEQUENCE: A 3-T, three-dimensional (3D) T1-weighted imaging by using gradient-echo-based sequence, single shell diffusion tensor imaging by using spin-echo-based echo-planar imaging. ASSESSMENT: Network metrics including global efficiency (Eg ), local efficiency (Eloc ), small world coefficient (Swc), rich-club coefficient (Φ), and nodal degree (ND) were estimated from diffusion MR (dMR) data. Connections among highly connected (hub) regions and less connected (peripheral) regions were also assessed. Correlation between the topological parameters and age for each group was also calculated to assess the development of the brain. STATISTICAL TESTS: Linear regression model, permutation test. P values estimated from the regression model for each brain region were adjusted by false discovery rate (FDR) correction. RESULTS: AS patients showed significantly lower Eg and higher swc compared to HC. Φn significantly increased at higher k-levels in AS patients. In addition, the connections among hub regions and peripheral regions were significantly interrupted in AS patients. DATA CONCLUSION: The AS brain showed diminished connectivity, reflected by reduced network efficiency compared to HC. Compared to densely connected regions, less connected regions were more vulnerable in AS. EVIDENCE LEVEL: 2 TECHNICAL EFFICACY: Stage 3.


Asunto(s)
Síndrome de Angelman , Sustancia Blanca , Femenino , Humanos , Preescolar , Niño , Masculino , Imagen de Difusión Tensora/métodos , Síndrome de Angelman/patología , Estudios Prospectivos , Encéfalo/patología
6.
Mol Biol Rep ; 50(1): 279-288, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36331752

RESUMEN

BACKGROUND: Postoperative abdominal adhesions (PAAs) represent a frequent condition occurring in more than 90% of patients undergoing abdomen and pelvic surgeries, which can cause chronic abdominal pain, female infertility, and repeated bowel obstruction, requiring repetitive surgical interventions causing morbidity and mortality, as well as high costs. It is therefore of paramount clinical importance and significance to develop practical and reliable strategies for preventing the occurrence of PAAs. METHODS AND RESULTS: In this study, we demonstrated that Nianfukang (NFK, composed of polyethylene glycol 1450 and diclofenac sodium) is highly effective in preventing PAAs, likely by reducing leukocytes and inflammatory factors in the abdominal cavity, and inhibiting intestinal fibrosis in a rat model of PAAs induced by postoperative cecum scraping. We further uncovered that NFK downregulates the expression of TGF-ß1, a key factor for adhesion formation, to suppress the TGF-ß1/TGF-ßRIII/Smad2 signaling pathway, thereby inhibiting the proliferation and migration of fibroblasts and provided evidences for the involvement of the TGF-ß1/TGF-ßRIII/Smad2 axis in the prevention of PAAs in normal human colon fibroblast CCD-18Co. CONCLUSIONS: Our findings support NFK as a potential anti-adhesive product that has the advantages of significant effectiveness, safety profile, and low cost, as well as clear mechanism of action.


Asunto(s)
Abdomen , Factor de Crecimiento Transformador beta1 , Humanos , Ratas , Femenino , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Abdomen/cirugía , Transducción de Señal , Fibroblastos/metabolismo , Fibrosis
7.
Acta Pharmacol Sin ; 44(5): 999-1013, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36347996

RESUMEN

Non-healing diabetic wounds (DW) are a serious clinical problem that remained poorly understood. We recently found that topical application of growth differentiation factor 11 (GDF11) accelerated skin wound healing in both Type 1 DM (T1DM) and genetically engineered Type 2 diabetic db/db (T2DM) mice. In the present study, we elucidated the cellular and molecular mechanisms underlying the action of GDF11 on healing of small skin wound. Single round-shape full-thickness wound of 5-mm diameter with muscle and bone exposed was made on mouse dorsum using a sterile punch biopsy 7 days following the onset of DM. Recombinant human GDF11 (rGDF11, 50 ng/mL, 10 µL) was topically applied onto the wound area twice a day until epidermal closure (maximum 14 days). Digital images of wound were obtained once a day from D0 to D14 post-wounding. We showed that topical application of GDF11 accelerated the healing of full-thickness skin wounds in both type 1 and type 2 diabetic mice, even after GDF8 (a muscle growth factor) had been silenced. At the cellular level, GDF11 significantly facilitated neovascularization to enhance regeneration of skin tissues by stimulating mobilization, migration and homing of endothelial progenitor cells (EPCs) to the wounded area. At the molecular level, GDF11 greatly increased HIF-1ɑ expression to enhance the activities of VEGF and SDF-1ɑ, thereby neovascularization. We found that endogenous GDF11 level was robustly decreased in skin tissue of diabetic wounds. The specific antibody against GDF11 or silence of GDF11 by siRNA in healthy mice mimicked the non-healing property of diabetic wound. Thus, we demonstrate that GDF11 promotes diabetic wound healing via stimulating endothelial progenitor cells mobilization and neovascularization mediated by HIF-1ɑ-VEGF/SDF-1ɑ pathway. Our results support the potential of GDF11 as a therapeutic agent for non-healing DW.


Asunto(s)
Diabetes Mellitus Experimental , Células Progenitoras Endoteliales , Factores de Diferenciación de Crecimiento , Cicatrización de Heridas , Animales , Humanos , Ratones , Proteínas Morfogenéticas Óseas/metabolismo , Quimiocina CXCL12/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/patología , Factores de Diferenciación de Crecimiento/uso terapéutico , Factores de Diferenciación de Crecimiento/metabolismo , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
8.
Pharmacol Res ; 182: 106284, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35661710

RESUMEN

Pathological cardiac hypertrophy is a process characterized by significant disturbance of protein turnover. Cullin-associated and Neddylation-dissociated 1 (CAND1) acts as a coordinator to modulate substrate protein degradation by promoting the formation of specific cullin-based ubiquitin ligase 3 complex in response to substrate accumulation, which thereby facilitate the maintaining of normal protein homeostasis. Accumulation of calcineurin is critical in the pathogenesis of cardiac hypertrophy and heart failure. However, whether CAND1 titrates the degradation of hypertrophy related protein eg. calcineurin and regulates cardiac hypertrophy remains unknown. Therefore, we aim to explore the role of CAND1 in cardiac hypertrophy and heart failure and the underlying molecular mechanism. Here, we found that the protein level of CAND1 was increased in cardiac tissues from heart failure (HF) patients and TAC mice, whereas the mRNA level did not change. CAND1-KO+ /- aggravated TAC-induced cardiac hypertrophic phenotypes; in contrast, CAND1-Tg attenuated the maladaptive cardiac remodeling. At the molecular level, CAND1 overexpression downregulated, whereas CAND1-KO+ /- or knockdown upregulated calcineurin expression at both in vivo and in vitro conditions. Mechanistically, CAND1 overexpression favored the assembly of Cul1/atrogin1/calcineurin complex and rendered the ubiquitination and degradation of calcineurin. Notably, CAND1 deficiency-induced hypertrophic phenotypes were partially rescued by knockdown of calcineurin, and application of exogenous CAND1 prevented TAC-induced cardiac hypertrophy. Taken together, our findings demonstrate that CAND1 exerts a protective effect against cardiac hypertrophy and heart failure partially by inducing the degradation of calcineurin.


Asunto(s)
Calcineurina , Cardiomegalia , Proteínas Cullin , Insuficiencia Cardíaca , Animales , Calcineurina/metabolismo , Cardiomegalia/genética , Proteínas Cullin/química , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Insuficiencia Cardíaca/genética , Humanos , Ratones , Factores de Transcripción
9.
Acta Pharmacol Sin ; 43(3): 613-623, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34035486

RESUMEN

Heart aging is characterized by structural and diastolic dysfunction of the heart. However, there is still no effective drug to prevent and treat the abnormal changes in cardiac function caused by aging. Here, we present the preventive effects of emodin and its derivative Kanglexin (KLX) against heart aging. We found that the diastolic dysfunction and cardiac remodeling in mice with D-galactose (D-gal)-induced aging were markedly mitigated by KLX and emodin. In addition, the senescence of neonatal mouse cardiomyocytes induced by D-gal was also reversed by KLX and emodin treatment. However, KLX exhibited better anti-heart aging effects than emodin at the same dose. Dysregulated mitophagy was observed in aging hearts and in senescent neonatal mouse cardiomyocytes, and KLX produced a greater increase in mitophagy than emodin. The mitophagy-promoting effects of KLX and emodin were ascribed to their abilities to enhance the protein stability of Parkin, a key modulator in mitophagy, with different potencies. Molecular docking and SPR analysis demonstrated that KLX has a higher affinity for the ubiquitin-like (UBL) domain of Parkin than emodin. The UBL domain might contribute to the stabilizing effects of KLX on Parkin. In conclusion, this study identifies KLX and emodin as effective anti-heart aging drugs that activate Parkin-mediated mitophagy and outlines their putative therapeutic importance.


Asunto(s)
Envejecimiento/efectos de los fármacos , Antraquinonas/farmacología , Emodina/farmacología , Cardiopatías/patología , Mitofagia/efectos de los fármacos , Animales , Benzofuranos , Modelos Animales de Enfermedad , Femenino , Galactosa/farmacología , Ratones , Simulación del Acoplamiento Molecular , Miocitos Cardíacos/efectos de los fármacos , Quinolinas , Distribución Aleatoria , Ubiquitina-Proteína Ligasas/efectos de los fármacos
10.
Cell Mol Life Sci ; 79(1): 43, 2021 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-34921634

RESUMEN

The initial identification of long non-coding RNA myocardial infarction associated transcript (MIAT) as a genetic risk factor of myocardial infarction has made this lncRNA (designated as lncR-MIAT here) a focus of intensive studies worldwide. Emerging evidence supports that lncR-MIAT is susceptible in its expression to multiple deleterious factors like angiotensin II, isoproterenol, hypoxia, and infection and is anomaly overexpressed in serum, plasma, blood cells and myocardial tissues under a variety of cardiovascular conditions including myocardial infarction, cardiac hypertrophy, diabetic cardiomyopathy, dilated cardiomyopathy, sepsis cardiomyopathy, atrial fibrillation and microvascular dysfunction. Experimental results consistently demonstrated that upregulation of lncR-MIAT plays active roles in the pathological processes of the cardiovascular system and knockdown of this lncRNA effectively ameliorates the adverse conditions. The available data revealed that lncR-MIAT acts through multiple mechanisms such as competitive endogenous RNA, natural antisense RNA and RNA/protein interactions. Moreover, the functional domains of lncR-MIAT accounting for certain specific cellular functions of the full-length transcript have been identified and characterized. These insights will not only tremendously advance our understanding of lncRNA biology and pathophysiology, but also offer good opportunities for more innovative and precise design of agents that have the potential to be developed into new drugs for better therapy of cardiovascular diseases (CVDs) in the future. Herein, we provide an overview of lncR-MIAT, focusing on its roles in cardiovascular diseases, underline the unique cellular/molecular mechanisms for its actions, and speculate the perspectives about the translational studies on the potential diagnostic and therapeutic applications of lncR-MIAT.


Asunto(s)
Enfermedades Cardiovasculares , Miocardio , ARN Largo no Codificante/fisiología , Animales , Biomarcadores/metabolismo , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Humanos , Miocardio/metabolismo , Miocardio/patología
11.
Acta Biochim Biophys Sin (Shanghai) ; 54(2): 199-208, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35130614

RESUMEN

Methyltransferase-like 3 (Mettl3) is a component of methyltransferase complex that mediates mA modification of RNAs, and participates in multiple biological processes. However, the role of Mettl3 in cardiac electrophysiology remains unknown. This study aims to explore the ventricular arrhythmia susceptibility of Mettl3 mice and the underlying mechanisms. Mice were anesthetized with 2% avertin (0.1 mL/ body weight) for echocardiography and programmed electrical pacing. Whole-cell patch clamp technique was used to examine the electrophysiological property of cardiomyocytes. The expression of Cav1.2 was determined by qRT-PCR and western blot analysis. The mA medication of mRNA was examined by MeRIP-Seq and MeRIP-qPCR. No differences are found in the morphology and function of the hearts between Mettl3 mice and wild-type (WT) controls. The QT and QTc intervals of Mettl3 mice are significantly longer. High-frequency electrical stimulation showed that heterozygous knockout of Mettl3 increases ventricular arrhythmia susceptibility. The whole-cell patch-clamp recordings showed that the APD is prolonged in Mettl3 ventricular myocytes and more EADs were observed. The density of is substantially increased in ventricular myocytes of Mettl3 mice. The pore-forming subunit of L-type calcium channel Cav1.2 is upregulated in Mettl3 mice, while the mRNA of its coding gene does not change. MeRIP-Seq and MeRIP-qPCR showed that the mA methylation of mRNA is decreased in cultured Mettl3-knockdown cardiomyocytes and Mettl3 hearts. Collectively, deficiency of Mettl3 increases ventricular arrhythmia susceptibility due to the upregulation of Cav1.2 by reducing mA modification onmRNA in mice. This study highlights the role of mA modification in the regulation of cardiac electrophysiology.


Asunto(s)
Arritmias Cardíacas , Canales de Calcio Tipo L/metabolismo , Miocitos Cardíacos , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Activación Transcripcional , Regulación hacia Arriba
12.
J Mol Cell Cardiol ; 158: 128-139, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34043986

RESUMEN

Myocardial infarction (MI)-induced the activation of NLRP3 inflammasome has been well known to aggravate myocardial injury and cardiac dysfunction by causing inflammation and pyroptosis in the heart. Circular RNAs (circRNAs) have been demonstrated to play critical roles in cardiovascular diseases. However, the functions and mechanisms of circRNAs in modulating cardiac inflammatory response and cardiomyocyte pyroptosis remain largely unknown. We revealed that circHelz, a novel circRNA transcribed from the helicase with zinc finger (Helz) gene, was significantly upregulated in both the ischemic myocardium of MI mouse and neonatal mouse ventricular cardiomyocytes (NMVCs) exposed to hypoxia. Overexpression of circHelz caused cardiomyocyte injury in NMVCs by activating the NLRP3 inflammasome and inducing pyroptosis, while circHelz silencing reduced these effects induced by hypoxia. Furthermore, knockdown of circHelz remarkably attenuated NLRP3 expression, decreased myocardial infarct size, pyroptosis, inflammation, and increased cardiac function in vivo after MI. Overexpression of miR-133a-3p in cardiomyocytes greatly prevented pyroptosis in the presence of hypoxia or circHelz by targeting NLRP3 in NMVCs. Mechanistically, circHelz functioned as an endogenous sponge for miR-133a-3p via suppressing its activity. Overall, our results demonstrate that circHelz causes myocardial injury by triggering the NLRP3 inflammasome-mediated pro-inflammatory response and subsequent pyroptosis in cardiomyocytes by inhibiting miR-133a-3p function. Therefore, interfering with circHelz/miR-133a-3p/NLRP3 axis might be a promising therapeutic approach for ischemic cardiac diseases.


Asunto(s)
Silenciador del Gen , Inflamasomas/metabolismo , MicroARNs/metabolismo , Infarto del Miocardio/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ARN Helicasas/genética , ARN Circular/metabolismo , Transducción de Señal/genética , Animales , Animales Recién Nacidos , Hipoxia de la Célula , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/metabolismo , Piroptosis/genética , ARN Circular/genética , Transfección , Regulación hacia Arriba
13.
Neuroimage ; 240: 118376, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34246768

RESUMEN

Quantitative susceptibility mapping (QSM) has demonstrated great potential in quantifying tissue susceptibility in various brain diseases. However, the intrinsic ill-posed inverse problem relating the tissue phase to the underlying susceptibility distribution affects the accuracy for quantifying tissue susceptibility. Recently, deep learning has shown promising results to improve accuracy by reducing the streaking artifacts. However, there exists a mismatch between the observed phase and the theoretical forward phase estimated by the susceptibility label. In this study, we proposed a model-based deep learning architecture that followed the STI (susceptibility tensor imaging) physical model, referred to as MoDL-QSM. Specifically, MoDL-QSM accounts for the relationship between STI-derived phase contrast induced by the susceptibility tensor terms (χ13, χ23 and χ33) and the acquired single-orientation phase. The convolutional neural networks are embedded into the physical model to learn a regularization term containing prior information. χ33 and phase induced by χ13 and χ23 terms were used as the labels for network training. Quantitative evaluation metrics were compared with recently developed deep learning QSM methods. The results showed that MoDL-QSM achieved superior performance, demonstrating its potential for future applications.


Asunto(s)
Mapeo Encefálico/métodos , Encéfalo/diagnóstico por imagen , Aprendizaje Profundo , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Redes Neurales de la Computación , Encéfalo/fisiología , Humanos
14.
Acta Pharmacol Sin ; 42(5): 691-700, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32759964

RESUMEN

Myocardial infarction (MI) leads to the loss of cardiomyocytes, left ventricle dilation and cardiac dysfunction, eventually developing into heart failure. Mzb1 (Marginal zone B and B1 cell specific protein 1) is a B-cell-specific and endoplasmic reticulum-localized protein. Mzb1 is an inflammation-associated factor that participates a series of inflammatory processes, including chronic periodontitis and several cancers. In this study we investigated the role of Mzb1 in experimental models of MI. MI was induced in mice by ligation of the left descending anterior coronary artery, and in neonatal mouse ventricular cardiomyocytes (NMVCs) by H2O2 treatment in vitro. We showed that Mzb1 expression was markedly reduced in the border zone of the infarct myocardium of MI mice and in H2O2-treated NMVCs. In H2O2-treated cardiomyocytes, knockdown of Mzb1 decreased mitochondrial membrane potential, impaired mitochondrial function and promoted apoptosis. On contrary, overexpression of Mzb1 improved mitochondrial membrane potential, ATP levels and mitochondrial oxygen consumption rate (OCR), and inhibited apoptosis. Direct injection of lentiviral vector carrying Len-Mzb1 into the myocardial tissue significantly improved cardiac function and alleviated apoptosis in MI mice. We showed that Mzb1 overexpression significantly decreased the levels of Bax/Bcl-2 and cytochrome c and improved mitochondrial function in MI mice via activating the AMPK-PGC1α pathway. In addition, we demonstrated that Mzb1 recruited the macrophages and alleviated inflammation in MI mice. We conclude that Mzb1 is a crucial regulator of cardiomyocytes after MI by improving mitochondrial function and reducing inflammatory signaling pathways, implying a promising therapeutic target in ischemic cardiomyopathy.


Asunto(s)
Inflamación/metabolismo , Mitocondrias/metabolismo , Chaperonas Moleculares/metabolismo , Infarto del Miocardio/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Regulación hacia Abajo , Corazón/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
15.
Acta Pharmacol Sin ; 42(11): 1780-1789, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33589793

RESUMEN

Interleukin-17 (IL-17), also called IL-17A, is an important regulator of cardiac diseases, but its role in calcium-related cardiac dysfunction remains to be explored. Thus, we investigated the influence of IL-17 on calcium handling process and its contribution to the development of heart failure. Mice were subjected to transaortic constriction (TAC) to induce heart failure. In these mice, the levels of IL-17 in the plasma and cardiac tissue were significantly increased compared with the sham group. In 77 heart failure patients, the plasma level of IL-17 was significantly higher than 49 non-failing subjects, and was negatively correlated with cardiac ejection fraction and fractional shortening. In IL-17 knockout mice, the shortening of isolated ventricular myocytes was increased compared with that in wild-type mice, which was accompanied by significantly increased amplitude of calcium transient and the upregulation of SERCA2a and Cav1.2. In cultured neonatal cardiac myocytes, treatment of with IL-17 (0.1, 1 ng/mL) concentration-dependently suppressed the amplitude of calcium transient and reduced the expression of SERCA2a and Cav1.2. Furthermore, IL-17 treatment increased the expression of the NF-κB subunits p50 and p65, whereas knockdown of p50 reversed the inhibitory effects of IL-17 on SERCA2a and Cav1.2 expression. In mice with TAC-induced mouse heart, IL-17 knockout restored the expression of SERCA2a and Cav1.2, increased the amplitude of calcium transient and cell shortening, and in turn improved cardiac function. In addition, IL-17 knockout attenuated cardiac hypertrophy with inhibition of calcium-related signaling pathway. In conclusion, upregulation of IL-17 impairs cardiac function through NF-κB-mediated disturbance of calcium handling and cardiac remodeling. Inhibition of IL-17 represents a potential therapeutic strategy for the treatment of heart failure.


Asunto(s)
Canales de Calcio Tipo L/biosíntesis , Insuficiencia Cardíaca/metabolismo , Interleucina-17/biosíntesis , FN-kappa B/biosíntesis , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/biosíntesis , Regulación hacia Arriba/fisiología , Animales , Animales Recién Nacidos , Canales de Calcio Tipo L/genética , Línea Celular , Células Cultivadas , Expresión Génica , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Humanos , Interleucina-17/deficiencia , Interleucina-17/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética
16.
Acta Biochim Biophys Sin (Shanghai) ; 53(1): 102-111, 2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33128543

RESUMEN

Currently, there remains a great need to elucidate the molecular mechanism of acute myocardial infarction in order to facilitate the development of novel therapy. Inhibitor of apoptosis-stimulating protein of p53 (iASPP) is a member of the ASPP family proteins and an evolutionarily preserved inhibitor of p53 that is involved in many cellular processes, including apoptosis of cancer cells. The purpose of this study was to investigate the possible role of iASPP in acute myocardial infarction. The protein level of iASPP was markedly reduced in the ischemic hearts in vivo and hydrogen peroxide-exposed cardiomyocytes in vitro. Overexpression of iASPP reduced the infarct size and cardiomyocyte apoptosis of mice subjected to 24 h of coronary artery ligation. Echocardiography showed that cardiac function was improved as indicated by the increase in ejection fraction and fractional shortening. In contrast, knockdown of iASPP exacerbated cardiac injury as manifested by impaired cardiac function, increased infarct size, and apoptosis rate. Mechanistically, overexpression of iASPP inhibited, while knockdown of iASPP increased the expressions of p53 and Bax, the key regulators of apoptosis. Taken together, our results suggested that iASPP is an important regulator of cardiomyocyte apoptosis, which represents a potential target in the therapy of myocardial infarction.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Animales , Apoptosis/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Lesiones Cardíacas/genética , Lesiones Cardíacas/patología , Lesiones Cardíacas/prevención & control , Peróxido de Hidrógeno/toxicidad , Masculino , Ratones Endogámicos C57BL , Isquemia Miocárdica/etiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Cultivo Primario de Células , Proteína X Asociada a bcl-2/metabolismo
17.
J Cell Mol Med ; 24(14): 7991-8005, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32468736

RESUMEN

Previous studies have confirmed that miR-195 expression is increased in cardiac hypertrophy, and the bioinformatics website predicted by Targetscan software shows that miR-195 can directly target CACNB1, KCNJ2 and KCND3 to regulate Cavß1, Kir2.1 and Kv4.3 proteins expression. The purpose of this study is to confirm the role of miR-195 in arrhythmia caused by cardiac hypertrophy. The protein levels of Cavß1, Kir2.1 and Kv4.3 in myocardium of HF mice were decreased. After miR-195 was overexpressed in neonatal mice cardiomyocytes, the expression of ANP, BNP and ß-MHC was up-regulated, and miR-195 inhibitor reversed this phenomenon. Overexpression of miR-195 reduced the estimated cardiac function of EF% and FS% in wild-type (WT) mice. Transmission electron microscopy showed that the ultrastructure of cardiac tissues was damaged after miR-195 overexpression by lentivirus in mice. miR-195 overexpression increased the likelihood of arrhythmia induction and duration of arrhythmia in WT mice. Lenti-miR-195 inhibitor carried by lentivirus can reverse the decreased EF% and FS%, the increased incidence of arrhythmia and prolonged duration of arrhythmia induced by TAC in mice. After miR-195 treatment, the protein expressions of Cavß1, Kir2.1 and Kv4.3 were decreased in mice. The results were consistent at animal and cellular levels, respectively. Luciferase assay results showed that miR-195 may directly target CACNB1, KCNJ2 and KCND3 to regulate the expression of Cavß1, Kir2.1 and Kv4.3 proteins. MiR-195 is involved in arrhythmia caused by cardiac hypertrophy by inhibiting Cavß1, Kir2.1 and Kv4.3.


Asunto(s)
Arritmias Cardíacas/etiología , Canales de Calcio/genética , Cardiomegalia/complicaciones , Cardiomegalia/genética , Regulación de la Expresión Génica , MicroARNs/genética , Canales de Potasio/genética , Animales , Arritmias Cardíacas/diagnóstico , Biomarcadores , Canales de Calcio/metabolismo , Cardiomegalia/diagnóstico , Modelos Animales de Enfermedad , Ecocardiografía , Técnica del Anticuerpo Fluorescente , Genes Reporteros , Vectores Genéticos/genética , Inmunohistoquímica , Ratones , Miocitos Cardíacos/metabolismo , Canales de Potasio/metabolismo , Isoformas de Proteínas , Transducción Genética , Regulación hacia Arriba
18.
J Pharmacol Exp Ther ; 373(3): 463-475, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32238453

RESUMEN

Leonurine (LEO) is a bioactive small molecular compound that has protective effects on the cardiovascular system and prevents the early progression of atherosclerosis; however, it is not clear whether LEO is effective for plaque stability. A novel mouse atherosclerosis model involving tandem stenosis (TS) of the right carotid artery combined with western diet (WD) feeding was used. Apolipoprotein E gene-deficient mice were fed with a WD and received LEO administration daily for 13 weeks. TS was introduced 6 weeks after the onset of experiments. We found that LEO enhanced plaque stability by increasing fibrous cap thickness and collagen content while decreasing the population of CD68-positive cells. Enhanced plaque stability by LEO was associated with the nitric oxide synthase (NOS)-nitric oxide (NO) system. LEO restored the balance between endothelial NOS(E)- and inducible NOS(iNOS)-derived NO production; suppressed the NF-κB signaling pathway; reduced the level of the inflammatory infiltration in plaque, including cytokine interleukin 6; and downregulated the expression of adhesion molecules. These findings support the distinct role of LEO in plaque stabilization. In vitro studies with oxidized low-density lipoprotein-challenged human umbilical vein endothelial cells revealed that LEO balanced NO production and inhibited NF-κB/P65 nuclear translocation, thus mitigating inflammation. In conclusion, the restored balance of the NOS-NO system and mitigated inflammation contribute to the plaque-stabilizing effect of LEO. SIGNIFICANCE STATEMENT: LEO restored the balance between endothelial NOS and inducible NOS in NO production and inhibited excessive inflammation in atherosclerotic "unstable" and rupture-prone plaques in apolipoprotein E gene-deficient mice. The protective effect of LEO for stabilizing atherosclerotic plaques was due to improved collagen content, increased fibrous cap thickness, and decreased accumulation of macrophages/foam cells. So far, LEO has passed the safety and feasibility test of phase I clinical trial.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Ácido Gálico/análogos & derivados , Inflamación/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Placa Aterosclerótica/tratamiento farmacológico , Animales , Aterosclerosis/metabolismo , Línea Celular , Ácido Gálico/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Placa Aterosclerótica/metabolismo , Transducción de Señal/efectos de los fármacos
19.
Basic Res Cardiol ; 115(2): 9, 2020 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-31900593

RESUMEN

Ventricular arrhythmia is the most common cause of sudden cardiac death in patients with myocardial infarction (MI). Fibroblast growth factor 21 (FGF21) has been shown to play an important role in cardiovascular and metabolic diseases. However, the effects of FGF21 on ventricular arrhythmias following MI have not been addressed yet. The present study was conducted to investigate the pharmacological action of FGF21 on ventricular arrhythmias after MI. Adult male mice were administrated with or without recombinant human basic FGF21 (rhbFGF21), and the susceptibility to arrhythmias was assessed by programmed electrical stimulation and optical mapping techniques. Here, we found that rhbFGF21 administration reduced the occurrence of ventricular tachycardia (VT), improved epicardial conduction velocity and shorted action potential duration at 90% (APD90) in infarcted mouse hearts. Mechanistically, FGF21 may improve cardiac electrophysiological remodeling as characterized by the decrease of INa and IK1 current density in border zone of infarcted mouse hearts. Consistently, in vitro study also demonstrated that FGF21 may rescue oxidant stress-induced dysfunction of INa and IK1 currents in cultured ventricular myocytes. We further found that oxidant stress-induced down-regulation of early growth response protein 1 (EGR1) contributed to INa and IK1 reduction in post-infarcted hearts, and FGF21 may recruit EGR1 into the SCN5A and KCNJ2 promoter regions to up-regulate NaV1.5 and Kir2.1 expression at transcriptional level. Moreover, miR-143 was identified as upstream of EGR1 and mediated FGF21-induced EGR1 up-regulation in cardiomyocytes. Collectively, rhbFGF21 administration effectively suppressed ventricular arrhythmias in post-infarcted hearts by regulating miR-143-EGR1-NaV1.5/Kir2.1 axis, which provides novel therapeutic strategies for ischemic arrhythmias in clinics.


Asunto(s)
Antiarrítmicos/administración & dosificación , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Factores de Crecimiento de Fibroblastos/administración & dosificación , Sistema de Conducción Cardíaco/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , MicroARNs/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Miocardio/metabolismo , Taquicardia Ventricular/prevención & control , Potenciales de Acción/efectos de los fármacos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Sistema de Conducción Cardíaco/metabolismo , Sistema de Conducción Cardíaco/fisiopatología , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/patología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Proteínas Recombinantes/administración & dosificación , Transducción de Señal , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología
20.
J Magn Reson Imaging ; 52(1): 197-206, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31755193

RESUMEN

BACKGROUND: Chronic pancreatitis (CP) is characterized by pancreatic fibrosis, in which a epithelial-mesenchymal transition (EMT)-like process is observed. However, few noninvasive approaches have been reported to evaluate pancreatic fibrosis and EMT in an animal model based on diffusion imaging. PURPOSE: To evaluate pancreatic fibrosis in CP by conventional diffusion-weighted imaging (DWI), intravoxel incoherent motion (IVIM), and diffusion kurtosis imaging (DKI) and then explore the correlation between diffusion parameters and the EMT markers in an animal model. STUDY TYPE: Prospective controlled imaging histological correlation. POPULATION: Forty-five rats with CP induced by injecting dibutyltin dichloride solution and 10 normal rats comprised the control group. FIELD STRENGTH/SEQUENCE: 11.7T MR, diffusion imaging with 10 b-values. ASSESSMENT: Apparent diffusion coefficient (ADC), IVIM-associated perfusion fraction (f), pseudodiffusion coefficient (D*), diffusion coefficient (D), DKI-associated mean kurtosis (MK), and mean corrected diffusion coefficient (MD) were quantitatively measured and correlated with pancreatic fibrosis stages as well as the EMT markers E-cadherin and α-smooth muscle actin (α-SMA) expression. The discriminative performance of diffusion parameters for staging fibrosis was compared. STATISTICAL TESTS: Spearman's correlation, Student's t-test, and a receiver operating characteristic curve was conducted for statistical analysis. RESULTS: ADC, D, and MD (r = -0.637, -0.688, and -0.535; P < 0.001) were negatively correlated with pancreatic fibrosis staging, but MK (r = 0.740, P < 0.001) had a positive correlation. ADC, D, MD, and MK were significantly correlated with α-SMA (r = -0.684, -0.728, -0.627, and 0.721, all P < 0.001), while MK was significantly correlated with E-cadherin (r = -0.606, P < 0.001). The area under the curve (AUC) was not significantly different (P > 0.05) among ADC (0.797, 0.816, 0.873), D (0.862, 0.810, 0.895), MD (0.767, 0.772, 0.801), and MK (0.836, 0.893, 0.951) for F1 or greater, F2 or greater, and F3 pancreatic fibrosis separately. DATA CONCLUSION: ADC, D, MD, and MK were helpful for assessing pancreatic fibrosis staging, and these diffusion parameters were also significantly correlated with the expression of EMT markers in pancreatic fibrosis. LEVEL OF EVIDENCE: 2 Technical Efficacy Stage: 2 J. Magn. Reson. Imaging 2020;52:197-206.


Asunto(s)
Transición Epitelial-Mesenquimal , Pancreatitis Crónica , Animales , Benchmarking , Imagen de Difusión por Resonancia Magnética , Fibrosis , Movimiento (Física) , Pancreatitis Crónica/inducido químicamente , Pancreatitis Crónica/diagnóstico por imagen , Estudios Prospectivos , Ratas , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA