Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 101(1): 119-130, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34757120

RESUMEN

Kidney fibrosis is associated with the progression of acute kidney injury to chronic kidney disease. MG53, a cell membrane repair protein, has been shown to protect against injury to kidney epithelial cells and acute kidney injury. Here, we evaluated the role of MG53 in modulation of kidney fibrosis in aging mice and in mice with unilateral ureteral obstruction (UUO) a known model of progressive kidney fibrosis. Mice with ablation of MG53 developed more interstitial fibrosis with age than MG53-intact mice of the same age. Similarly, in the absence of MG53, kidney fibrosis was exaggerated compared to mice with intact MG53 in the obstructed kidney compared to the contralateral unobstructed kidney or the kidneys of sham operated mice. The ureteral obstructed kidneys from MG53 deficient mice also showed significantly more inflammation than ureteral obstructed kidneys from MG53 intact mice. In vitro experiments demonstrated that MG53 could enter the nuclei of proximal tubular epithelial cells and directly interact with the p65 component of transcription factor NF-κB, providing a possible explanation of enhanced inflammation in the absence of MG53. To test this, enhanced MG53 expression through engineered cells or direct recombinant protein delivery was given to mice subject to UUO. This reduced NF-κB activation and inflammation and attenuated kidney fibrosis. Thus, MG53 may have a therapeutic role in treating chronic kidney inflammation and thereby provide protection against fibrosis that leads to the chronic kidney disease phenotype.


Asunto(s)
Lesión Renal Aguda , Obstrucción Ureteral , Lesión Renal Aguda/genética , Lesión Renal Aguda/prevención & control , Animales , Membrana Celular/metabolismo , Fibrosis , Riñón/patología , Proteínas de la Membrana/metabolismo , Ratones , FN-kappa B/metabolismo , Obstrucción Ureteral/metabolismo
2.
Circ Res ; 126(4): 417-435, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31805819

RESUMEN

RATIONALE: Trimeric intracellular cation (TRIC)-A and B are distributed to endoplasmic reticulum/sarcoplasmic reticulum intracellular Ca2+ stores. The crystal structure of TRIC has been determined, confirming the homotrimeric structure of a potassium channel. While the pore architectures of TRIC-A and TRIC-B are conserved, the carboxyl-terminal tail (CTT) domains of TRIC-A and TRIC-B are different from each other. Aside from its recognized role as a counterion channel that participates in excitation-contraction coupling of striated muscles, the physiological function of TRIC-A in heart physiology and disease has remained largely unexplored. OBJECTIVE: In cardiomyocytes, spontaneous Ca2+ waves, triggered by store overload-induced Ca2+ release mediated by the RyR2 (type 2 ryanodine receptor), develop extrasystolic contractions often associated with arrhythmic events. Here, we test the hypothesis that TRIC-A is a physiological component of RyR2-mediated Ca2+ release machinery that directly modulates store overload-induced Ca2+ release activity via CTT. METHODS AND RESULTS: We show that cardiomyocytes derived from the TRIC-A-/- (TRIC-A knockout) mice display dysregulated Ca2+ movement across sarcoplasmic reticulum. Biochemical studies demonstrate a direct interaction between CTT-A and RyR2. Modeling and docking studies reveal potential sites on RyR2 that show differential interactions with CTT-A and CTT-B. In HEK293 (human embryonic kidney) cells with stable expression of RyR2, transient expression of TRIC-A, but not TRIC-B, leads to apparent suppression of spontaneous Ca2+ oscillations. Ca2+ measurements using the cytosolic indicator Fura-2 and the endoplasmic reticulum luminal store indicator D1ER suggest that TRIC-A enhances Ca2+ leak across the endoplasmic reticulum by directly targeting RyR2 to modulate store overload-induced Ca2+ release. Moreover, synthetic CTT-A peptide facilitates RyR2 activity in lipid bilayer reconstitution system, enhances Ca2+ sparks in permeabilized TRIC-A-/- cardiomyocytes, and induces intracellular Ca2+ release after microinjection into isolated cardiomyocytes, whereas such effects were not observed with the CTT-B peptide. In response to isoproterenol stimulation, the TRIC-A-/- mice display irregular ECG and develop more fibrosis than the WT (wild type) littermates. CONCLUSIONS: In addition to the ion-conducting function, TRIC-A functions as an accessory protein of RyR2 to modulate sarcoplasmic reticulum Ca2+ handling in cardiac muscle.


Asunto(s)
Calcio/metabolismo , Canales Iónicos/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Señalización del Calcio , Cardiotónicos/farmacología , Electrocardiografía/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Fibrosis/genética , Fibrosis/fisiopatología , Células HEK293 , Corazón/efectos de los fármacos , Corazón/fisiopatología , Humanos , Canales Iónicos/química , Canales Iónicos/genética , Isoproterenol/farmacología , Ratones Noqueados , Simulación del Acoplamiento Molecular , Miocardio/citología , Unión Proteica , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/genética , Retículo Sarcoplasmático/metabolismo
3.
Pflugers Arch ; 473(3): 547-556, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33474637

RESUMEN

Trimeric intracellular cation (TRIC) channels have been identified as monovalent cation channels that are located in the ER/SR membrane. Two isoforms discovered in mammals are TRIC-A (TMEM38a) and TRIC-B (TMEM38b). TRIC-B ubiquitously expresses in all tissues, and TRIC-B-/- mice is lethal at the neonatal stage. TRIC-A mainly expresses in excitable cells. TRIC-A-/- mice survive normally but show abnormal SR Ca2+ handling in both skeletal and cardiac muscle cells. Importantly, TRIC-A mutations have been identified in human patients with stress-induced arrhythmia. In the past decade, important discoveries have been made to understand the structure and function of TRIC channels, especially its role in regulating intracellular Ca2+ homeostasis. In this review article, we focus on the potential roles of TRIC-A in regulating cardiac function, particularly its effects on intracellular Ca2+ signaling of cardiomyocytes and discuss the current knowledge gaps.


Asunto(s)
Señalización del Calcio/fisiología , Homeostasis/fisiología , Canales Iónicos/fisiología , Miocitos Cardíacos/metabolismo , Animales , Humanos
4.
Mol Cancer ; 20(1): 118, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34521423

RESUMEN

BACKGROUND: Cancer cells develop resistance to chemotherapeutic intervention by excessive formation of stress granules (SGs), which are modulated by an oncogenic protein G3BP2. Selective control of G3BP2/SG signaling is a potential means to treat non-small cell lung cancer (NSCLC). METHODS: Co-immunoprecipitation was conducted to identify the interaction of MG53 and G3BP2. Immunohistochemistry and live cell imaging were performed to visualize the subcellular expression or co-localization. We used shRNA to knock-down the expression MG53 or G3BP2 to test the cell migration and colony formation. The expression level of MG53 and G3BP2 in human NSCLC tissues was tested by western blot analysis. The ATO-induced oxidative stress model was used to examine the effect of rhMG53 on SG formation. Moue NSCLC allograft experiments were performed on wild type and transgenic mice with either knockout of MG53, or overexpression of MG53. Human NSCLC xenograft model in mice was used to evaluate the effect of MG53 overexpression on tumorigenesis. RESULTS: We show that MG53, a member of the TRIM protein family (TRIM72), modulates G3BP2 activity to control lung cancer progression. Loss of MG53 results in the progressive development of lung cancer in mg53-/- mice. Transgenic mice with sustained elevation of MG53 in the bloodstream demonstrate reduced tumor growth following allograft transplantation of mouse NSCLC cells. Biochemical assay reveals physical interaction between G3BP2 and MG53 through the TRIM domain of MG53. Knockdown of MG53 enhances proliferation and migration of NSCLC cells, whereas reduced tumorigenicity is seen in NSCLC cells with knockdown of G3BP2 expression. The recombinant human MG53 (rhMG53) protein can enter the NSCLC cells to induce nuclear translation of G3BP2 and block arsenic trioxide-induced SG formation. The anti-proliferative effect of rhMG53 on NSCLC cells was abolished with knockout of G3BP2. rhMG53 can enhance sensitivity of NSCLC cells to undergo cell death upon treatment with cisplatin. Tailored induction of MG53 expression in NSCLC cells suppresses lung cancer growth via reduced SG formation in a xenograft model. CONCLUSION: Overall, these findings support the notion that MG53 functions as a tumor suppressor by targeting G3BP2/SG activity in NSCLCs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/etiología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Gránulos de Estrés/metabolismo , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Neoplasias Pulmonares/patología , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Gránulos de Estrés/patología
5.
Pharmacol Res ; 169: 105617, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33872811

RESUMEN

Traditional Chinese multi-herb-combined prescriptions usually show better performance than a single agent since a group of effective compounds interfere multiple disease-relevant targets simultaneously. Huang-Lian-Jie-Du decoction is a remedy made of four herbs that are widely used to treat oral ulcers, gingivitis, and periodontitis. However, the active ingredients and underlying mechanisms are not clear. To address these questions, we prepared a water extract solution of Huang-Lian-Jie-Du decoction (HLJDD), called it as WEH (Water Extract Solution of HLJDD), and used it to treat LPS-induced systemic inflammation in mice. We observed that WEH attenuated inflammatory responses including reducing production of cytokines, chemokines and interferons (IFNs), further attenuating emergency myelopoiesis, and preventing mice septic lethality. Upon LPS stimulation, mice pretreated with WEH increased circulating Ly6C- patrolling and splenic Ly6C+ inflammatory monocytes. The acute myelopoiesis related transcriptional factor profile was rearranged by WEH. Mechanistically we confirmed that WEH interrupted LPS/TLR4/CD14 signaling-mediated downstream signaling pathways through its nine principal ingredients, which blocked LPS stimulated divergent signaling cascades, such as activation of NF-κB, p38 MAPK, and ERK1/2. We conclude that the old remedy blunts LPS-induced "danger" signal recognition and transduction process at multiple sites. To translate our findings into clinical applications, we refined the crude extract into a pure multicomponent drug by directly mixing these nine chemical entities, which completely reproduced the effect of protecting mice from lethal septic shock. Finally, we reduced a large number of compounds within a multi-herb water extract to seven-chemical combination that exhibited superior therapeutic efficacy compared with WEH.


Asunto(s)
Medicamentos Herbarios Chinos/uso terapéutico , Inflamación/tratamiento farmacológico , Monocitos/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Factores de Transcripción/efectos de los fármacos , Animales , Reprogramación Celular/efectos de los fármacos , Coptis chinensis , Medicamentos Herbarios Chinos/administración & dosificación , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Extractos Vegetales/administración & dosificación , Células RAW 264.7/efectos de los fármacos , Factores de Transcripción/metabolismo
6.
Acta Pharmacol Sin ; 41(11): 1457-1464, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32424239

RESUMEN

Mitsugumin 53 (MG53) is a tripartite motif family protein that has been reported to attenuate injury via membrane repair in different organs. Contrast-induced acute kidney injury (CI-AKI) is a common complication caused by the administration of iodinated contrast media (CM). While the cytotoxicity induced by CM leading to tubular cell death may be initiated by cell membrane damage, we wondered whether MG53 alleviates CI-AKI. This study was designed to investigate the effect of MG53 on CI-AKI and the underlying mechanism. A rat model of CI-AKI was established, and CI-AKI induced the translocation of MG53 from serum to injury sites on the renal proximal tubular (RPT) epithelia, as illustrated by immunoblot analysis and immunohistochemical staining. Moreover, pretreatment of rats with recombinant human MG53 protein (rhMG53, 2 mg/mL) alleviated iopromide-induced injury in the kidney, which was determined by measuring serum creatinine, blood urea nitrogen and renal histological changes. In vitro studies demonstrated that exposure of RPT cells to iopromide (20, 40, and 80 mg/mL) caused cell membrane injury and cell death, which were attenuated by rhMG53 (10 and 50 µg/mL). Mechanistically, MG53 translocated to the injury site on RPT cells and bound to phosphatidylserine to protect RPT cells from iopromide-induced injury. In conclusion, MG53 protects against CI-AKI through cell membrane repair and reducing cell apoptosis; therefore, rhMG53 might be a potential effective means to treat or prevent CI-AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Apoptosis/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Sustancias Protectoras/uso terapéutico , Proteínas de Motivos Tripartitos/uso terapéutico , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Animales , Membrana Celular/metabolismo , Células Epiteliales , Femenino , Humanos , Yohexol/análogos & derivados , Riñón/patología , Túbulos Renales Proximales/citología , Masculino , Fosfatidilserinas/metabolismo , Sustancias Protectoras/metabolismo , Ratas Endogámicas WKY , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Proteínas de Motivos Tripartitos/metabolismo
7.
Biomacromolecules ; 18(9): 2820-2829, 2017 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-28731675

RESUMEN

Following myocardial infarction (MI), degradation of extracellular matrix (ECM) by upregulated matrix metalloproteinases (MMPs) especially MMP-2 decreases tissue mechanical properties, leading to cardiac function deterioration. Attenuation of cardiac ECM degradation at the early stage of MI has the potential to preserve tissue mechanical properties, resulting in cardiac function increase. Yet the strategy for efficiently preventing cardiac ECM degradation remains to be established. Current preclinical approaches have shown limited efficacy because of low drug dosage allocated to the heart tissue, dose-limiting side effects, and cardiac fibrosis. To address these limitations, we have developed a MMP-2 inhibitor delivery system that can be specifically delivered into infarcted hearts at early stage of MI to efficiently prevent MMP-2-mediated ECM degradation. The system was based on an injectable, degradable, fast gelation, and thermosensitive hydrogel, and a MMP-2 specific inhibitor, peptide CTTHWGFTLC (CTT). The use of fast gelation hydrogel allowed to completely retain CTT in the heart tissue. The system was able to release low molecular weight CTT over 4 weeks possibly due to the strong hydrogen bonding between the hydrogel and CTT. The release kinetics was modulated by amount of CTT loaded into the hydrogel, and using chondroitin sulfate and heparin that can interact with CTT and the hydrogel. Both glycosaminoglycans augmented CTT release, while heparin more greatly accelerated the release. After it was injected into the infarcted hearts for 4 weeks, the released CTT efficiently prevented cardiac ECM degradation as it not only increased tissue thickness but also preserved collagen composition similar to that in the normal heart tissue. In addition, the delivery system significantly improved cardiac function. Importantly, the delivery system did not induce cardiac fibrosis. These results demonstrate that the developed MMP-2 inhibitor delivery system has potential to efficiently reduce adverse myocardial remodeling and improve cardiac function.


Asunto(s)
Portadores de Fármacos/síntesis química , Liberación de Fármacos , Hidrogeles/síntesis química , Inhibidores de la Metaloproteinasa de la Matriz/farmacocinética , Infarto del Miocardio/tratamiento farmacológico , Péptidos Cíclicos/farmacocinética , Animales , Sulfatos de Condroitina/química , Portadores de Fármacos/efectos adversos , Portadores de Fármacos/química , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Heparina/química , Humanos , Hidrogeles/efectos adversos , Hidrogeles/química , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/administración & dosificación , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Péptidos Cíclicos/administración & dosificación , Péptidos Cíclicos/uso terapéutico , Ratas , Ratas Sprague-Dawley
8.
Adv Exp Med Biol ; 982: 529-551, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28551805

RESUMEN

The kidney is a vital organ that demands an extraordinary amount of energy to actively maintain the body's metabolism, plasma hemodynamics, electrolytes and water homeostasis, nutrients reabsorption, and hormone secretion. Kidney is only second to the heart in mitochondrial count and oxygen consumption. As such, the health and status of the energy power house, the mitochondria, is pivotal to the health and proper function of the kidney. Mitochondria are heterogeneous and highly dynamic organelles and their functions are subject to complex regulations through modulation of its biogenesis, bioenergetics, dynamics and clearance within cell. Kidney diseases, either acute kidney injury (AKI) or chronic kidney disease (CKD), are important clinical issues and global public health concerns with high mortality rate and socioeconomic burden due to lack of effective therapeutic strategies to cure or retard the progression of the diseases. Mitochondria-targeted therapeutics has become a major focus for modern research with the belief that maintaining mitochondria homeostasis can prevent kidney pathogenesis and disease progression. A better understanding of the cellular and molecular events that govern mitochondria functions in health and disease will potentially lead to improved therapeutics development.


Asunto(s)
Metabolismo Energético , Enfermedades Renales/metabolismo , Riñón/metabolismo , Mitocondrias/metabolismo , Animales , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Metabolismo Energético/efectos de los fármacos , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/genética , Enfermedades Renales/fisiopatología , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Dinámicas Mitocondriales , Mitofagia , Transducción de Señal
9.
J Sci Food Agric ; 97(10): 3323-3332, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27981601

RESUMEN

BACKGROUND: Among active components in Rabdosia rubescens, oridonin has been considered a key component and the most valuable compound because it has a wide range of activities beneficial to human health. To produce a high-quality oridonin extract, an alternative hyphenated procedure involving an ultrasound-assisted and supercritical carbon dioxide (HSC-CO2 ) extraction method to extract oridonin from R. rubescens was developed in this study. Fictitious solubilities of oridonin in supercritical CO2 (SC-CO2 ) with ultrasound assistance were measured by using the dynamic method at temperatures ranging from 305.15 K to 342.15 K over a pressure range of 11.5 to 33.5 MPa. RESULTS: Fictitious solubilities of oridonin at different temperatures and pressures were over the range of 2.13 × 10-6 to 10.09 × 10-6 (mole fraction) and correlated well with the density-based models, including the Bartle model, the Chrastil model, the Kumar and Johnston model and the Mendez-Santiago and Teja model, with overall average absolute relative deviations (AARDs) of 6.29%, 4.39%, 3.12% and 5.07%, respectively. CONCLUSION: Oridonin exhibits retrograde solubility behaviour in the supercritical state. Fictitious solubility data were further determined and obtained a good fit with four semi-empirical models. Simultaneously, the values of the total heat of solution, vaporisation and solvation of oridonin were estimated. © 2016 Society of Chemical Industry.


Asunto(s)
Cromatografía con Fluido Supercrítico/métodos , Diterpenos de Tipo Kaurano/análisis , Diterpenos de Tipo Kaurano/aislamiento & purificación , Isodon/química , Extractos Vegetales/análisis , Extractos Vegetales/aislamiento & purificación , Ultrasonido/métodos , Solubilidad
10.
J Biol Chem ; 290(22): 13830-9, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25869134

RESUMEN

Zinc is an essential trace element that participates in a wide range of biological functions, including wound healing. Although Zn(2+) deficiency has been linked to compromised wound healing and tissue repair in human diseases, the molecular mechanisms underlying Zn(2+)-mediated tissue repair remain unknown. Our previous studies established that MG53, a TRIM (tripartite motif) family protein, is an essential component of the cell membrane repair machinery. Domain homology analysis revealed that MG53 contains two Zn(2+)-binding motifs. Here, we show that Zn(2+) binding to MG53 is indispensable to assembly of the cell membrane repair machinery. Live cell imaging illustrated that Zn(2+) entry from extracellular space is essential for translocation of MG53-containing vesicles to the acute membrane injury sites for formation of a repair patch. The effect of Zn(2+) on membrane repair is abolished in mg53(-/-) muscle fibers, suggesting that MG53 functions as a potential target for Zn(2+) during membrane repair. Mutagenesis studies suggested that both RING and B-box motifs of MG53 constitute Zn(2+)-binding domains that contribute to MG53-mediated membrane repair. Overall, this study establishes a base for Zn(2+) interaction with MG53 in protection against injury to the cell membrane.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Zinc/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular , Reparación del ADN , Electrodos , Escherichia coli/metabolismo , Humanos , Proteínas de la Membrana , Ratones , Ratones Transgénicos , Microscopía Confocal , Músculo Esquelético/metabolismo , Mutación , Oxidación-Reducción , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Medicina Regenerativa , Transducción de Señal , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/metabolismo , Cicatrización de Heridas
11.
J Biol Chem ; 290(40): 24592-603, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26306047

RESUMEN

Cell membrane repair is an important aspect of physiology, and disruption of this process can result in pathophysiology in a number of different tissues, including wound healing, chronic ulcer and scarring. We have previously identified a novel tripartite motif family protein, MG53, as an essential component of the cell membrane repair machinery. Here we report the functional role of MG53 in the modulation of wound healing and scarring. Although MG53 is absent from keratinocytes and fibroblasts, remarkable defects in skin architecture and collagen overproduction are observed in mg53(-/-) mice, and these animals display delayed wound healing and abnormal scarring. Recombinant human MG53 (rhMG53) protein, encapsulated in a hydrogel formulation, facilitates wound healing and prevents scarring in rodent models of dermal injuries. An in vitro study shows that rhMG53 protects against acute injury to keratinocytes and facilitates the migration of fibroblasts in response to scratch wounding. During fibrotic remodeling, rhMG53 interferes with TGF-ß-dependent activation of myofibroblast differentiation. The resulting down-regulation of α smooth muscle actin and extracellular matrix proteins contributes to reduced scarring. Overall, these studies establish a trifunctional role for MG53 as a facilitator of rapid injury repair, a mediator of cell migration, and a modulator of myofibroblast differentiation during wound healing. Targeting the functional interaction between MG53 and TGF-ß signaling may present a potentially effective means for promoting scarless wound healing.


Asunto(s)
Proteínas Portadoras/fisiología , Membrana Celular/metabolismo , Proteínas Musculares/fisiología , Proteínas de Transporte Vesicular/fisiología , Cicatrización de Heridas/fisiología , Células 3T3 , Actinas/metabolismo , Animales , Diferenciación Celular , Movimiento Celular , Cicatriz/patología , Colágeno Tipo I/metabolismo , Fibroblastos/citología , Fibronectinas/metabolismo , Fibrosis/patología , Regulación de la Expresión Génica , Humanos , Hidrogeles/química , Queratinocitos/metabolismo , Proteínas de la Membrana , Ratones , Músculo Liso/metabolismo , Miofibroblastos/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Piel/patología , Proteínas de Motivos Tripartitos
12.
J Biol Chem ; 290(6): 3377-89, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25480788

RESUMEN

Postnatal skeletal muscle mass is regulated by the balance between anabolic protein synthesis and catabolic protein degradation, and muscle atrophy occurs when protein homeostasis is disrupted. Autophagy has emerged as critical in clearing dysfunctional organelles and thus in regulating protein turnover. Here we show that endolysosomal two-pore channel subtype 2 (TPC2) contributes to autophagy signaling and protein homeostasis in skeletal muscle. Muscles derived from Tpcn2(-/-) mice exhibit an atrophic phenotype with exacerbated autophagy under starvation. Compared with wild types, animals lacking TPC2 demonstrated an enhanced autophagy flux characterized by increased accumulation of autophagosomes upon combined stress induction by starvation and colchicine treatment. In addition, deletion of TPC2 in muscle caused aberrant lysosomal pH homeostasis and reduced lysosomal protease activity. Association between mammalian target of rapamycin and TPC2 was detected in skeletal muscle, allowing for appropriate adjustments to cellular metabolic states and subsequent execution of autophagy. TPC2 therefore impacts mammalian target of rapamycin reactivation during the process of autophagy and contributes to maintenance of muscle homeostasis.


Asunto(s)
Autofagia , Canales de Calcio/metabolismo , Músculo Esquelético/metabolismo , Transducción de Señal , Animales , Canales de Calcio/genética , Homeostasis , Concentración de Iones de Hidrógeno , Lisosomas/metabolismo , Lisosomas/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Péptido Hidrolasas/metabolismo , Fagosomas/metabolismo , Fagosomas/ultraestructura , Estrés Fisiológico , Serina-Treonina Quinasas TOR/metabolismo
13.
Circ Res ; 114(4): 706-16, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24526676

RESUMEN

Trimeric intracellular cation channels (TRIC) represents a novel class of trimeric intracellular cation channels. Two TRIC isoforms have been identified in both the human and the mouse genomes: TRIC-A, a subtype predominantly expressed in the sarcoplasmic reticulum (SR) of muscle cells, and TRIC-B, a ubiquitous subtype expressed in the endoplasmic reticulum (ER) of all tissues. Genetic ablation of either TRIC-A or TRIC-B leads to compromised K(+) permeation and Ca(2+) release across the SR/ER membrane, supporting the hypothesis that TRIC channels provide a counter balancing K(+) flux that reduces SR/ER membrane depolarization for maintenance of the electrochemical gradient that drives SR/ER Ca(2+) release. TRIC-A and TRIC-B seem to have differential functions in Ca(2+) signaling in excitable and nonexcitable cells. Tric-a(-/-) mice display defective Ca(2+) sparks and spontaneous transient outward currents in arterial smooth muscle and develop hypertension, in addition to skeletal muscle dysfunction. Knockout of TRIC-B results in abnormal IP3 receptor-mediated Ca(2+) release in airway epithelial cells, respiratory defects, and neonatal lethality. Double knockout mice lacking both TRIC-A and TRIC-B show embryonic lethality as a result of cardiac arrest. Such an aggravated lethality indicates that TRIC-A and TRIC-B share complementary physiological functions in Ca(2+) signaling in embryonic cardiomyocytes. Tric-a(-/-) and Tric-b(+/-) mice are viable and susceptible to stress-induced heart failure. Recent evidence suggests that TRIC-A directly modulates the function of the cardiac ryanodine receptor 2 Ca(2+) release channel, which in turn controls store-overload-induced Ca(2+) release from the SR. Thus, the TRIC channels, in addition to providing a countercurrent for SR/ER Ca(2+) release, may also function as accessory proteins that directly modulate the ryanodine receptor/IP3 receptor channel functions.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Retículo Endoplásmico/fisiología , Canales Iónicos/metabolismo , Retículo Sarcoplasmático/fisiología , Animales , Homeostasis/fisiología , Humanos , Canales Iónicos/genética , Ratones
14.
Int J Mol Sci ; 17(5)2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27153058

RESUMEN

Kidney is a vital organ with high energy demands to actively maintain plasma hemodynamics, electrolytes and water homeostasis. Among the nephron segments, the renal tubular epithelium is endowed with high mitochondria density for their function in active transport. Acute kidney injury (AKI) is an important clinical syndrome and a global public health issue with high mortality rate and socioeconomic burden due to lack of effective therapy. AKI results in acute cell death and necrosis of renal tubule epithelial cells accompanied with leakage of tubular fluid and inflammation. The inflammatory immune response triggered by the tubular cell death, mitochondrial damage, associative oxidative stress, and the release of many tissue damage factors have been identified as key elements driving the pathophysiology of AKI. Autophagy, the cellular mechanism that removes damaged organelles via lysosome-mediated degradation, had been proposed to be renoprotective. An in-depth understanding of the intricate interplay between autophagy and innate immune response, and their roles in AKI pathology could lead to novel therapies in AKI. This review addresses the current pathophysiology of AKI in aspects of mitochondrial dysfunction, innate immunity, and molecular mechanisms of autophagy. Recent advances in renal tissue regeneration and potential therapeutic interventions are also discussed.


Asunto(s)
Lesión Renal Aguda/metabolismo , Autofagia , Inmunidad Innata , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/patología , Lesión Renal Aguda/terapia , Animales , Apoptosis , Humanos , Regeneración
15.
J Mol Cell Cardiol ; 80: 10-19, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25533937

RESUMEN

Ischemic heart disease is a leading cause of death in human population and protection of myocardial infarction (MI) associated with ischemia-reperfusion (I/R) remains a challenge. MG53 is an essential component of the cell membrane repair machinery that protects injury to the myocardium. We investigated the therapeutic value of using the recombinant human MG53 (rhMG53) protein for treatment of MI. Using Langendorff perfusion of isolated mouse heart, we found that I/R caused injury to cardiomyocytes and release of endogenous MG53 into the extracellular solution. rhMG53 protein was applied to the perfusion solution concentrated at injury sites on cardiomyocytes to facilitate cardioprotection. With rodent models of I/R-induced MI, we established the in vivo dosing range for rhMG53 in cardioprotection. Using a porcine model of angioplasty-induced MI, the cardioprotective effect of rhMG53 was evaluated. Intravenous administration of rhMG53, either prior to or post-ischemia, reduced infarct size and troponin I release in the porcine model when examined at 24h post-reperfusion. Echocardiogram and histological analyses revealed that the protective effects of rhMG53 observed following acute MI led to long-term improvement in cardiac structure and function in the porcine model when examined at 4weeks post-operation. Our study supports the concept that rhMG53 could have potential therapeutic value for treatment of MI in human patients with ischemic heart diseases.


Asunto(s)
Cardiotónicos/farmacología , Proteínas Portadoras/farmacología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Proteínas Recombinantes/farmacología , Animales , Cardiotónicos/administración & dosificación , Cardiotónicos/farmacocinética , Proteínas Portadoras/administración & dosificación , Proteínas Portadoras/farmacocinética , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Técnicas In Vitro , Masculino , Ratones , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/mortalidad , Miocardio/metabolismo , Miocardio/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Transducción de Señal/efectos de los fármacos , Porcinos , Proteínas de Motivos Tripartitos
16.
Nature ; 459(7246): 596-600, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19387438

RESUMEN

Ca(2+) mobilization from intracellular stores represents an important cell signalling process that is regulated, in mammalian cells, by inositol-1,4,5-trisphosphate (InsP(3)), cyclic ADP ribose and nicotinic acid adenine dinucleotide phosphate (NAADP). InsP(3) and cyclic ADP ribose cause the release of Ca(2+) from sarcoplasmic/endoplasmic reticulum stores by the activation of InsP(3) and ryanodine receptors (InsP(3)Rs and RyRs). In contrast, the nature of the intracellular stores targeted by NAADP and the molecular identity of the NAADP receptors remain controversial, although evidence indicates that NAADP mobilizes Ca(2+) from lysosome-related acidic compartments. Here we show that two-pore channels (TPCs) comprise a family of NAADP receptors, with human TPC1 (also known as TPCN1) and chicken TPC3 (TPCN3) being expressed on endosomal membranes, and human TPC2 (TPCN2) on lysosomal membranes when expressed in HEK293 cells. Membranes enriched with TPC2 show high affinity NAADP binding, and TPC2 underpins NAADP-induced Ca(2+) release from lysosome-related stores that is subsequently amplified by Ca(2+)-induced Ca(2+) release by InsP(3)Rs. Responses to NAADP were abolished by disrupting the lysosomal proton gradient and by ablating TPC2 expression, but were only attenuated by depleting endoplasmic reticulum Ca(2+) stores or by blocking InsP(3)Rs. Thus, TPCs form NAADP receptors that release Ca(2+) from acidic organelles, which can trigger further Ca(2+) signals via sarcoplasmic/endoplasmic reticulum. TPCs therefore provide new insights into the regulation and organization of Ca(2+) signals in animal cells, and will advance our understanding of the physiological role of NAADP.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Calcio/metabolismo , NADP/análogos & derivados , Orgánulos/metabolismo , Animales , Canales de Calcio/genética , Señalización del Calcio/efectos de los fármacos , Línea Celular , Pollos , Humanos , Concentración de Iones de Hidrógeno , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , NADP/metabolismo , NADP/farmacología , Orgánulos/efectos de los fármacos , Unión Proteica
17.
J Biol Chem ; 288(4): 2103-9, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23223241

RESUMEN

Functional coupling between inositol (1,4,5)-trisphosphate receptor (IP(3)R) and ryanodine receptor (RyR) represents a critical component of intracellular Ca(2+) signaling in many excitable cells; however, the role of this mechanism in skeletal muscle remains elusive. In skeletal muscle, RyR-mediated Ca(2+) sparks are suppressed in resting conditions, whereas application of transient osmotic stress can trigger activation of Ca(2+) sparks that are restricted to the periphery of the fiber. Here we show that onset of these spatially confined Ca(2+) sparks involves interaction between activation of IP(3)R and RyR near the sarcolemmal membrane. Pharmacological prevention of IP(3) production or inhibition of IP(3)R channel activity abolishes stress-induced Ca(2+) sparks in skeletal muscle. Although genetic ablation of the type 2 IP(3)R does not appear to affect Ca(2+) sparks in skeletal muscle, specific silencing of the type 1 IP(3)R leads to ablation of stress-induced Ca(2+) sparks. Our data indicate that membrane-delimited signaling involving cross-talk between IP(3)R1 and RyR1 contributes to Ca(2+) spark activation in skeletal muscle.


Asunto(s)
Calcio/metabolismo , Regulación de la Expresión Génica , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Calcio/química , Señalización del Calcio , Ratones , Microscopía Confocal/métodos , Modelos Biológicos , Modelos Genéticos , Ósmosis , Técnicas de Placa-Clamp , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal
18.
Opt Express ; 22(10): 12160-76, 2014 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-24921337

RESUMEN

One key factor that limits resolution of single-molecule superresolution microscopy relates to the localization accuracy of the activated emitters, which is usually deteriorated by two factors. One originates from the background noise due to out-of-focus signals, sample auto-fluorescence, and camera acquisition noise; and the other is due to the low photon count of emitters at a single frame. With fast acquisition rate, the activated emitters can last multiple frames before they transiently switch off or permanently bleach. Effectively incorporating the temporal information of these emitters is critical to improve the spatial resolution. However, majority of the existing reconstruction algorithms locate the emitters frame by frame, discarding or underusing the temporal information. Here we present a new image reconstruction algorithm based on tracklets, short trajectories of the same objects. We improve the localization accuracy by associating the same emitters from multiple frames to form tracklets and by aggregating signals to enhance the signal to noise ratio. We also introduce a weighted mean-shift algorithm (WMS) to automatically detect the number of modes (emitters) in overlapping regions of tracklets so that not only well-separated single emitters but also individual emitters within multi-emitter groups can be identified and tracked. In combination with a maximum likelihood estimator method (MLE), we are able to resolve low to medium density of overlapping emitters with improved localization accuracy. We evaluate the performance of our method with both synthetic and experimental data, and show that the tracklet-based reconstruction is superior in localization accuracy, particularly for weak signals embedded in a strong background. Using this method, for the first time, we resolve the transverse tubule structure of the mammalian skeletal muscle.


Asunto(s)
Algoritmos , Procesamiento de Imagen Asistido por Computador , Músculo Esquelético/citología , Nanotecnología/métodos , Fotones , Animales , Células Cultivadas , Relación Señal-Ruido
19.
J Biol Chem ; 287(40): 33523-32, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22872646

RESUMEN

Of the TRIM/RBCC family proteins taking part in a variety of cellular processes, TRIM50 is a stomach-specific member with no defined biological function. Our biochemical data demonstrated that TRIM50 is specifically expressed in gastric parietal cells and is predominantly localized in the tubulovesicular and canalicular membranes. In cultured cells ectopically expressing GFP-TRIM50, confocal microscopic imaging revealed dynamic movement of TRIM50-associated vesicles in a phosphoinositide 3-kinase-dependent manner. A protein overlay assay detected preferential binding of the PRY-SPRY domain from the TRIM50 C-terminal region to phosphatidylinositol species, suggesting that TRIM50 is involved in vesicular dynamics by sensing the phosphorylated state of phosphoinositol lipids. Trim50 knock-out mice retained normal histology in the gastric mucosa but exhibited impaired secretion of gastric acid. In response to histamine, Trim50 knock-out parietal cells generated deranged canaliculi, swollen microvilli lacking actin filaments, and excess multilamellar membrane complexes. Therefore, TRIM50 seems to play an essential role in tubulovesicular dynamics, promoting the formation of sophisticated canaliculi and microvilli during acid secretion in parietal cells.


Asunto(s)
Ácidos/química , Mucosa Gástrica/metabolismo , Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Células Parietales Gástricas/citología , Animales , Linfocitos/citología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microvellosidades/metabolismo , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositoles/química , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Ratas Wistar
20.
FASEB J ; 26(5): 1875-83, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22253476

RESUMEN

Repair of injury to the plasma membrane is an essential mechanism for maintenance of cellular homeostasis and integrity that involves coordinated movement of intracellular vesicles to membrane injury sites to facilitate patch formation. We have previously identified MG53 as an essential component of the cell membrane repair machinery. In order for MG53 and intracellular vesicles to translocate to membrane injury sites, motor proteins must be involved. Here, we show that nonmuscle myosin type IIA (NM-IIA) interacts with MG53 to regulate vesicle trafficking during cell membrane repair. In cells that are deficient for NM-IIA expression, MG53 cannot translocate to acute injury sites, whereas rescue of NM-IIA expression in these cells can restore MG53-mediated membrane repair. Compromised cell membrane repair is observed in cells with RNAi-mediated knockdown of NM-IIA expression, or following pharmacological alteration of NM-IIA motor function. Together, our data reveal NM-IIA as a key cytoskeleton motor protein that facilitates vesicle trafficking during MG53-mediated cell membrane repair.


Asunto(s)
Proteínas Portadoras/fisiología , Miosina Tipo IIA no Muscular/fisiología , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular , Membrana Celular/fisiología , Chlorocebus aethiops , Cartilla de ADN , Humanos , Datos de Secuencia Molecular , Miosina Tipo IIA no Muscular/química , Miosina Tipo IIA no Muscular/genética , Transporte de Proteínas , Interferencia de ARN , Proteínas de Motivos Tripartitos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA