Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Cell Physiol ; 323(6): C1860-C1871, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36374170

RESUMEN

Mutations in tripartite motif-containing protein 32 (TRIM32), especially in NHL repeats, have been found in skeletal muscle in patients with type 2H limb-girdle muscular dystrophy (LGMD2H). However, the roles of the NHL repeats of TRIM32 in skeletal muscle functions have not been well addressed. In the present study, to examine the functional role(s) of the TRIM32 NHL repeats in skeletal muscle, TRIM32-binding proteins in skeletal muscle were first searched using a binding assay and MALDI-TOF/TOF. Sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 1a (SERCA1a) was found to be a TRIM32-binding protein. Next, a deletion mutant of TRIM32 missing the NHL repeats (NHL-Del) was expressed in mouse primary skeletal myotubes during myoblast differentiation into myotubes. Ca2+ movement in the myotubes was examined using single-cell Ca2+ imaging. Unlike wild-type (WT) TRIM32, NHL-Del did not enhance the amount of Ca2+ release from the sarcoplasmic reticulum (SR), Ca2+ release for excitation-contraction (EC) coupling, or extracellular Ca2+ entry via store-operated Ca2+ entry (SOCE). In addition, even compared with the vector control, NHL-Del resulted in reduced SOCE due to reduced expression of extracellular Ca2+ entry channels. Transmission electron microscopy (TEM) observation of the myotubes revealed that NHL-Del induced the formation of abnormal vacuoles and tubular structures in the cytosol. Therefore, by binding to SERCA1a via its NHL repeats, TRIM32 may participate in the regulation of Ca2+ movement for skeletal muscle contraction and the formation of cellular vacuoles and tubular structures in skeletal muscle. Functional defects in TRIM32 due to mutations in NHL repeats may be pathogenic toward LGMD2H.


Asunto(s)
Calcio , Músculo Esquelético , Distrofia Muscular de Cinturas , Secuencias Repetitivas de Aminoácido , Animales , Ratones , Calcio/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/patología , Retículo Sarcoplasmático/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Secuencias Repetitivas de Aminoácido/genética , Secuencias Repetitivas de Aminoácido/fisiología
2.
J Korean Med Sci ; 36(14): e107, 2021 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-33847085

RESUMEN

Coronavirus disease 2019 vaccinations for healthcare workers (HCWs) have begun in South Korea. To investigate adverse events (AEs) of the first dose of each vaccine, any symptom was collected daily for seven days after vaccination in a tertiary hospital. We found that 1,301 of 1,403 ChAdOx1 nCoV-19 recipients and 38 of 80 BNT162b2 recipients reported AEs respectively (90.9% vs. 52.5%): injection-site pain (77.7% vs. 51.2%), myalgia (60.5% vs. 11.2%), fatigue (50.7% vs. 7.5%), headache (47.4% vs. 7.5%), and fever (36.1% vs. 5%; P < 0.001 for all). Young HCWs reported more AEs with ChAdOx1 nCoV-19 than with BNT162b2. No incidences of anaphylaxis were observed. Only one serious AE required hospitalization for serious vomiting, and completely recovered. In conclusion, reported AEs were more common in recipients with ChAdOx1 nCoV-19 than in those with BNT162b2. However, most of the reported AEs were mild to moderate in severity. Sufficient explanation and preparation for expected AEs required to promote widespread vaccination.


Asunto(s)
Vacunas contra la COVID-19/efectos adversos , COVID-19/prevención & control , Personal de Salud , Adulto , Vacuna BNT162 , ChAdOx1 nCoV-19 , Femenino , Hospitalización , Humanos , Incidencia , Masculino , Persona de Mediana Edad , República de Corea/epidemiología , Centros de Atención Terciaria , Vacunación/efectos adversos
3.
J Cell Physiol ; 234(4): 3837-3849, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30256387

RESUMEN

Transforming growth factor-ß (TGF-ß) is a multifunctional cytokine that is known to modulate various aspects of endothelial cell (EC) biology. Retinal pigment epithelium (RPE) is important for regulating angiogenesis of choriocapillaris and one of the main cell sources of TGF-ß secretion, particularly TGF-ß2. However, it is largely unclear whether and how TGF-ß2 affects angiogenic responses of ECs. In the current study, we demonstrated that TGF-ß2 reduces vascular endothelial growth factor receptor-2 (VEGFR-2) expression in ECs and thereby inhibits vascular endothelial growth factor (VEGF) signaling and VEGF-induced angiogenic responses such as EC migration and tube formation. We also demonstrated that the reduction of VEGFR-2 expression by TGF-ß2 is due to the suppression of JNK signaling. In coculture of RPE cells and ECs, RPE cells decreased VEGFR-2 levels in ECs and EC migration. In addition, we showed that TGF-ß2 derived from RPE cells is involved in the reduction of VEGFR-2 expression and inhibition of EC migration. These results suggest that TGF-ß2 plays an important role in inhibiting the angiogenic responses of ECs during the interaction between RPE cells and ECs and that angiogenic responses of ECs may be amplified by a decrease in TGF-ß2 expression in RPE cells under pathologic conditions.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica , Comunicación Paracrina , Epitelio Pigmentado de la Retina/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Regulación hacia Abajo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fosforilación , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Vías Secretoras , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
4.
FASEB J ; 32(5): 2324-2338, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29269397

RESUMEN

Pericytes (PCs) are crucial in maintaining the quiescence of endothelial cells (ECs) and the integrity of EC tight junctions. Especially in diabetic retinopathy (DR), PC loss is one of the early pathologic changes in capillaries of diabetic retinas. Thus, preventing PC loss is beneficial for attenuating vision impairment in patients with DR. Although many studies have revealed the mechanism of PC loss in retinas, little is known about the mechanisms that increase PC survival. We focused on the effect of ß-adrenergic receptor agonists (ß-agonists) on PC loss in diabetic retinas. In this study, ß-agonists increased the cell viability of PCs by increasing PC survival and proliferation. Mechanistically, ß-agonist-induced protein kinase B activation in PCs reduced PC apoptosis in response to various stimuli. ß2-agonists more potently increased PC survival than ß1-agonists. ß2-Agonist reduced vascular leakage and PC loss in retinas of mice with streptozotocin-induced diabetes. In cocultures of PCs and ECs, ß2-agonists restored the altered permeability and ZO-1 expression in ECs induced by PC loss. We concluded that ß-agonists, especially ß2-agonists, increase PC survival, thereby preventing diabetes-induced PC loss in retinas. These results provide a potential therapeutic benefit of ß-agonists for preventing PC loss in DR.-Yun, J.-H., Jeong, H.-S., Kim, K.-J., Han, M. H., Lee, E. H., Lee, K., Cho, C.-H. ß-Adrenergic receptor agonists attenuate pericyte loss in diabetic retinas through Akt activation.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Retinopatía Diabética/tratamiento farmacológico , Pericitos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Retina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Ratones , Pericitos/patología , Retina/patología , Proteína de la Zonula Occludens-1/biosíntesis
5.
Biochem Biophys Res Commun ; 503(4): 2792-2799, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30100069

RESUMEN

Retinopathy of prematurity (ROP) is an eye disease that causes blindness due to delayed vascular growth, retinal ischemia, and resulting abnormal angiogenesis. Nonselective ß-antagonist propranolol is in clinical trials for the treatment of ROP due to its effect of reducing VEGF expression and inhibiting retinal angiogenesis in oxygen-induced ROP models (OIR), but the mechanism by which propranolol acts on ROP vessels is still unclear. In the present study, we have focused on the effect of propranolol on pericyte survival and vascular permeability. We demonstrated that propranolol increases pericyte apoptosis more sensitively than endothelial cells (ECs), thereby weakening EC tight junctions to increase endothelial permeability in co-cultures of pericytes and ECs. Mechanistically, pericyte apoptosis by propranolol was due to the inhibition of Akt signaling pathway. We also demonstrated that propranolol increases pericyte loss and vascular permeability of retinal vessels in a mouse model of OIR. These results suggest that propranolol may be negative for blood vessels in retinas of OIR, and that the efficacy of propranolol for the treatment of ROP needs to be more thoroughly verified.


Asunto(s)
Apoptosis/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Hiperoxia/inducido químicamente , Propranolol/farmacología , Retinopatía de la Prematuridad/inducido químicamente , Vasodilatadores/farmacología , Animales , Animales Recién Nacidos , Apoptosis/genética , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hiperoxia/genética , Hiperoxia/metabolismo , Hiperoxia/patología , Ratones , Ratones Endogámicos C57BL , Oxígeno/administración & dosificación , Pericitos/citología , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neovascularización Retiniana/inducido químicamente , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Retinopatía de la Prematuridad/genética , Retinopatía de la Prematuridad/metabolismo , Retinopatía de la Prematuridad/patología , Transducción de Señal , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
J Cell Physiol ; 232(5): 1123-1134, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27580405

RESUMEN

Vascular inflammation is characteristic feature of diabetic retinopathy. In diabetic retina, a variety of the pro-inflammatory cytokines are elevated and involved in endothelial dysfunction. STAT3 transcription factor has been implicated in mediating cytokine signaling during vascular inflammation. However, whether and how STAT3 is involved in the direct regulation of the endothelial permeability is currently undefined. Our studies revealed that IL-6-induced STAT3 activation increases retinal endothelial permeability and vascular leakage in retinas of mice through the reduced expression of the tight junction proteins ZO-1 and occludin. In a co-culture model with microglia and endothelial cells under a high glucose condition, the microglia-derived IL-6 induced STAT3 activation in the retinal endothelial cells, leading to increasing endothelial permeability. In addition, IL-6-induced STAT3 activation was independent of ROS generation in the retinal endothelial cells. Moreover, we demonstrated that STAT3 activation downregulates the ZO-1 and occludin levels and increases the endothelial permeability through the induction of VEGF production in retinal endothelial cells. These results suggest the potential importance of IL-6/STAT3 signaling in regulating endothelial permeability and provide a therapeutic target to prevent the pathology of diabetic retinopathy. J. Cell. Physiol. 232: 1123-1134, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Regulación hacia Abajo , Células Endoteliales/metabolismo , Ocludina/metabolismo , Vasos Retinianos/patología , Factor de Transcripción STAT3/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Animales , Línea Celular , Permeabilidad de la Membrana Celular , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Glucosa/toxicidad , Humanos , Interleucina-6/metabolismo , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Retina/efectos de los fármacos , Retina/patología , Vasos Retinianos/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
J Am Chem Soc ; 139(22): 7432-7435, 2017 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-28538099

RESUMEN

We report here a synthetic ion channel developed from a shape-persistent porphyrin-based covalent organic cage. The cage was synthesized by employing a synthetically economical dynamic covalent chemistry (DCC) approach. The organic cage selectively transports biologically relevant iodide ions over other inorganic anions by a dehydration-driven, channel mechanism as evidenced by vesicle-based fluorescence assays and planar lipid bilayer-based single channel recordings. Furthermore, the organic cage appears to facilitate iodide transport across the membrane of a living cell, suggesting that the cage could be useful as a biological tool that may replace defective iodide channels in living systems.

8.
Biochem Biophys Res Commun ; 464(1): 133-9, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26141232

RESUMEN

Mitsugumin 29 (MG29) is related to the fatigue and aging processes of skeletal muscle. To examine the roles of MG29 in conjunction with its binding protein, the canonical-type transient receptor potential cation channel 3 (TRPC3), in skeletal muscle, the binding region of MG29 to TRPC3 was studied along with the functional relevance of the binding in mouse primary skeletal myotubes using co-immunoprecipitation assays and Ca(2+) imaging experiments. The N-terminus and the I-II loop of MG29 constitute the binding region for TRPC3. The myotubes that expressed the MG29 mutant missing the entire TRPC3-binding region showed a disrupted binding between endogenous MG29 and TRPC3 and a reduction in Ca(2+) transients in response to membrane depolarization without affecting ryanodine receptor 1 activity, the resting cytosolic Ca(2+) level, and the amount of releasable Ca(2+) from the sarcoplasmic reticulum. Among the proteins mediating Ca(2+) movements in skeletal muscle, TRPC4 expression was significantly decreased by the MG29 mutant. Therefore, MG29 could be a new factor for regulating Ca(2+) transients during skeletal muscle contraction possibly via a correlation with TRPC3 and TRPC4.


Asunto(s)
Calcio/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Retículo Sarcoplasmático/metabolismo , Sinaptofisina/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Sitios de Unión , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Transporte Iónico , Ratones , Imagen Molecular , Fibras Musculares Esqueléticas/citología , Proteínas Musculares/genética , Unión Proteica , Estructura Secundaria de Proteína , Conejos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal , Sinaptofisina/genética , Canales Catiónicos TRPC/genética
9.
Pflugers Arch ; 466(5): 987-1001, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24077737

RESUMEN

Stromal interaction molecule 1 (STIM1) mediates Ca2+ movements from the extracellular space to the cytosol through a store-operated Ca2+ entry (SOCE) mechanism in various cells including skeletal muscle cells. In the present study, to reveal the unidentified functional role of the STIM1 C terminus from 449 to 671 amino acids in skeletal muscle, binding assays and quadrupole time-of-flight mass spectrometry were used to identify proteins binding in this region along with proteins that mediate skeletal muscle contraction and relaxation. STIM1 binds to sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 1a (SERCA1a) via this region (called STIM1-SBR). The binding was confirmed in endogenous full-length STIM1 in rabbit skeletal muscle and mouse primary skeletal myotubes via co-immunoprecipitation assay and immunocytochemistry. STIM1 knockdown in mouse primary skeletal myotubes decreased Ca2+ uptake from the cytosol to the sarcoplasmic reticulum (SR) through SERCA1a only at micromolar cytosolic Ca2+ concentrations, suggesting that STIM1 could be required for the full activity of SERCA1a possibly during the relaxation of skeletal muscle. Various Ca2+ imaging experiments using myotubes expressing STIM1-SBR suggest that STIM1 is involved in intracellular Ca2+ distributions between the SR and the cytosol via regulating SERCA1a activity without affecting SOCE. Therefore, in skeletal muscle, STIM1 could play an important role in regulating Ca2+ movements between the SR and the cytosol.


Asunto(s)
Canales de Calcio/metabolismo , Fibras Musculares Esqueléticas/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Calcio/metabolismo , Canales de Calcio/química , Canales de Calcio/genética , Señalización del Calcio , Ratones , Datos de Secuencia Molecular , Unión Proteica , Conejos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Molécula de Interacción Estromal 1
10.
Biochem Biophys Res Commun ; 446(2): 454-9, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24613381

RESUMEN

The main tasks of skeletal muscle are muscle contraction and relaxation, which are mediated by changes in cytosolic Ca(2+) levels. Canonical-type transient receptor potential 3 (TRPC3) contains an ankyrin repeat (AR) region at the N-terminus (38-188 amino acids) and forms extracellular Ca(2+)-entry channels by homo or heteromerization with other TRP subtypes in various cells including skeletal myotubes. However, previous research has not determined which region(s) of TRPC3 is responsible for the heteromerization, whether the AR region participates in the heteromerizations, or what is the role of heteromeric TRPC3s in skeletal muscle. In the present study, the heteromerization of TRPC3 with TRPC1 was first examined by GST pull-down assays of TRPC3 portions with TRPC1. The portion containing the AR region of TRPC3 was bound to the TRPC1, but the binding was inhibited by the very end sub-region of the TRPC3 (1-37 amino acids). In-silico studies have suggested that the very end sub-region possibly induces a structural change in the AR region. Second, the very end sub-region of TRPC3 was expressed in mouse primary skeletal myotubes, resulting in a dominant-negative inhibition of heteromeric TRPC3/1 formation. In addition, the skeletal myotubes expressing the very end sub-region showed a decrease in resting cytosolic Ca(2+) levels. These results suggest that the AR region of TRPC3 could mediate the heteromeric TRPC3/1 formation, and the heteromeric TRPC3/1 could participate in regulating the resting cytosolic Ca(2+) levels in skeletal muscle.


Asunto(s)
Repetición de Anquirina/fisiología , Calcio/metabolismo , Citosol/metabolismo , Regulación de la Expresión Génica/fisiología , Fibras Musculares Esqueléticas/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Ratones , Unión Proteica , Multimerización de Proteína
11.
Biochem Biophys Res Commun ; 454(2): 282-8, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25450390

RESUMEN

Hemangioma is a benign tumor derived from abnormal blood vessel growth. Unlike other vascular tumor counterparts, a hemangioma is known to proliferate during its early stage but it is followed by a stage of involution where regression of the tumor occurs. The critical onset leading to the involution of hemangioma is currently not well understood. This study focused on the molecular identities of the involution of hemangioma. We demonstrated that a soluble factor released from the involuting phase of hemangioma-derived endothelial cells (HemECs) and identified pigment epithelium-derived factor (PEDF) as an anti-angiogenic factor that was associated with the growth inhibition of the involuting HemECs. The growth inhibition of the involuting HemECs was reversed by suppression of PEDF in the involuting HemECs. Furthermore, we found that PEDF was more up-regulated in the involuting phase of hemangioma tissues than in the proliferating or the involuted. Taken together, we propose that PEDF accelerates the involution of hemangioma by growth inhibition of HemECs in an autocrine manner. The regulatory mechanism of PEDF expression could be a potential therapeutic target to treat hemangiomas.


Asunto(s)
Vasos Sanguíneos/patología , Proliferación Celular , Células Endoteliales/patología , Proteínas del Ojo/metabolismo , Hemangioma/metabolismo , Hemangioma/patología , Factores de Crecimiento Nervioso/metabolismo , Serpinas/metabolismo , Apoptosis , Vasos Sanguíneos/metabolismo , Células Endoteliales/metabolismo , Proteínas del Ojo/genética , Hemangioma/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neovascularización Patológica , Factores de Crecimiento Nervioso/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Serpinas/genética , Células Tumorales Cultivadas
12.
Biochem J ; 453(2): 187-200, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23668188

RESUMEN

STIM1 (stromal interaction molecule 1) mediates SOCE (store-operated Ca²âº entry) in skeletal muscle. However, the direct role(s) of STIM1 in skeletal muscle, such as Ca²âº release from the SR (sarcoplasmic reticulum) for muscle contraction, have not been identified. The times required for the maximal expression of endogenous STIM1 or Orai1, or for the appearance of puncta during the differentiation of mouse primary skeletal myoblasts to myotubes, were all different, and the formation of puncta was detected with no stimulus during differentiation, suggesting that, in skeletal muscle, the formation of puncta is a part of the differentiation. Wild-type STIM1 and two STIM1 mutants (Triple mutant, missing Ca²âº-sensing residues but possessing the intact C-terminus; and E136X, missing the C-terminus) were overexpressed in the myotubes. The wild-type STIM1 increased SOCE, whereas neither mutant had an effect on SOCE. It was interesting that increases in the formation of puncta were observed in the Triple mutant as well as in wild-type STIM1, suggesting that SOCE-irrelevant puncta could exist in skeletal muscle. On the other hand, overexpression of wild-type or Triple mutant, but not E136X, attenuated Ca²âº releases from the SR in response to KCl [evoking ECC (excitation-contraction coupling) via activating DHPR (dihydropyridine receptor)] in a dominant-negative manner. The attenuation was removed by STIM1 knockdown, and STIM1 was co-immunoprecipitated with DHRP in a Ca²âº-independent manner. These results suggest that STIM1 negatively regulates Ca²âº release from the SR through the direct interaction of the STIM1 C-terminus with DHPR, and that STIM1 is involved in both ECC and SOCE in skeletal muscle.


Asunto(s)
Huesos/metabolismo , Calcio/metabolismo , Proteínas de la Membrana/fisiología , Fibras Musculares Esqueléticas/metabolismo , Proteínas de Neoplasias/fisiología , Retículo Sarcoplasmático/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , ADN Complementario , Humanos , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Molécula de Interacción Estromal 1
13.
Exp Mol Med ; 56(5): 1164-1177, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38689088

RESUMEN

Recent evidence of gut microbiota dysbiosis in the context of psoriasis and the increased cooccurrence of inflammatory bowel disease and psoriasis suggest a close relationship between skin and gut immune responses. Using a mouse model of psoriasis induced by the Toll-like receptor (TLR) 7 ligand imiquimod, we found that psoriatic dermatitis was accompanied by inflammatory changes in the small intestine associated with eosinophil degranulation, which impaired intestinal barrier integrity. Inflammatory responses in the skin and small intestine were increased in mice prone to eosinophil degranulation. Caco-2 human intestinal epithelial cells were treated with media containing eosinophil granule proteins and exhibited signs of inflammation and damage. Imiquimod-induced skin and intestinal changes were attenuated in eosinophil-deficient mice, and this attenuation was counteracted by the transfer of eosinophils. Imiquimod levels and the distribution of eosinophils were positively correlated in the intestine. TLR7-deficient mice did not exhibit intestinal eosinophil degranulation but did exhibit attenuated inflammation in the skin and small intestine following imiquimod administration. These results suggest that TLR7-dependent bidirectional skin-to-gut communication occurs in psoriatic inflammation and that inflammatory changes in the intestine can accelerate psoriasis.


Asunto(s)
Degranulación de la Célula , Modelos Animales de Enfermedad , Eosinófilos , Imiquimod , Intestino Delgado , Psoriasis , Receptor Toll-Like 7 , Animales , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 7/genética , Psoriasis/patología , Psoriasis/metabolismo , Ratones , Eosinófilos/metabolismo , Eosinófilos/inmunología , Humanos , Intestino Delgado/patología , Intestino Delgado/metabolismo , Piel/patología , Piel/metabolismo , Inflamación/patología , Inflamación/metabolismo , Ratones Noqueados , Células CACO-2 , Glicoproteínas de Membrana
14.
J Biol Chem ; 287(18): 14336-48, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22389502

RESUMEN

Junctophilins (JPs) play an important role in the formation of junctional membrane complexes (JMC) in striated muscle by physically linking the transverse-tubule and sarcoplasmic reticulum (SR) membranes. Researchers have found five JP2 mutants in humans with hypertrophic cardiomyopathy. Among these, Y141H and S165F are associated with severely altered Ca(2+) signaling in cardiomyocytes. We previously reported that S165F also induced both hypertrophy and altered intracellular Ca(2+) signaling in mouse skeletal myotubes. In the present study, we attempted to identify the dominant-negative role(s) of Y141H in primary mouse skeletal myotubes. Consistent with S165F, Y141H led to hypertrophy and altered Ca(2+) signaling (a decrease in the gain of excitation-contraction coupling and an increase in the resting level of myoplasmic Ca(2+)). However, unlike S165F, neither ryanodine receptor 1-mediated Ca(2+) release from the SR nor the phosphorylation of the mutated JP2 by protein kinase C was related to the altered Ca(2+) signaling by Y141H. Instead, abnormal JMC and increased SOCE via Orai1 were found, suggesting that the hypertrophy caused by Y141H progressed differently from S165F. Therefore JP2 can be linked to skeletal muscle hypertrophy via various Ca(2+) signaling pathways, and SOCE could be one of the causes of altered Ca(2+) signaling observed in muscle hypertrophy.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Mutación Missense , Sustitución de Aminoácidos , Animales , Canales de Calcio/genética , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/patología , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patología , Proteínas de la Membrana/genética , Ratones , Fibras Musculares Esqueléticas/patología , Proteínas Musculares/genética , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología , Proteína ORAI1 , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/patología
15.
J Cell Physiol ; 228(5): 1038-44, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23041942

RESUMEN

Angiopoietin 1 (Ang1) plays an important role in various endothelial functions, such as vascular integrity and angiogenesis; however, less is known about its function outside of the endothelium. In this study, we examined whether Ang1 has direct effects on skeletal muscle cells. We found that Ang1 exhibited myogenic potential, as it promoted the proliferation, migration, and differentiation of mouse primary skeletal myoblasts. The positive effect of Ang1 on myoblast proliferation could have been mediated by the α7 and ß1 integrins. We also found that Ang1 potentiated cellular Ca(2+) movements in differentiated myotubes in response to stimuli, possibly through the increased expression of two Ca(2+) -related proteins, namely, Orai1 and calmodulin. Ang1 also increased Orai1 and calmodulin expression in mouse hearts in vivo. These results provide an insight into the molecular mechanisms by which Ang1 directly affects the myogenesis of striated muscle.


Asunto(s)
Angiopoyetina 1/metabolismo , Diferenciación Celular , Proliferación Celular , Desarrollo de Músculos , Mioblastos Esqueléticos , Animales , Antígenos CD/metabolismo , Calcio/metabolismo , Canales de Calcio/metabolismo , Calmodulina/metabolismo , Movimiento Celular , Regulación de la Expresión Génica , Cadenas alfa de Integrinas/metabolismo , Integrina beta1/metabolismo , Ratones , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Neovascularización Fisiológica , Proteína ORAI1
16.
Biochem Biophys Res Commun ; 436(4): 691-7, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23770419

RESUMEN

Despite the altered expression of Tie receptors and angiopoietin ligands during hypoxic conditions, the effect of hypoxia on Tie-mediated endothelial responses has not been elucidated. In this study, we found that hypoxia increased Tie receptor expression but attenuated angiopoietin-1 (Ang1)-induced Tie2 activity, including Tie2 phosphorylation, Tie2 downstream signaling activation, and endothelial cell tube formation. However, Ang1 binding to endothelial cells was increased during hypoxic conditions. We demonstrated that Tie1 suppression restored the Tie2 activity and that Tie1-mediated Tie2 suppression was independent of tyrosine phosphatase activity. These results suggest that under hypoxic conditions, Tie1 is critical for reducing Ang1-induced Tie2 activity and angiogenesis.


Asunto(s)
Angiopoyetina 1/metabolismo , Hipoxia de la Célula , Endotelio Vascular/metabolismo , Receptor TIE-1/metabolismo , Células Cultivadas , Endotelio Vascular/enzimología , Humanos , Transducción de Señal
17.
Cells ; 12(16)2023 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-37626915

RESUMEN

Tripartite motif-containing protein 32 (TRIM32) is a member of the tripartite motif family and is highly conserved from flies to humans. Via its E3 ubiquitin ligase activity, TRIM32 mediates and regulates many physiological and pathophysiological processes, such as growth, differentiation, muscle regeneration, immunity, and carcinogenesis. TRIM32 plays multifunctional roles in the maintenance of skeletal muscle. Genetic variations in the TRIM32 gene are associated with skeletal muscular dystrophies in humans, including limb-girdle muscular dystrophy type 2H (LGMD2H). LGMD2H-causing genetic variations of TRIM32 occur most frequently in the C-terminal NHL (ncl-1, HT2A, and lin-41) repeats of TRIM32. LGMD2H is characterized by skeletal muscle dystrophy, myopathy, and atrophy. Surprisingly, most patients with LGMD2H show minimal or no dysfunction in other tissues or organs, despite the broad expression of TRIM32 in various tissues. This suggests more prominent roles for TRIM32 in skeletal muscle than in other tissues or organs. This review is focused on understanding the physiological roles of TRIM32 in skeletal muscle, the pathophysiological mechanisms mediated by TRIM32 genetic variants in LGMD2H patients, and the correlations between TRIM32 and Duchenne muscular dystrophy (DMD).


Asunto(s)
Distrofia Muscular de Cinturas , Distrofia Muscular de Duchenne , Humanos , Músculo Esquelético , Distrofia Muscular de Cinturas/genética , Atrofia , Proteínas de Motivos Tripartitos/genética , Factores de Transcripción , Ubiquitina-Proteína Ligasas/genética
18.
Biochem Biophys Res Commun ; 428(3): 383-8, 2012 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-23103543

RESUMEN

Mitsugumin 53 (MG53) is a member of the membrane repair system in skeletal muscle. However, the roles of MG53 in the unique functions of skeletal muscle have not been addressed, although it is known that MG53 is expressed only in skeletal and cardiac muscle. In the present study, MG53-binding proteins were examined along with proteins that mediate skeletal muscle contraction and relaxation using the binding assays of various MG53 domains and quadrupole time-of-flight mass spectrometry. MG53 binds to sarcoplasmic reticulum Ca(2+)-ATPase 1a (SERCA1a) via its tripartite motif (TRIM) and PRY domains. The binding was confirmed in rabbit skeletal muscle and mouse primary skeletal myotubes by co-immunoprecipitation and immunocytochemistry. MG53 knockdown in mouse primary skeletal myotubes increased Ca(2+)-uptake through SERCA1a (more than 35%) at micromolar Ca(2+) but not at nanomolar Ca(2+), suggesting that MG53 attenuates SERCA1a activity possibly during skeletal muscle contraction or relaxation but not during the resting state of skeletal muscle. Therefore MG53 could be a new candidate for the diagnosis and treatment of patients with Brody syndrome, which is not related to the mutations in the gene coding for SERCA1a, but still accompanies exercise-induced muscle stiffness and delayed muscle relaxation due to a reduction in SERCA1a activity.


Asunto(s)
Proteínas Portadoras/metabolismo , Músculo Esquelético/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Proteínas Portadoras/genética , Técnicas de Silenciamiento del Gen , Inmunoprecipitación , Proteínas de la Membrana , Ratones , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Estructura Terciaria de Proteína , Proteolípidos/metabolismo , Conejos
19.
Immune Netw ; 22(2): e20, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35573151

RESUMEN

Despite the high prevalence of chronic dermatitis and the accompanied intractable itch, therapeutics that specifically target itching have low efficacy. Increasing evidence suggests that TLRs contribute to immune activation and neural sensitization; however, their roles in chronic itch remain elusive. Here, we show that the RBL-2H3 mast cell line expresses TLR4 and that treatment with a TLR4 antagonist opposes the LPS dependent increase in mRNA levels of Th2 and innate cytokines. The pathological role of TLR4 activation in itching was studied in neonate rats that developed chronic itch due to neuronal damage after receiving subcutaneous capsaicin injections. Treatment with a TLR4 antagonist protected these rats with chronic itch against scratching behavior and chronic dermatitis. TLR4 antagonist treatment also restored the density of cutaneous nerve fibers and inhibited the histopathological changes that are associated with mast cell activation after capsaicin injection. Additionally, the expression of IL-1ß, IL-4, IL-5, IL-10, and IL-13 mRNA in the lesional skin decreased after TLR4 antagonist treatment. Based on these data, we propose that inhibiting TLR4 alleviated itch in a rat model of chronic relapsing itch, and the reduction in the itch was associated with TLR4 signaling in mast cells and nerve fibers.

20.
Biochem J ; 427(1): 125-34, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20095964

RESUMEN

JPs (junctophilins) contribute to the formation of junctional membrane complexes in muscle cells by physically linking the t-tubule (transverse-tubule) and SR (sarcoplasmic reticulum) membranes. In humans with HCM (hypertrophic cardiomyopathy), mutations in JP2 are linked to altered Ca2+ signalling in cardiomyocytes; however, the effects of these mutations on skeletal muscle function have not been examined. In the present study, we investigated the role of the dominant-negative JP2-S165F mutation (which is associated with human HCM) in skeletal muscle. Consistent with the hypertrophy observed in human cardiac muscle, overexpression of JP2-S165F in primary mouse skeletal myotubes led to a significant increase in myotube diameter and resting cytosolic Ca2+ concentration. Single myotube Ca2+ imaging experiments showed reductions in both the excitation-contraction coupling gain and RyR (ryanodine receptor) 1-mediated Ca2+ release from the SR. Immunoprecipitation assays revealed defects in the PKC (protein kinase C)-mediated phosphorylation of the JP2-S165F mutant protein at Ser165 and in binding of JP2-S165F to the Ca2+ channel TRPC3 (transient receptor potential cation canonical-type channel 3) on the t-tubule membrane. Therefore both the hypertrophy and altered intracellular Ca2+ signalling in the JP2-S165F-expressing skeletal myotubes can be linked to altered phosphorylation of JP2 and/or altered cross-talk among Ca2+ channels on the t-tubule and SR membranes.


Asunto(s)
Señalización del Calcio/fisiología , Proteínas de la Membrana/genética , Músculo Esquelético/metabolismo , Mutación/genética , Transducción de Señal , Animales , Cafeína/farmacología , Células Cultivadas , Citosol/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Fibras Musculares Esqueléticas/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación , Proteína Quinasa C/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Canales Catiónicos TRPC/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA