Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
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
2.
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
3.
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
4.
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.

5.
Cells ; 10(11)2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34831044

RESUMEN

Calsequestrin 1 (CASQ1) in skeletal muscle buffers and senses Ca2+ in the sarcoplasmic reticulum (SR). CASQ1 also regulates store-operated Ca2+ entry (SOCE) by binding to stromal interaction molecule 1 (STIM1). Abnormal SOCE and/or abnormal expression or mutations in CASQ1, STIM1, or STIM2 are associated with human skeletal, cardiac, or smooth muscle diseases. However, the functional relevance of CASQ1 along with STIM2 has not been studied in any tissue, including skeletal muscle. First, in the present study, it was found by biochemical approaches that CASQ1 is bound to STIM2 via its 92 N-terminal amino acids (C1 region). Next, to examine the functional relevance of the CASQ1-STIM2 interaction in skeletal muscle, the full-length wild-type CASQ1 or the C1 region was expressed in mouse primary skeletal myotubes, and the myotubes were examined using single-myotube Ca2+ imaging experiments and transmission electron microscopy observations. The CASQ1-STIM2 interaction via the C1 region decreased SOCE, increased intracellular Ca2+ release for skeletal muscle contraction, and changed intracellular Ca2+ distributions (high Ca2+ in the SR and low Ca2+ in the cytosol were observed). Furthermore, the C1 region itself (which lacks Ca2+-buffering ability but has STIM2-binding ability) decreased the expression of Ca2+-related proteins (canonical-type transient receptor potential cation channel type 6 and calmodulin 1) and induced mitochondrial shape abnormalities. Therefore, in skeletal muscle, CASQ1 plays active roles in Ca2+ movement and distribution by interacting with STIM2 as well as Ca2+ sensing and buffering.


Asunto(s)
Calsecuestrina/metabolismo , Músculo Esquelético/metabolismo , Molécula de Interacción Estromal 2/metabolismo , Animales , Calcio/metabolismo , Calsecuestrina/química , Citosol/metabolismo , Dinaminas/metabolismo , GTP Fosfohidrolasas/metabolismo , Humanos , Espacio Intracelular/metabolismo , Ratones , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Modelos Moleculares , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/ultraestructura , Unión Proteica , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
6.
Biomolecules ; 11(8)2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34439731

RESUMEN

Stromal interaction molecule 1 (STIM1) is the main protein that, along with Orai1, mediates store-operated Ca2+ entry (SOCE) in skeletal muscle. Abnormal SOCE due to mutations in STIM1 is one of the causes of human skeletal muscle diseases. STIM1-R304Q (a constitutively active form of STIM1) has been found in human patients with skeletal muscle phenotypes such as muscle weakness, myalgia, muscle stiffness, and contracture. However, the pathological mechanism(s) of STIM1-R304Q in skeletal muscle have not been well studied. To examine the pathological mechanism(s) of STIM1-R304Q in skeletal muscle, STIM1-R304Q was expressed in mouse primary skeletal myotubes, and the properties of the skeletal myotubes were examined using single-myotube Ca2+ imaging, transmission electron microscopy (TEM), and biochemical approaches. STIM1-R304Q did not interfere with the terminal differentiation of skeletal myoblasts to myotubes and retained the ability of STIM1 to attenuate dihydropyridine receptor (DHPR) activity. STIM1-R304Q induced hyper-SOCE (that exceeded the SOCE by wild-type STIM1) by affecting both the amplitude and the onset rate of SOCE. Unlike that by wild-type STIM1, hyper-SOCE by STIM1-R304Q contributed to a disturbance in Ca2+ distribution between the cytosol and the sarcoplasmic reticulum (SR) (high Ca2+ in the cytosol and low Ca2+ in the SR). Moreover, the hyper-SOCE and the high cytosolic Ca2+ level induced by STIM1-R304Q involve changes in mitochondrial shape. Therefore, a series of these cellular defects induced by STIM1-R304Q could induce deleterious skeletal muscle phenotypes in human patients carrying STIM1-R304Q.


Asunto(s)
Fibras Musculares Esqueléticas/metabolismo , Mioblastos Esqueléticos/metabolismo , Proteínas de Neoplasias/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Humanos , Ratones , Fibras Musculares Esqueléticas/citología , Mioblastos Esqueléticos/citología
7.
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
8.
Cell Death Dis ; 12(3): 243, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33664229

RESUMEN

Atopic dermatitis is a chronic skin inflammatory disease mediated by Th2-type immune responses. Although intestinal immune responses have been shown to play a critical role in the development or prevention of atopic dermatitis, the precise influence of intestinal immunity on atopic dermatitis is incompletely understood. We show here that orally tolerized mice are protected from experimental atopic dermatitis induced by sensitization and epicutaneous (EC) challenge to ovalbumin. Although the expression of Th2-type cytokines in the small intestine of orally tolerized and EC-challenged mice did not change significantly, these mice showed decreased inflammatory responses in the small intestine with restoration of microbial change elicited by the EC challenge. Interestingly, an increase in small intestinal eosinophils was observed with the EC challenge, which was also inhibited by oral tolerance. The role of small intestinal eosinophils and microbiota in the pathogenesis of experimental atopic dermatitis was further substantiated by decreased inflammatory mediators in the small intestine and attenuated Th2-type inflammation in the skin of eosinophil-deficient and microbiota-ablated mice with EC challenges. Based on these data, we propose that the bidirectional interaction between the skin and the intestine has a role in the pathogenesis of atopic dermatitis and that modulation of the intestinal microenvironments could be a therapeutic approach to atopic dermatitis.


Asunto(s)
Dermatitis Atópica/prevención & control , Desensibilización Inmunológica , Tolerancia Inmunológica , Intestino Delgado/inmunología , Leucocitos/inmunología , Ovalbúmina/administración & dosificación , Piel/inmunología , Administración Oral , Animales , Bacterias/inmunología , Claudina-4/genética , Claudina-4/metabolismo , Citocinas/genética , Citocinas/metabolismo , Dermatitis Atópica/inmunología , Dermatitis Atópica/metabolismo , Dermatitis Atópica/microbiología , Modelos Animales de Enfermedad , Disbiosis , Femenino , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Leucocitos/metabolismo , Ratones Endogámicos BALB C , Piel/metabolismo
9.
Exp Mol Med ; 52(12): 1908-1925, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33288873

RESUMEN

Calsequestrin (CASQ) was discovered in rabbit skeletal muscle tissues in 1971 and has been considered simply a passive Ca2+-buffering protein in the sarcoplasmic reticulum (SR) that provides Ca2+ ions for various Ca2+ signals. For the past three decades, physiologists, biochemists, and structural biologists have examined the roles of the skeletal muscle type of CASQ (CASQ1) in skeletal muscle and revealed that CASQ1 has various important functions as (1) a major Ca2+-buffering protein to maintain the SR with a suitable amount of Ca2+ at each moment, (2) a dynamic Ca2+ sensor in the SR that regulates Ca2+ release from the SR to the cytosol, (3) a structural regulator for the proper formation of terminal cisternae, (4) a reverse-directional regulator of extracellular Ca2+ entries, and (5) a cause of human skeletal muscle diseases. This review is focused on understanding these functions of CASQ1 in the physiological or pathophysiological status of skeletal muscle.


Asunto(s)
Calsecuestrina/metabolismo , Músculo Esquelético/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio/metabolismo , Calsecuestrina/química , Calsecuestrina/genética , Susceptibilidad a Enfermedades , Acoplamiento Excitación-Contracción , Regulación de la Expresión Génica , Humanos , Fosforilación , Isoformas de Proteínas , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo , Transducción de Señal , Relación Estructura-Actividad
10.
Cells ; 9(4)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32244622

RESUMEN

Ca2+ itself or Ca2+-dependent signaling pathways play fundamental roles in various cellular processes from cell growth to death. The most representative example can be found in skeletal muscle cells where a well-timed and adequate supply of Ca2+ is required for coordinated Ca2+-dependent skeletal muscle functions, such as the interactions of contractile proteins during contraction. Intracellular Ca2+ movements between the cytosol and sarcoplasmic reticulum (SR) are strictly regulated to maintain the appropriate Ca2+ supply in skeletal muscle cells. Added to intracellular Ca2+ movements, the contribution of extracellular Ca2+ entry to skeletal muscle functions and its significance have been continuously studied since the early 1990s. Here, studies on the roles of channel proteins that mediate extracellular Ca2+ entry into skeletal muscle cells using skeletal myoblasts, myotubes, fibers, tissue, or skeletal muscle-originated cell lines are reviewed with special attention to the proposed functions of transient receptor potential canonical proteins (TRPCs) as store-operated Ca2+ entry (SOCE) channels under normal conditions and the potential abnormal properties of TRPCs in muscle diseases such as Duchenne muscular dystrophy (DMD).


Asunto(s)
Músculo Esquelético/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Humanos , Modelos Biológicos , Distrofias Musculares/metabolismo
11.
Sci Rep ; 9(1): 19140, 2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31844136

RESUMEN

Stromal interaction molecule 1 (STIM1) mediates extracellular Ca2+ entry into the cytosol through a store-operated Ca2+ entry (SOCE) mechanism, which is involved in the physiological functions of various tissues, including skeletal muscle. STIM1 is also associated with skeletal muscle diseases, but its pathological mechanisms have not been well addressed. The present study focused on examining the pathological mechanism(s) of a mutant STIM1 (R429C) that causes human muscular hypotonia. R429C was expressed in mouse primary skeletal myotubes, and the properties of the skeletal myotubes were examined using single-cell Ca2+ imaging of myotubes and transmission electron microscopy (TEM) along with biochemical approaches. R429C did not interfere with the terminal differentiation of myoblasts to myotubes. Unlike wild-type STIM1, there was no further increase of SOCE by R429C. R429C bound to endogenous STIM1 and slowed down the initial rate of SOCE that were mediated by endogenous STIM1. Moreover, R429C increased intracellular Ca2+ movement in response to membrane depolarization by eliminating the attenuation on dihydropyridine receptor-ryanodine receptor (DHPR-RyR1) coupling by endogenous STIM1. The cytosolic Ca2+ level was also increased due to the reduction in SR Ca2+ level. In addition, R429C-expressing myotubes showed abnormalities in mitochondrial shape, a significant decrease in ATP levels, and the higher expression levels of mitochondrial fission-mediating proteins. Therefore, serial defects in SOCE, intracellular Ca2+ movement, and cytosolic Ca2+ level along with mitochondrial abnormalities in shape and ATP level could be a pathological mechanism of R429C for human skeletal muscular hypotonia. This study also suggests a novel clue that STIM1 in skeletal muscle could be related to mitochondria via regulating intra and extracellular Ca2+ movements.


Asunto(s)
Calcio/metabolismo , Espacio Extracelular/metabolismo , Espacio Intracelular/metabolismo , Hipotonía Muscular/genética , Músculo Esquelético/patología , Mutación/genética , Proteínas de Neoplasias/genética , Molécula de Interacción Estromal 1/genética , Canales de Calcio Tipo L/metabolismo , Citosol/metabolismo , Humanos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Modelos Biológicos , Fibras Musculares Esqueléticas/metabolismo , Hipotonía Muscular/patología , Proteínas de Neoplasias/química , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Molécula de Interacción Estromal 1/química
13.
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
14.
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
15.
Sci Rep ; 8(1): 9894, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29967467

RESUMEN

Accumulating data have indicated a fundamental role of eosinophils in regulating adipose tissue homeostasis. Here, we performed whole-genome RNA sequencing of the small intestinal tract, which suggested the presence of impaired lipid metabolism in eosinophil-deficient ΔdblGATA mice. ΔdblGATA mice fed a high-fat diet (HFD) showed reduced body fat mass, impaired enlargement of adipocytes, decreased expression of adipogenic genes, and developed glucose intolerance. HFD induced accumulation of eosinophils in the perigonadal white adipose tissue. Concordantly, adipocyte-differentiated 3T3-L1 cells promoted the migration of eosinophils through the expression of CCL11 (eotaxin-1) and likely promoted their survival through the expression of interleukin (IL)-3, IL-5, and granulocyte-macrophage colony-stimulating factor. HFD-fed ΔdblGATA mice showed increased infiltration of macrophages, CD4+ T-cells, and B-cells, increased expression of interferon-γ, and decreased expression of IL-4 and IL-13 in white adipose tissue. Interferon-γ treatment significantly decreased lipid deposition in adipocyte-differentiated 3T3-L1 cells, while IL-4 treatment promoted lipid accumulation. Notably, HFD-fed ΔdblGATA mice showed increased lipid storage in the liver as compared with wild-type mice. We propose that obesity promotes the infiltration of eosinophils into adipose tissue that subsequently contribute to the metabolic homeostasis by promoting adipocyte maturation.


Asunto(s)
Adipocitos/patología , Eosinófilos/patología , Obesidad/patología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo Blanco/patología , Animales , Diferenciación Celular , Quimiocina CCL11/genética , Quimiocina CCL11/metabolismo , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Eosinófilos/metabolismo , Factores de Transcripción GATA/genética , Prueba de Tolerancia a la Glucosa , Interferón gamma/farmacología , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Obesidad/etiología , Obesidad/metabolismo
16.
Mol Cells ; 41(8): 771-780, 2018 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-30037214

RESUMEN

Angiogenesis must be precisely controlled because uncontrolled angiogenesis is involved in aggravation of disease symptoms. Vascular endothelial growth factor (VEGF)/VEGF receptor 2 (VEGFR-2) signaling is a key pathway leading to angiogenic responses in vascular endothelial cells (ECs). Therefore, targeting VEGF/VEGFR-2 signaling may be effective at modulating angiogenesis to alleviate various disease symptoms. Oleanolic acid was verified as a VEGFR-2 binding chemical from anticancer herbs with similar binding affinity as a reference drug in the Protein Data Bank (PDB) entry 3CJG of model A coordination. Oleanolic acid effectively inhibited VEGF-induced VEGFR-2 activation and angiogenesis in HU-VECs without cytotoxicity. We also verified that oleanolic acid inhibits in vivo angiogenesis during the development and the course of the retinopathy of prematurity (ROP) model in the mouse retina. Taken together, our results suggest a potential therapeutic benefit of oleanolic acid for inhibiting angiogenesis in proangiogenic diseases, including retinopathy.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Ácido Oleanólico/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Recombinantes/farmacología , Neovascularización Retiniana/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
17.
BMB Rep ; 51(8): 378-387, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29898810

RESUMEN

Skeletal muscle contracts or relaxes to maintain the body position and locomotion. For the contraction and relaxation of skeletal muscle, Ca2+ in the cytosol of skeletal muscle fibers acts as a switch to turn on and off a series of contractile proteins. The cytosolic Ca2+ level in skeletal muscle fibers is governed mainly by movements of Ca2+ between the cytosol and the sarcoplasmic reticulum (SR). Store-operated Ca2+ entry (SOCE), a Ca2+ entryway from the extracellular space to the cytosol, has gained a significant amount of attention from muscle physiologists. Orai1 and stromal interaction molecule 1 (STIM1) are the main protein identities of SOCE. This mini-review focuses on the roles of STIM proteins and SOCE in the physiological and pathophysiological functions of skeletal muscle and in their correlations with recently identified proteins, as well as historical proteins that are known to mediate skeletal muscle function. [BMB Reports 2018; 51(8): 378-387].


Asunto(s)
Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Moléculas de Interacción Estromal/metabolismo , Moléculas de Interacción Estromal/fisiología , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Espacio Extracelular/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Fibras Musculares Esqueléticas/fisiología , Retículo Sarcoplasmático/metabolismo
18.
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
19.
Sci Rep ; 7(1): 17936, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-29263348

RESUMEN

Stromal interaction molecule 1 (STIM1) along with Orai1 mediates extracellular Ca2+ entry into the cytosol through a store-operated Ca2+ entry (SOCE) mechanism in various tissues including skeletal muscle. However, the role(s) of STIM2, a homolog of STIM1, in skeletal muscle has not been well addressed. The present study, first, was focused on searching for STIM2-binding proteins from among proteins mediating skeletal muscle functions. This study used a binding assay, quadrupole time-of-flight mass spectrometry, and co-immunoprecipitation assay with bona-fide STIM2- and SERCA1a-expressing rabbit skeletal muscle. The region for amino acids from 453 to 729 of STIM2 binds to sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 1a (SERCA1a). Next, oxalate-supported 45Ca2+-uptake experiments and various single-myotube Ca2+ imaging experiments using STIM2-knockdown mouse primary skeletal myotubes have suggested that STIM2 attenuates SERCA1a activity during skeletal muscle contraction, which contributes to the intracellular Ca2+ distribution between the cytosol and the SR at rest. In addition, STIM2 regulates Ca2+ movement through RyR1 during skeletal muscle contraction as well as SOCE. Therefore, via regulation of SERCA1a activity, STIM2 regulates both intracellular Ca2+ distribution and Ca2+ movement in skeletal muscle, which makes it both similar to, yet different from, STIM1.


Asunto(s)
Calcio/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Molécula de Interacción Estromal 2/fisiología , Animales , Retículo Endoplásmico/metabolismo , Inmunoprecipitación , Espectrometría de Masas , Ratones , Ratones Noqueados , Contracción Muscular , Fibras Musculares Esqueléticas/fisiología , Conejos , Retículo Sarcoplasmático/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Molécula de Interacción Estromal 2/metabolismo
20.
Exp Mol Med ; 49(9): e378, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28912570

RESUMEN

The main task of skeletal muscle is contraction and relaxation for body movement and posture maintenance. During contraction and relaxation, Ca2+ in the cytosol has a critical role in activating and deactivating a series of contractile proteins. In skeletal muscle, the cytosolic Ca2+ level is mainly determined by Ca2+ movements between the cytosol and the sarcoplasmic reticulum. The importance of Ca2+ entry from extracellular spaces to the cytosol has gained significant attention over the past decade. Store-operated Ca2+ entry with a low amplitude and relatively slow kinetics is a main extracellular Ca2+ entryway into skeletal muscle. Herein, recent studies on extracellular Ca2+ entry into skeletal muscle are reviewed along with descriptions of the proteins that are related to extracellular Ca2+ entry and their influences on skeletal muscle function and disease.


Asunto(s)
Calcio/metabolismo , Músculo Esquelético/metabolismo , Animales , Transporte Biológico , Canales de Calcio/metabolismo , Senescencia Celular , Susceptibilidad a Enfermedades , Espacio Extracelular/metabolismo , Humanos , Espacio Intracelular/metabolismo , Fatiga Muscular , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...