Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Biol Chem ; 299(1): 102716, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36403856

RESUMEN

Adipocyte hyperplasia and hypertrophy are the two main processes contributing to adipose tissue expansion, yet the mechanisms that regulate and balance their involvement in obesity are incompletely understood. Activin B/GDF-3 receptor ALK7 is expressed in mature adipocytes and promotes adipocyte hypertrophy upon nutrient overload by suppressing adrenergic signaling and lipolysis. In contrast, the role of ALK4, the canonical pan-activin receptor, in adipose tissue is unknown. Here, we report that, unlike ALK7, ALK4 is preferentially expressed in adipocyte precursors, where it suppresses differentiation, allowing proliferation and adipose tissue expansion. ALK4 expression in adipose tissue increases upon nutrient overload and positively correlates with fat depot mass and body weight, suggesting a role in adipose tissue hyperplasia during obesity. Mechanistically, ALK4 signaling suppresses expression of CEBPα and PPARγ, two master regulators of adipocyte differentiation. Conversely, ALK4 deletion enhances CEBPα/PPARγ expression and induces premature adipocyte differentiation, which can be rescued by CEBPα knockdown. These results clarify the function of ALK4 in adipose tissue and highlight the contrasting roles of the two activin receptors in the regulation of adipocyte hyperplasia and hypertrophy during obesity.


Asunto(s)
Receptores de Activinas Tipo I , Adipocitos , Tejido Adiposo , Humanos , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Hiperplasia/metabolismo , Hipertrofia/metabolismo , Obesidad/metabolismo , PPAR gamma/metabolismo , Diferenciación Celular , Receptores de Activinas Tipo I/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo
2.
FASEB J ; 35(8): e21759, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34245608

RESUMEN

Life-style change and anti-inflammatory interventions have only transient effects in obesity. It is not clear how benefits obtained by these treatments can be maintained longer term, especially during sustained high caloric intake. Constitutive ablation of the activin receptor ALK7 in adipose tissue enhances catecholamine signaling and lipolysis in adipocytes, and protects mice from diet-induced obesity. Here, we investigated the consequences of conditional ALK7 ablation in adipocytes of adult mice with pre-existing obesity. Although ALK7 deletion had little effect on its own, it synergized strongly with a transient switch to low-fat diet (life-style change) or anti-inflammatory treatment (Na-salicylate), resulting in enhanced lipolysis, increased energy expenditure, and reduced adipose tissue mass and body weight gain, even under sustained high caloric intake. By themselves, diet-switch and salicylate had only a temporary effect on weight gain. Mechanistically, combination of ALK7 ablation with either treatment strongly enhanced the levels of ß3-AR, the main adrenergic receptor for catecholamine stimulation of lipolysis, and C/EBPα, an upstream regulator of ß3-AR expression. These results suggest that inhibition of ALK7 can be combined with simple interventions to produce longer-lasting benefits in obesity.


Asunto(s)
Receptores de Activinas Tipo I/deficiencia , Adipocitos/metabolismo , Ingestión de Alimentos , Lipólisis , Obesidad/metabolismo , Receptores de Activinas Tipo I/metabolismo , Adipocitos/patología , Animales , Ratones , Ratones Transgénicos , Obesidad/genética , Obesidad/patología , Salicilatos/farmacología
3.
J Biol Chem ; 295(7): 2034-2042, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31919095

RESUMEN

Prolonged cold exposure stimulates the formation of brownlike adipocytes expressing UCP1 (uncoupling-protein-1) in subcutaneous white adipose tissue which, together with classical brown adipose tissue, contributes to maintaining body temperature in mammals through nonshivering thermogenesis. The mechanisms that regulate the formation of these cells, alternatively called beige or brite adipocytes, are incompletely understood. Here we report that mice lacking CD137, a cell surface protein used in several studies as a marker for beige adipocytes, showed elevated levels of thermogenic markers, including UCP1, increased numbers of beige adipocyte precursors, and expanded UCP1-expressing cell clusters in inguinal white adipose tissue after chronic cold exposure. CD137 knockout mice also showed enhanced cold resistance. These results indicate that CD137 functions as a negative regulator of "browning" in white adipose tissue and call into question the use of this protein as a functional marker for beige adipocytes.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Proteína Desacopladora 1/genética , Adipocitos Beige/metabolismo , Animales , Temperatura Corporal/genética , Frío , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Noqueados , Termogénesis/genética
5.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38740432

RESUMEN

Subclinical vascular impairment can be exacerbated in individuals who experience sustained inflammation after COVID-19 infection. Our study explores the prevalence and impact of autoantibodies on vascular dysfunction in healthy COVID-19 survivors, an area that remains inadequately investigated. Focusing on autoantibodies against the atypical chemokine receptor 1 (ACKR1), COVID-19 survivors demonstrated significantly elevated anti-ACKR1 autoantibodies, correlating with systemic cytokines, circulating damaged endothelial cells, and endothelial dysfunction. An independent cohort linked these autoantibodies to increased vascular disease outcomes during a median 6.7-yr follow-up. We analyzed a single-cell transcriptome atlas of endothelial cells from diverse mouse tissues, identifying enriched Ackr1 expressions in venous regions of the brain and soleus muscle vasculatures, which holds intriguing implications for tissue-specific venous thromboembolism manifestations reported in COVID-19. Functionally, purified immunoglobulin G (IgG) extracted from patient plasma did not trigger cell apoptosis or increase barrier permeability in human vein endothelial cells. Instead, plasma IgG enhanced antibody-dependent cellular cytotoxicity mediated by patient PBMCs, a phenomenon alleviated by blocking peptide or liposome ACKR1 recombinant protein. The blocking peptide uncovered that purified IgG from COVID-19 survivors possessed potential epitopes in the N-terminal extracellular domain of ACKR1, which effectively averted antibody-dependent cellular cytotoxicity. Our findings offer insights into therapeutic development to mitigate autoantibody reactivity in blood vessels in chronic inflammation.


Asunto(s)
Autoanticuerpos , COVID-19 , SARS-CoV-2 , Humanos , Autoanticuerpos/inmunología , COVID-19/inmunología , Animales , Ratones , Femenino , Masculino , SARS-CoV-2/inmunología , Inflamación/inmunología , Persona de Mediana Edad , Endotelio Vascular/metabolismo , Endotelio Vascular/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Células Endoteliales/metabolismo , Células Endoteliales/inmunología , Adulto , Anciano
6.
Biomaterials ; 302: 122325, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37751670

RESUMEN

A major bottleneck in drug/gene delivery to enhance tissue regeneration after injuries is to achieve targeted delivery to the cells of interest. Unfortunately, we have not been able to attain effective targeted drug delivery in tissues due to the lack of efficient delivery platforms. Since specific cell-cell interactions exist to impart the unique structure and functionality of tissues and organs, we hypothesize that such specific cellular interactions may also be harnessed for drug delivery applications in the form of cell membrane coatings. Here, we employed neural cell-derived membrane coating technique on DNA nanogels to improve target specificity. The efficacy of neural cell membrane-coated DNA nanogels (NCM-nanogels) was demonstrated by using four types of cell membranes derived from the central nervous system (CNS), namely, astrocytes, microglia, cortical neurons, and oligodendrocyte progenitor cells (OPCs). A successful coating of NCMs over DNA nanogels was confirmed by dynamic light scattering, zeta potential measurements and transmission electron microscopy. Subsequently, an overall improvement in cellular uptake of NCM-nanogels over uncoated DNA nanogels (p < 0.005) was seen. Additionally, we observed a selective uptake of OPC membrane-coated DNA nanogels (NCM-O mem) by oligodendrocytes over other cell types both in vitro and in vivo. Our quantitative polymerase chain reaction (qPCR) results also showed selective and effective gene knockdown capacity of NCM-O mem for OPC transfection. The findings in this work may be beneficial for future drug delivery applications targeted at the CNS.


Asunto(s)
Sistema Nervioso Central , Sistemas de Liberación de Medicamentos , Nanogeles , Sistemas de Liberación de Medicamentos/métodos , Neuronas , Membrana Celular , ADN , Portadores de Fármacos/química
7.
Pathobiology ; 77(2): 96-105, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20332669

RESUMEN

OBJECTIVES: This study aimed to examine (1) the expression of P16 protein relative to sites of presentation, immunophenotypic subgroups and proliferative indices of tumour cells, and (2) the relationship between p16 gene alterations and P16 protein overexpression in 70 cases of diffuse large B cell lymphoma (DLBCL). METHODS: Expression of P16, CD10, BCL-6, MUM-1 and proliferation marker (Ki-67) was demonstrated by immunohistochemistry. Fluorescence in situ hybridization (FISH) was employed to detect p16 alterations. RESULTS: P16 overexpression was shown in 45.7% (32/70) of the DLBCL cases, and was significantly correlated with CD10 (p = 0.022) and germinal centre B-cell-like (GCB) phenotype (p = 0.022). High expression of P16 was inversely associated with high proliferative activity (Ki-67 index greater than 75%) (p = 0.020). Of the 47 cases that yielded interpretable FISH results, 57.4% (27/47) showed deletions of p16 and 27.7% (13/47) showed gains of p16. P16 overexpression and p16 deletions were mutually exclusive (p = 0.019). There was no correlation between P16 overexpression and p16 gains (p = 0.621). CONCLUSIONS: The GCB and non-GCB subgroups of DLBCLs show different patterns of P16 expression. High levels of P16 may mitigate tumour cell proliferation. Gains of p16 do not necessarily increase P16 protein expression.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes p16 , Linfoma de Células B Grandes Difuso/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Proliferación Celular , Niño , Preescolar , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Femenino , Humanos , Técnicas para Inmunoenzimas , Hibridación Fluorescente in Situ , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Masculino , Persona de Mediana Edad
8.
Malays J Pathol ; 31(1): 53-6, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19694314

RESUMEN

The retinoblastoma-related gene Rb2/p130 has been reported to be mutated in several malignancies such as lung cancer and Burkitt's lymphoma. Nasopharyngeal carcinoma (NPC) is a common cancer in Malaysia especially amongst the ethnic Chinese. We screened for Rb2/p130 gene (exons 19 to 21) mutations in 53 archival NPC samples via PCR-SSCP-direct sequencing approach. Only one sample had a base change which involved a serine to glycine substitution at codon 995 (S995G). We conclude that Rb2/p130 genetic alterations are infrequent in NPC and may not be essential for the pathogenesis of the disease.


Asunto(s)
Carcinoma/genética , Neoplasias Nasofaríngeas/genética , Proteína p130 Similar a la del Retinoblastoma/genética , Carcinoma/patología , Análisis Mutacional de ADN , ADN de Neoplasias/análisis , Humanos , Neoplasias Nasofaríngeas/patología , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple
9.
Sci Rep ; 7: 42487, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28209993

RESUMEN

Endothelial dysfunction is characterised by aberrant redox signalling and an inflammatory phenotype. Shear stress antagonises endothelial dysfunction by increasing nitric oxide formation, activating anti-inflammatory pathways and suppressing inflammatory pathways. The TAK1 (MAP3K7) is a key mediator of inflammation and non-canonical TGF-ß signalling. While the individual roles of TAK1, ERK5 (MAPK7) and TGF-ß pathways in endothelial cell regulation are well characterised, an integrative understanding of the orchestration of these pathways and their crosstalk with the redox system under shear stress is lacking. We hypothesised that shear stress counteracts the inflammatory effects of oxidative stress and TGF-ß1 on endothelial cells by restoring redox balance and repressing the TAK1 pathway. Using human umbilical vein endothelial cells, we here show that TGF-ß1 aggravates oxidative stress-mediated inflammatory activation and that shear stress activates ERK5 signalling while attenuating TGF-ß signalling. ERK5 activation restores redox balance, but fails to repress the inflammatory effect of TGF-ß1 which is suppressed upon TAK1 inhibition. In conclusion, shear stress counteracts endothelial dysfunction by suppressing the pro-inflammatory non-canonical TGF-ß pathway and by activating the ERK5 pathway which restores redox signalling. We propose that a pharmacological compound that abates TGF-ß signalling and enhances ERK5 signalling may be useful to counteract endothelial dysfunction.


Asunto(s)
Células Endoteliales/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Estrés Oxidativo , Fenotipo , Resistencia al Corte , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Biomarcadores , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mediadores de Inflamación/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Modelos Biológicos , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología
10.
Cardiovasc Res ; 108(3): 377-86, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26084310

RESUMEN

AIMS: Neointimal hyperplasia is a common feature of fibro-proliferative vascular disease and characterizes initial stages of atherosclerosis. Neointimal lesions mainly comprise smooth muscle-like cells. The presence of these lesions is related to local differences in shear stress. Neointimal cells may arise through migration and proliferation of smooth muscle cells from the media. However, a role for the endothelium as a source of smooth muscle-like cells has largely been disregarded. Here, we investigated the role of endothelial-to-mesenchymal transition (EndMT) in neointimal hyperplasia and atherogenesis, and studied its modulation by shear stress. METHODS AND RESULTS: In human atherosclerotic plaques and porcine aortic tissues, myo-endothelial cells were identified, suggestive for EndMT. Flow disturbance by thoracic-aortic constriction in mice similarly showed the presence of myo-endothelial cells specifically in regions exposed to disturbed flow. While uniform laminar shear stress (LSS) was found to inhibit EndMT, endothelial cells exposed to disturbed flow underwent EndMT, in vitro and in vivo, and showed atherogenic differentiation. Gain- and loss-of-function studies using a constitutive active mutant of MEK5 and short hairpins targeting ERK5 established a pivotal role for ERK5 signalling in the inhibition of EndMT. CONCLUSION: Together, these data suggest that EndMT contributes to neointimal hyperplasia and induces atherogenic differentiation of endothelial cells. Importantly, we uncovered that EndMT is modulated by shear stress in an ERK5-dependent manner. These findings provide new insights in the role of adverse endothelial plasticity in vascular disease and identify a novel atheroprotective mechanism of uniform LSS, namely inhibition of EndMT.


Asunto(s)
Enfermedades de la Aorta/patología , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/patología , Proliferación Celular , Células Endoteliales/patología , Transición Epitelial-Mesenquimal , Mecanotransducción Celular , Placa Aterosclerótica , Remodelación Vascular , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Aorta Torácica/fisiopatología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/fisiopatología , Arterias Carótidas/metabolismo , Arterias Carótidas/fisiopatología , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/fisiopatología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Fibrosis , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , MAP Quinasa Quinasa 5/genética , MAP Quinasa Quinasa 5/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteína Quinasa 7 Activada por Mitógenos/genética , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Neointima , Interferencia de ARN , Flujo Sanguíneo Regional , Estrés Mecánico , Porcinos , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA