Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain Res ; 1827: 148756, 2024 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-38199307

RESUMEN

Neural stem/progenitor cells (NSPCs) hold immense promise in clinical applications, yet the harsh conditions resulting from central nervous system (CNS) injuries, particularly oxidative stress, lead to the demise of both native and transplanted NSPCs. Cellular communication network factor 3 (CCN3) exhibits a protective effect against oxidative stress in various cell types. This study investigates the impact of CCN3 on NSPCs apoptosis induced by oxidative stress. To establish models of primary cultured mouse NSPCs under oxidative stress, we exposed them to 50 µM H2O2 for 4 h. Remarkably, pre-exposing CCN3 exacerbated the H2O2-induced decline in cell viability in a concentration-dependent manner. However, employing gene-targeted siRNA to inhibit CCN3 protected NSPCs against H2O2-induced cell death. Conversely, CCN3 replenishment reversed this protective effect, as evidenced by TUNEL staining, the ratio of Cleaved-caspase-3 to Pro-caspase-3, and Bcl-2/Bax. Further investigations revealed that CCN3 pretreatment increased the phosphorylation level of p38 MAPK, while silencing CCN3 diminished p38 MAPK activation. Ultimately, the impact of changes in CCN3 protein expression on H2O2-induced apoptosis was nullified using anisomycin (a p38 activator) and SB 203580 (a p38 inhibitor). Our findings suggest that CCN3 inhibition prevents H2O2-induced cell death in cultured mouse NSPCs via the p38 pathway. These discoveries may contribute to the development of strategies aimed at enhancing the survival of both endogenous and transplanted NSPCs following CNS oxidative stress insults.


Asunto(s)
Peróxido de Hidrógeno , Proteínas Quinasas p38 Activadas por Mitógenos , Ratones , Animales , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Peróxido de Hidrógeno/farmacología , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Proteína Hiperexpresada del Nefroblastoma/farmacología , Estrés Oxidativo , Apoptosis , Células Madre/metabolismo
2.
Int J Mol Sci ; 20(13)2019 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-31284378

RESUMEN

CCN3, otherwise known as the nephroblastoma overexpressed (NOV) protein, is a cysteine-rich protein that belongs to the CCN family and regulates several cellular functions. Osteoblasts are major bone-forming cells that undergo proliferation, mineralization, renewal, and repair during the bone formation process. We have previously reported that CCN3 increases bone morphogenetic protein 4 (BMP-4) production and bone mineralization in osteoblasts, although the role of CCN3 remains unclear with regard to osteogenic transcription factors (runt-related transcription factor 2 (Runx2) and osterix). Here, we used alizarin red-S and alkaline phosphatase staining to show that CCN3 enhances osteoblast differentiation. Stimulation of osteoblasts with CCN3 increases expression of osteogenic factors such as BMPs, Runx2, and osterix. Moreover, we found that the inhibition of miR-608 expression is involved in the effects of CCN3 and that incubation of osteoblasts with CCN3 promotes focal adhesion kinase (FAK) and Akt phosphorylation. Our results indicate that CCN3 promotes the expression of Runx2 and osterix in osteoblasts by inhibiting miR-608 expression via the FAK and Akt signaling pathways.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , MicroARNs/metabolismo , Proteína Hiperexpresada del Nefroblastoma/farmacología , Osteoblastos/metabolismo , Osteogénesis , Transducción de Señal , Factor de Transcripción Sp7/metabolismo , Regiones no Traducidas 3'/genética , Animales , Secuencia de Bases , Línea Celular , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , MicroARNs/genética , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos
3.
Mol Med Rep ; 13(3): 2017-22, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26795879

RESUMEN

The presence of apoptotic cells and loss of extracellular matrix (ECM) are common characteristics of degenerated cartilage endplates (CEPs). In addition, therapeutic efficacy is hampered by an incomplete understanding regarding the mechanisms underlying CEP homeostasis and degeneration. The CCN proteins have recently emerged as important regulators of cell­ECM interactions, and have been identified as key mediators of nucleus pulposus ECM composition and tissue homeostasis. However, whether CCN3 is associated with CEP homeostasis has yet to be elucidated. The present study aimed to investigate the effects of CCN3 on the apoptosis and ECM synthesis of CEP cells cultured under serum deprivation. Rat CEP cells were confirmed to be of the chondrocytic phenotype by toluidine blue staining. The mRNA expression levels of CCN3 were markedly increased, and a dose­dependent increase of apoptotic rate was detected under serum deprivation conditions following treatment with recombinant CCN3, whereas CCN3 did not exert a proapoptotic effect on cells cultured under normal conditions. Furthermore, CCN3­treated cells exhibited a decrease in the expression levels of aggrecan and collagen II in both groups. These results suggested that CCN3 may act as a regulator, rather than an initiator, of serum deprivation­induced cellular apoptosis, and that CCN3 has a catabolic effect on the mediation of ECM synthesis under both normal and serum deprivation conditions. Therefore, CCN3 may represent a novel therapeutic target for the prevention of CEP degeneration.


Asunto(s)
Cartílago Articular/patología , Condrocitos/metabolismo , Proteína Hiperexpresada del Nefroblastoma/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Células Cultivadas , Condrocitos/efectos de los fármacos , Medio de Cultivo Libre de Suero , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Fenotipo , Ratas Sprague-Dawley
4.
Oncol Rep ; 34(4): 2011-21, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26238193

RESUMEN

The nephroblastoma overexpressed (NOV) gene, a member of the CCN gene family that encodes secreted proteins involved in a variety of processes including tumorigenesis, is often altered in a variety of tumors, including osteosarcoma. Recent studies indicated that NOV promotes osteosarcoma metastasis, but its biological functions and molecular mechanisms on osteosarcoma proliferation have yet to be fully elucidated. The aim of the present study was to examine the role of NOV in osteosarcoma biology. Reverse transcription-polymerase chain reaction (RT-PCR) and western blot analysis were performed to characterize the endogenous expression of NOV in osteosarcoma cell lines. Recombinant adenovirus expressing NOV/siNOV (AdNOV/AdsiNOV) was used to infect osteosarcoma cell lines with a relatively low/high endogenous NOV expression to determine the functional relevance of NOV expression to osteosarcoma cell growth and migration in vitro, respectively. As a result, osteosarcoma cell proliferation was significantly reduced by NOV upregulation, indicated by 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltrazolium bromide (MTT), colony forming assay and cell cycle analysis. Cell apoptosis was markedly induced, as indicated by Hoechst 33258 staining assay and flow cytometry (FCM) detection. Despite the antiproliferative effect, NOV-transfected osteosarcoma cells exhibited increased migration ability. The possible molecular mechanisms underlying the biological role of NOV were also investigated. The results demonstrated that NOV increased the phosphorylation of p38 and c-Jun N-terminal kinase (JNK) mitogen-actived protein kinases (MAPKs) in osteosarcoma cell lines. When the phosphorylation of p38 and JNK were inhibited by SB203580 (p38 inhibitor) or SP600125 (JNK inhibitor), respectively, the NOV-induced proliferation inhibition and cell apoptosis were reversed. In conclusion, the results revealed that NOV regulates the tumor growth of osteosarcoma cells through activation of the MAPK signaling pathway and promotes osteosarcoma cell migration in vitro.


Asunto(s)
Neoplasias Óseas/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Hiperexpresada del Nefroblastoma/genética , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Osteosarcoma/metabolismo , Antracenos/farmacología , Apoptosis , Neoplasias Óseas/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Imidazoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Hiperexpresada del Nefroblastoma/farmacología , Osteosarcoma/genética , Fosforilación/efectos de los fármacos , Piridinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Am J Pathol ; 184(11): 2908-21, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25193594

RESUMEN

Fibrosis is at the core of the high morbidity and mortality rates associated with the complications of diabetes and obesity, including diabetic nephropathy (DN), without any US Food and Drug Administration-approved drugs with this specific target. We recently provided the first evidence that the matricellular protein CCN3 (official symbol NOV) functions in a reciprocal manner, acting on the profibrotic family member CCN2 to inhibit fibrosis in a mesangial cell model of DN. Herein, we used the BT/BR ob/ob mouse as a best model of human obesity and DN progression to determine whether recombinant human CCN3 could be used therapeutically, and the mechanisms involved. Eight weeks of thrice-weekly i.p. injections (0.604 and 6.04 µg/kg of recombinant human CCN3) beginning in early-stage DN completely blocked and/or reversed the up-regulation of mRNA expression of kidney cortex fibrosis genes (CCN2, Col1a2, TGF-ß1, and PAI-1) seen in placebo-treated diabetic mice. The treatment completely blocked glomerular fibrosis, as determined by altered mesangial expansion and deposition of laminin. Furthermore, it protected against, or reversed, podocyte loss and kidney function reduction (rise in plasma creatinine concentration); albuminuria was also greatly reduced. This study demonstrates the potential efficacy of recombinant human CCN3 treatment in DN and points to mechanisms operating at multiple levels or pathways, upstream (eg, protecting against cell injury) and downstream (eg, regulating CCN2 activity and extracellular matrix metabolism).


Asunto(s)
Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/patología , Fibrosis/tratamiento farmacológico , Riñón/efectos de los fármacos , Proteína Hiperexpresada del Nefroblastoma/uso terapéutico , Animales , Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/complicaciones , Fibrosis/patología , Fibrosis/prevención & control , Riñón/patología , Masculino , Ratones , Proteína Hiperexpresada del Nefroblastoma/farmacología , Obesidad/complicaciones , Obesidad/patología , Resultado del Tratamiento
6.
Int J Hematol ; 99(4): 393-406, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24563081

RESUMEN

Throughout life, hematopoietic stem cells (HSCs) sustain the blood cell supply through their capacities for self-renewal and multilineage differentiation. These processes are regulated within a specialized microenvironment termed the 'niche'. Here, we show a novel mechanism for regulating HSC function that is mediated by nephroblastoma overexpressed (Nov/CCN3), a matricellular protein member of the CCN family. We found that Nov contributes to the maintenance of long-term repopulating (LTR) activity through association with integrin αvß3 on HSCs. The resultant ß3 integrin outside-in signaling is dependent on thrombopoietin (TPO), a crucial cytokine involved in HSC maintenance. TPO was required for Nov binding to integrin αvß3, and stimulated Nov expression in HSCs. However, in the presence of IFNγ, a cytokine known to impair HSC function, not only was TPO-induced expression of Nov suppressed, but the LTR activity was conversely impaired by TPO-mediated ligation of integrin αvß3 with exogenous ligands, including Nov, as well. Thus, Nov/integrin αvß3-mediated maintenance of HSCs appears to be modulated by simultaneous stimulation by other cytokines. Our finding suggests that this system contributes to the regulation of HSCs within the bone marrow niche.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Integrina alfaVbeta3/metabolismo , Proteína Hiperexpresada del Nefroblastoma/genética , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Interferón gamma/farmacología , Ratones , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Proteína Hiperexpresada del Nefroblastoma/farmacología , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Trombopoyetina/farmacología
7.
Am J Pathol ; 180(5): 1979-90, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22538190

RESUMEN

In contrast to factors that promote mesangial cell proliferation, little is known about their endogenous inhibitors. During experimental mesangioproliferative nephritis, expression of the glomerular CCN3 (nephroblastoma overexpressed gene [NOV]) gene is reduced before the proliferative phase and increased in glomeruli and serum when mesangial cell proliferation subsides. To further elucidate its role in mesangioproliferative glomerulonephritis, CCN3 systemically was overexpressed by muscle electroporation in healthy or nephritic rats. This increased CCN3 serum concentrations more than threefold for up to 56 days. At day 5 after disease induction, CCN3-transfected rats showed an increase in glomerular endothelial area and in mRNA levels of the pro-angiogenic factors vascular endothelial growth factor and PDGF-C. At day 7, CCN3 overexpression decreased mesangial cell proliferation, including expression of α-smooth muscle actin and matrix accumulation of fibronectin and type IV collagen. In progressive nephritis (day 56), overexpression of CCN3 resulted in decreased albuminuria, glomerulosclerosis, and reduced cortical collagen type I accumulation. In healthy rat kidneys, overexpression of CCN3 induced no morphologic changes but regulated glomerular gene transcripts (reduced transcription of PDGF-B, PDGF-D, PDGF-receptor-ß, and fibronectin, and increased PDGF-receptor-α and PDGF-C mRNA). These data identify a dual role for CCN3 in experimental glomerulonephritis with pro-angiogenic and antimesangioproliferative effects. Manipulation of CCN3 may represent a novel approach to help repair glomerular endothelial damage and mesangioproliferative changes.


Asunto(s)
Glomerulonefritis Membranoproliferativa/metabolismo , Neovascularización Fisiológica/fisiología , Proteína Hiperexpresada del Nefroblastoma/fisiología , Actinas/metabolismo , Enfermedad Aguda , Inductores de la Angiogénesis/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo IV/metabolismo , Progresión de la Enfermedad , Electroquimioterapia/métodos , Células Endoteliales/efectos de los fármacos , Fibronectinas/metabolismo , Terapia Genética/métodos , Mesangio Glomerular/patología , Glomerulonefritis Membranoproliferativa/patología , Glomerulonefritis Membranoproliferativa/fisiopatología , Glomerulonefritis Membranoproliferativa/terapia , Humanos , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/efectos de los fármacos , Masculino , Células Mesangiales/patología , Músculo Esquelético/metabolismo , Proteína Hiperexpresada del Nefroblastoma/sangre , Proteína Hiperexpresada del Nefroblastoma/genética , Proteína Hiperexpresada del Nefroblastoma/farmacología , ARN Mensajero/genética , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Proteínas Recombinantes/farmacología
8.
Arthritis Rheum ; 63(10): 3022-31, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21618206

RESUMEN

OBJECTIVE: To investigate transforming growth factor ß (TGFß) regulation of CCN3 expression in cells of the nucleus pulposus. METHODS: Real-time reverse transcription-polymerase chain reaction and Western blot analyses were used to measure CCN3 expression in the nucleus pulposus. Transfections were used to measure the effect of Smad3, MAPKs, and activator protein 1 (AP-1) on TGFß-mediated CCN3 promoter activity. Lentiviral knockdown of Smad3 was performed to assess the role of Smad3 in CCN3 expression. RESULTS: CCN3 was expressed in embryonic and adult intervertebral discs. TGFß decreased the expression of CCN3 and suppressed its promoter activity in nucleus pulposus cells. DN-Smad3, Smad3 small interfering RNA, or DN-AP-1 had little effect on TGFß suppression of CCN3 promoter activity. However, p38 and ERK inhibitors blocked suppression of CCN3 by TGFß, suggesting involvement of these signaling pathways in the regulation of CCN3. Interestingly, overexpression of Smad3 in the absence of TGFß increased CCN3 promoter activity. We validated the role of Smad3 in controlling CCN3 expression in Smad3-null mice and in nucleus pulposus cells transduced with lentiviral short hairpin Smad3. In terms of function, treatment with recombinant CCN3 showed a dose-dependent decrease in the proliferation of nucleus pulposus cells. Moreover, CCN3-treated cells showed a decrease in aggrecan, versican, CCN2, and type I collagen expression. CONCLUSION: The opposing effect of TGFß on CCN2 and CCN3 expression and the suppression of CCN2 by CCN3 in nucleus pulposus cells further the paradigm that these CCN proteins form an interacting triad, which is possibly important in maintaining extracellular matrix homeostasis and cell numbers.


Asunto(s)
Disco Intervertebral/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Disco Intervertebral/citología , Disco Intervertebral/efectos de los fármacos , Ratones , Proteína Hiperexpresada del Nefroblastoma/genética , Proteína Hiperexpresada del Nefroblastoma/farmacología , Regiones Promotoras Genéticas , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factor de Crecimiento Transformador beta/genética
9.
Glia ; 58(12): 1510-21, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20648642

RESUMEN

Increasing evidence suggests that CCN matricellular proteins play important roles in inflammation. One of the major cell types that handle inflammation in the brain is the astrocyte, which, upon activation, dramatically increases its production of cytokines and chemokines. Here, we report that NOV/CCN3, added to primary cultured rat brain astrocytes, markedly increased the expression of CCL2 and CXCL1 chemokines, as indicated by ELISA and RT-qPCR assays. This effect was selective, as the production of thirteen other cytokines and chemokines was not affected by NOV. NOV expression by astrocytes was demonstrated by immunocytochemistry and Western blot analysis, and astrocyte transfection with NOV small interfering RNA (siRNA) markedly decreased CXCL1 and CCL2 production, indicating that endogenous NOV played a major role in the control of astrocytic chemokine synthesis. NOV was shown to mediate several of its actions through integrins. Here, we observed that siRNAs against integrins beta1 and beta5 decreased basal and abrogated NOV-stimulated astrocyte expression of CCL2 and CXCL1, respectively. Using a panel of kinase inhibitors, we demonstrated that NOV action on CCL2 and CXCL1 production involved a Rho/ROCK/JNK/NF-kappaB and a Rho/qROCK/p38/NF-kappaB pathway, respectively. Thus, distinct integrins and signaling mechanisms are involved in NOV-induced production of CCL2 and CXCL1 in astrocytes. Finally, astrocytic expression of NOV was detected in rat brain tissue sections, and NOV intracerebral injection increased CCL2 and CXCL1 brain levels in vivo. Altogether, our data shed light on the signaling pathways operated by NOV and strongly suggest that NOV mediates astrocyte activation and, therefore, might play a role in neuroinflammation.


Asunto(s)
Astrocitos/efectos de los fármacos , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/metabolismo , Cadenas beta de Integrinas/metabolismo , Integrina beta1/metabolismo , Proteína Hiperexpresada del Nefroblastoma/farmacología , Regulación hacia Arriba/efectos de los fármacos , Animales , Astrocitos/metabolismo , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Movimiento Celular , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CXCL1/genética , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Masculino , Proteína Hiperexpresada del Nefroblastoma/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA