Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 236
Filtrar
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 ; 24(12)2023 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-37373471

RESUMEN

Neural stem cells (NSCs) persist in the subgranular zone (SGZ) throughout the lifespan and hold immense potential for the repair and regeneration of the central nervous system, including hippocampal-related diseases. Several studies have demonstrated that cellular communication network protein 3 (CCN3) regulates multiple types of stem cells. However, the role of CCN3 in NSCs remains unknown. In this study, we identified CCN3 expression in mouse hippocampal NSCs and observed that supplementing CCN3 improved cell viability in a concentration-dependent manner. Additionally, in vivo results showed that the injection of CCN3 in the dentate gyrus (DG) increased Ki-67- and SOX2-positive cells while decreasing neuron-specific class III beta-tubulin (Tuj1) and doublecortin (DCX)-positive cells. Consistently with the in vivo results, supplementing CCN3 in the medium increased the number of BrdU and Ki-67 cells and the proliferation index but decreased the number of Tuj1 and DCX cells. Conversely, both the in vivo and in vitro knockdown of the Ccn3 gene in NSCs had opposite effects. Further investigations revealed that CCN3 promoted cleaved Notch1 (NICD) expression, leading to the suppression of PTEN expression and eventual promotion of AKT activation. In contrast, Ccn3 knockdown inhibited the activation of the Notch/PTEN/AKT pathway. Finally, the effects of changes in CCN3 protein expression on NSC proliferation and differentiation were eliminated by FLI-06 (a Notch inhibitor) and VO-OH (a PTEN inhibitor). Our findings imply that while promoting proliferation, CCN3 inhibits the neuronal differentiation of mouse hippocampal NSCs and that the Notch/PTEN/AKT pathway may be a potential intracellular target of CCN3. Our findings may help develop strategies to enhance the intrinsic potential for brain regeneration after injuries, particularly stem cell treatment for hippocampal-related diseases.


Asunto(s)
Proteína Hiperexpresada del Nefroblastoma , Células-Madre Neurales , Proteínas Proto-Oncogénicas c-akt , Animales , Ratones , Diferenciación Celular , Proliferación Celular , Hipocampo/metabolismo , Antígeno Ki-67/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
3.
Am J Physiol Lung Cell Mol Physiol ; 324(2): L154-L168, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36573684

RESUMEN

Aberrant vascular remodeling contributes to the progression of many aging-associated diseases, including idiopathic pulmonary fibrosis (IPF), where heterogeneous capillary density, endothelial transcriptional alterations, and increased vascular permeability correlate with poor disease outcomes. Thus, identifying disease-driving mechanisms in the pulmonary vasculature may be a promising strategy to limit IPF progression. Here, we identified Ccn3 as an endothelial-derived factor that is upregulated in resolving but not in persistent lung fibrosis in mice, and whose function is critical for vascular homeostasis and repair. Loss and gain of function experiments were carried out to test the role of CCN3 in lung microvascular endothelial function in vitro through RNAi and the addition of recombinant human CCN3 protein, respectively. Endothelial migration, permeability, proliferation, and in vitro angiogenesis were tested in cultured human lung microvascular endothelial cells (ECs). Loss of CCN3 in lung ECs resulted in transcriptional alterations along with impaired wound-healing responses, in vitro angiogenesis, barrier integrity as well as an increased profibrotic activity through paracrine signals, whereas the addition of recombinant CCN3 augmented endothelial function. Altogether, our results demonstrate that the matricellular protein CCN3 plays an important role in lung endothelial function and could serve as a promising therapeutic target to facilitate vascular repair and promote lung fibrosis resolution.


Asunto(s)
Fibrosis Pulmonar , Ratones , Humanos , Animales , Células Endoteliales/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Células Cultivadas , Pulmón/metabolismo
4.
Int J Mol Sci ; 23(11)2022 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-35682564

RESUMEN

Cellular communication network factor (CCN) 2 and 3 are the members of the CCN family that conduct the harmonized development of a variety of tissues and organs under interaction with multiple biomolecules in the microenvironment. Despite their striking structural similarities, these two members show contrastive molecular functions as well as temporospatial emergence in living tissues. Typically, CCN2 promotes cell growth, whereas CCN3 restrains it. Where CCN2 is produced, CCN3 disappears. Nevertheless, these two proteins collaborate together to execute their mission in a yin-yang fashion. The apparent functional counteractions of CCN2 and CCN3 can be ascribed to their direct molecular interaction and interference over the cofactors that are shared by the two. Recent studies have revealed the mutual negative regulation systems between CCN2 and CCN3. Moreover, the simultaneous and bidirectional regulatory system of CCN2 and CCN3 is also being clarified. It is of particular note that these regulations were found to be closely associated with glycolysis, a fundamental procedure of energy metabolism. Here, the molecular interplay and metabolic gene regulation that enable the yin-yang collaboration of CCN2 and CCN3 typically found in cartilage development/regeneration and fibrosis are described.


Asunto(s)
Proteína Hiperexpresada del Nefroblastoma , Yin-Yang , Proliferación Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibrosis , Regulación de la Expresión Génica , Humanos , Proteína Hiperexpresada del Nefroblastoma/genética , Proteína Hiperexpresada del Nefroblastoma/metabolismo
5.
Genes (Basel) ; 12(11)2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34828265

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) is a common type of human cancer with a poor prognosis, and investigating the potential molecular mechanisms that can contribute to gene diagnosis and therapy. Herein, based on the recently concerned vertebrate-specific Cyr61/CTGF/NOV (CCN) gene family because of its important roles in diverse diseases, we obtained NOV/CCN3 to query for its potential roles in tumorigenesis via bioinformatics analysis. Experimental validations confirmed that both NOV mRNA and protein are up-regulated in two ICC cell lines, suggesting that it may promote cell migration and invasion by promoting EMT. To elucidate the detailed regulatory mechanism, miR-92a-3p is screened and identified as a negative regulatory small RNA targeting NOV, and further experimental validation demonstrates that miR-92a-3p contributes to NOV-mediated migration and invasion of ICC via the Notch signaling pathway. Our study reveals that NOV may be a potential target for diagnosing and treating ICC, which will provide experimental data and molecular theoretical foundation for cancer treatment, particularly for future precision medicine.


Asunto(s)
Neoplasias de los Conductos Biliares/patología , Movimiento Celular/genética , Colangiocarcinoma/patología , MicroARNs/fisiología , Proteína Hiperexpresada del Nefroblastoma/fisiología , Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos/efectos de los fármacos , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Colangiocarcinoma/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/genética , Invasividad Neoplásica , Proteína Hiperexpresada del Nefroblastoma/genética , ARN Interferente Pequeño/farmacología
6.
Mediators Inflamm ; 2021: 5576059, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34393649

RESUMEN

The CCN proteins are a family of extracellular matrix- (ECM-) associated proteins which currently consist of six secreted proteins (CCN1-6). CCN3 protein, also known as nephroblastoma overexpressed protein (NOV), is a member of the CCN family with multiple biological functions, implicated in major cellular processes such as cell growth, migration, and differentiation. Recently, CCN3 has emerged as a critical regulator in a variety of diseases, including immune-related diseases, including rheumatology arthritis, osteoarthritis, and systemic sclerosis. In this review, we will briefly introduce the structure and function of the CCN3 protein and summarize the roles of CCN3 in immune-related diseases, which is essential to understand the functions of the CCN3 in immune-related diseases.


Asunto(s)
Sistema Inmunológico/fisiopatología , Proteína Hiperexpresada del Nefroblastoma/fisiología , Animales , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Fibrosis , Glomerulonefritis/inmunología , Glomerulonefritis/metabolismo , Humanos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Osteoartritis/inmunología , Osteoartritis/metabolismo , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/metabolismo
7.
Mol Immunol ; 137: 163-173, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34256324

RESUMEN

BACKGROUND: Resveratrol improves insulin-resistance (IR) of gestational diabetes mellitus (GDM) mice. Low-expressed miR-23a-3p in diabetes patients regulates IR of adipocytes. Hence, we speculated the effect of Res on GDM mice was realized through regulating miR-23a-3p. METHODS: The GDM model was established in mice by high-fat diet, treated with miR-23a-3p antagomiR, and further performed with glucose and insulin tolerance tests. The bodyweight, serum glucose and serum insulin, and the expressions of miR-23a-3p and nephroblastoma overexpressed (NOV) in mouse adipose tissues were detected. MiR-23a-3p target was identified by Starbase and dual-luciferase reporter. Then, an IR adipocyte model was established by dexamethasone-inducing and further treated with Resveratrol or transfected with miR-23a-3p inhibitor or siNOV. The cell glucose intake was detected by radioimmunoassay. The expressions of miR-23a-3p, NOV, Adiponectin, Leptin, p-PI3K, PI3K, p-Akt, and Akt in the adipocytes were determined by qPCR or Western blot. RESULTS: Resveratrol decreased bodyweight, glucose level, insulin level, and the expressions of miR-23a-3p and NOV in the GDM mice, which was reversed by miR-23a-3p antagomiR. MiR-23a-3p targeted NOV. Resveratrol increased the glucose intake and the expressions of miR-23a-3p, Adiponectin, Leptin, p-PI3K, and p-Akt, decreased NOV expression in the IR adipocytes. The effect of the miR-23a-3p inhibitor on adipocytes with IR was opposite to Resveratrol, and the effects siNOV was the same as Resveratrol, except for its effect on miR-23a-3p expression. Effect of Res on the adipocytes with IR was counteracted by miR-23a-3p inhibitor whose effect was reversed by siNOV. CONCLUSION: Resveratrol ameliorated glucose uptake and lipid metabolism of the GDM mice and adipocytes with IR by regulating miR-23a-3p/NOV axis.


Asunto(s)
Adipocitos/efectos de los fármacos , Diabetes Gestacional/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos/efectos de los fármacos , MicroARNs/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Resveratrol/farmacología , Adipocitos/metabolismo , Animales , Diabetes Mellitus/metabolismo , Diabetes Mellitus Experimental/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Insulina/metabolismo , Ratones , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tumor de Wilms/tratamiento farmacológico , Tumor de Wilms/metabolismo
8.
Cancer Treat Res Commun ; 28: 100381, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33946014

RESUMEN

BACKGROUND: Osteosarcoma is the most prevalent type of primary bone sarcoma and is the major cause of deaths associated with cancer in children and adolescents. Despite novel and innovative therapies, early diagnosis of the osteosarcoma is still critically needed. Our study aimed to analyse the CCN3 proteins as a diagnostic marker and correlate their expression level with the severity of primary osteosarcoma patients. METHODS: In this prospective case-control study, after ethical clearance and informed consent, a total of 35 cases with primary osteosarcoma and ten otherwise healthy controls were enroled according to our strict inclusion-exclusion criteria. Tissue samples were collected during biopsy procedures in suspected cases and in controls during bone grafting procedures. The CCN3 expression level was measured by the western blotting assay. The clinic-radiological examinations were done in cases and graded according to the AJCC classification. Comparisons of CCN3 expression were measured between cases and controls, followed by correlation of their expression level with severity/grade of osteosarcoma in cases. RESULTS: All the demographic parameters showed insignificant differences. The CCN3 protein expressions were significantly upregulated in tissue samples of osteosarcoma patients (cases) compared to controls. The mean difference (p<0.0001) in CCN3 protein expression between cases' and controls' bony tissues was significant but showed insignificant correlation with the different grades of osteosarcoma. CONCLUSIONS: The upregulated CCN3 protein expression in osteosarcoma tissue along with significant differential manifestation in accordance with different grades of osteosarcoma make CCN3 suitable for a potential diagnostic biomarker. However, the author recommends further extensive multi-centric collaborative studies to increase our study reliability and generalizability.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/diagnóstico , Neoplasias Pulmonares/diagnóstico , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Osteosarcoma/diagnóstico , Adolescente , Adulto , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/cirugía , Estudios de Casos y Controles , Niño , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/cirugía , Índice de Severidad de la Enfermedad , Tomografía Computarizada por Rayos X , Adulto Joven
9.
J Cell Physiol ; 236(10): 6884-6896, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33655492

RESUMEN

Cellular communication network factor (CCN) family members are multifunctional matricellular proteins that manipulate and integrate extracellular signals. In our previous studies investigating the role of CCN family members in cellular metabolism, we found three members that might be under the regulation of energy metabolism. In this study, we confirmed that CCN2 and CCN3 are the only members that are tightly regulated by glycolysis in human chondrocytic cells. Interestingly, CCN3 was induced under a variety of impaired glycolytic conditions. This CCN3 induction was also observed in two breast cancer cell lines with a distinct phenotype, suggesting a basic role of CCN3 in cellular metabolism. Reporter gene assays indicated a transcriptional regulation mediated by an enhancer in the proximal promoter region. As a result of analyses in silico, we specified regulatory factor binding to the X-box 1 (RFX1) as a candidate that mediated the transcriptional activation by impaired glycolysis. Indeed, the inhibition of glycolysis induced the expression of RFX1, and RFX1 silencing nullified the CCN3 induction by impaired glycolysis. Subsequent experiments with an anti-CCN3 antibody indicated that CCN3 supported the survival of chondrocytes under impaired glycolysis. Consistent with these findings in vitro, abundant CCN3 production by chondrocytes in the deep zones of developing epiphysial cartilage, which are located far away from the synovial fluid, was confirmed in vivo. Our present study uncovered that RFX1 is the mediator that enables CCN3 induction upon cellular starvation, which may eventually assist chondrocytes in retaining their viability, even when there is an energy supply shortage.


Asunto(s)
Condrocitos/metabolismo , Glucólisis , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Factor Regulador X1/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Condrocitos/efectos de los fármacos , Regulación de la Expresión Génica , Edad Gestacional , Glucólisis/efectos de los fármacos , Humanos , Articulaciones/embriología , Articulaciones/metabolismo , Ratones Endogámicos BALB C , Proteína Hiperexpresada del Nefroblastoma/genética , Factor Regulador X1/genética , Fluoruro de Sodio/farmacología
10.
Am J Physiol Endocrinol Metab ; 320(4): E786-E796, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33586490

RESUMEN

We investigated the expression levels of nephroblastoma overexpressed [NOV or CCN3 (cellular communication network factor 3)] in the serum and placenta of pregnant women and of pregnant mice fed a high-fat diet (HFD), and its effect on placental glucose transporter 3 (GLUT3) expression, to examine its role in gestational diabetes mellitus (GDM). NOV/CCN3 expression was increased in the mouse serum during pregnancy. At gestational day 18, NOV/CCN3 protein expression was increased in the serum and placenta of the HFD mice compared with that of mice fed a normal diet. Compared with non-GDM patients, the patients with GDM had significantly increased serum NOV/CCN3 protein expression and placental NOV/CCN3 mRNA expression. Therefore, we hypothesized that NOV/CCN3 signaling may be involved in the pathogenesis of GDM. We administered NOV/CCN3 recombinant protein via intraperitoneal injections to pregnant mice fed HFD or normal diet. NOV/CCN3 overexpression led to glucose intolerance. Combined with the HFD, NOV/CCN3 exacerbated glucose intolerance and caused insulin resistance. NOV/CCN3 upregulates GLUT3 expression and affects the mammalian target of rapamycin (mTOR) pathway in the GDM environment in vivo and in vitro. In summary, our results demonstrate, for the first time, the molecular mechanism of NOV/CCN3 signaling in maternal metabolism to regulate glucose balance during pregnancy. NOV/CCN3 may be a potential target for detecting and treating GDM.NEW & NOTEWORTHY NOV/CCN3 regulates glucose homeostasis in mice during pregnancy. NOV/CCN3 upregulates GLUT3 expression and affects the mTOR pathway in the GDM environment in vivo and in vitro.


Asunto(s)
Dieta Alta en Grasa , Transportador de Glucosa de Tipo 3/genética , Proteína Hiperexpresada del Nefroblastoma/genética , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Cultivadas , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Grasas de la Dieta/farmacología , Femenino , Glucosa/metabolismo , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Humanos , Fenómenos Fisiologicos Nutricionales Maternos/efectos de los fármacos , Fenómenos Fisiologicos Nutricionales Maternos/genética , Ratones , Ratones Endogámicos C57BL , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Embarazo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 41(1): 79-86, 2021 Jan 30.
Artículo en Chino | MEDLINE | ID: mdl-33509757

RESUMEN

OBJECTIVE: To investigate the role of NOV/CCN3 in regulating the proliferation of mesenchymal stem cells (MSCs) and its regulatory mechanism and assess the value of CCN3 as a proliferative factor in bone tissue engineering. METHODS: Mouse embryonic fibroblasts (MEFs) were used as the MSC model, in which CCN3 expression was up-regulated and downregulated by transfection with the recombinant adenovirus vectors Ad-CCN3 and Ad-siCCN3, respectively. Flow cytometry was used to analyze the changes in cell cycle and apoptosis of the transfected cells. Western blotting was used to detect the expression levels of the proliferation indicators (PCNA, cyclin E, and cyclin B1) and the apoptosis indicators (Bax and Bcl-2) to assess the effect of modulation of CCN3 expression on MEF proliferation and apoptosis. CCN3 protein secretion by the cells was detected using ELISA. RT-qPCR and Western blotting were employed to analyze the changes in the expressions of Notch1, ligand DLL1, the downstream key proteins or genes (Hey1, P300, H3K9) and MAPK pathway-related proteins ERK1+2 and p-ERK1+2. RESULTS: Flow cytometry showed that compared with the control cells, MEFs transfected with Ad-CCN3 exhibited significantly increased cell proliferation index (P < 0.01) and lowered cell apoptosis rate (P < 0.05) with obviously enhanced expressions of PCNA, cyclin E and Bcl-2 proteins (P < 0.05). The results of RT-qPCR and Western blotting demonstrated that CCN3 overexpression significantly promoted the expression of Notch1 in the Notch signaling pathway (P < 0.001), inhibited the expressions of DLL1, Hey1, P300, and H3K9 (P < 0.05), and increased the protein expressions of ERK1+2 and P-ERk1+2 in the MAPK pathway (P < 0.01). CONCLUSIONS: CCN3 over-expression promotes the proliferation and inhibits apoptosis of MEFs possibly by inhibiting the classical Notch signaling pathway and activating the MAPK pathway via binding to Notch1, suggesting the potential value of CCN3 as a proliferative factor of MSCs in bone tissue engineering.


Asunto(s)
Apoptosis , Proteína Hiperexpresada del Nefroblastoma , Animales , Ciclo Celular , Proliferación Celular , Fibroblastos , Ratones
12.
Carcinogenesis ; 42(2): 190-201, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33313663

RESUMEN

Among cancer cells, there are specific cell populations of whose activities are comparable to those of stem cells in normal tissues, and for whom the levels of cell dedifferentiation are reported to correlate with poor prognosis. Information concerning the mechanisms that modulate the stemness like traits of cancer cells is limited. Therefore, we examined five gastric cancer cell lines and isolated gastric oncospheres from three gastric cancer cell lines. The gastric cancer cells that expanded in the spheres expressed relatively elevated proportion of CD44, which is a marker of gastric cancer stem cells (CSCs), and displayed many properties of CSCs, for example: chemoresistance, tumorigenicity and epithelial-mesenchymal transition (EMT) acquisition. SNAIL, which is a key factor in EMT, was highly expressed in the gastric spheres. Microarray analysis in gastric cancer cell line HGC27 showed that CCN3 and NEFL displayed the greatest differential expression by knocking down of SNAIL; the former was upregulated and the latter downregulated, respectively. Downregulation of CCN3 and upregulation of NEFL gene expression impaired the SNAIL-dependent EMT activity: high tumorigenicity, and chemoresistance in gastric cancer cells. Thus, approach that disrupts SNAIL/CCN3/NEFL axis may be credible in inhibiting gastric cancer development.


Asunto(s)
Carcinogénesis/genética , Proteína Hiperexpresada del Nefroblastoma/genética , Proteínas de Neurofilamentos/genética , Factores de Transcripción de la Familia Snail/metabolismo , Neoplasias Gástricas/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/genética , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Células Madre Neoplásicas/patología , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Proteínas de Neurofilamentos/metabolismo , Transducción de Señal/genética , Factores de Transcripción de la Familia Snail/genética , Estómago/patología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Environ Toxicol ; 36(3): 425-432, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33107671

RESUMEN

Prostate cancer osteoblastic bone metastases are incurable and associated with chronic bone pain and a high mortality rate. Osteoclast-targeting drugs intended to prevent skeletal-related events associated with prostate cancer bone metastases do not prolong overall survival. Improved understanding of the bone-derived factors that contribute to prostate cancer osteoblastic bone metastases is required to design treatments that will improve morbidities and overall survival. Activated osteoblasts stimulate prostate cancer growth in bone. In this study, we report that prostate cancer conditioned medium (CM) promoted bone morphogenetic protein (BMP)-2, -4 and -7 production and the expression of osteogenic transcription factors Runx2 and osterix in osteoblasts. Treating the prostate cancer CM with antibody against CCN3 (nephroblastoma-overexpressed), a cysteine-rich protein that belongs to the CCN family, reduced all of these increases. Incubation of osteoblasts with CCN3 facilitated phosphorylation of GSK3ß and ß-catenin. GSK3ß and ß-catenin inhibitors or siRNAs all abolished CCN3-induced promotion of BMPs, Runx2 and osterix expression in osteoblasts. Our results indicate that prostate cancer-secreted CCN3 enhances BMP, Runx2 and osterix expression in osteoblasts via the GSK3ß and ß-catenin signaling pathways. This understanding of the role played by CCN3 in osteoblastic prostate bone metastasis may lead to more efficient targeted therapies.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Neoplasias de la Próstata/metabolismo , beta Catenina/metabolismo , Animales , Neoplasias Óseas/patología , Diferenciación Celular , Humanos , Masculino , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Fosforilación , Transducción de Señal
14.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-880831

RESUMEN

OBJECTIVE@#To investigate the role of NOV/CCN3 in regulating the proliferation of mesenchymal stem cells (MSCs) and its regulatory mechanism and assess the value of CCN3 as a proliferative factor in bone tissue engineering.@*METHODS@#Mouse embryonic fibroblasts (MEFs) were used as the MSC model, in which CCN3 expression was up-regulated and downregulated by transfection with the recombinant adenovirus vectors Ad-CCN3 and Ad-siCCN3, respectively. Flow cytometry was used to analyze the changes in cell cycle and apoptosis of the transfected cells. Western blotting was used to detect the expression levels of the proliferation indicators (PCNA, cyclin E, and cyclin B1) and the apoptosis indicators (Bax and Bcl-2) to assess the effect of modulation of CCN3 expression on MEF proliferation and apoptosis. CCN3 protein secretion by the cells was detected using ELISA. RT-qPCR and Western blotting were employed to analyze the changes in the expressions of Notch1, ligand DLL1, the downstream key proteins or genes (Hey1, P300, H3K9) and MAPK pathway-related proteins ERK1+2 and p-ERK1+2.@*RESULTS@#Flow cytometry showed that compared with the control cells, MEFs transfected with Ad-CCN3 exhibited significantly increased cell proliferation index (@*CONCLUSIONS@#CCN3 over-expression promotes the proliferation and inhibits apoptosis of MEFs possibly by inhibiting the classical Notch signaling pathway and activating the MAPK pathway


Asunto(s)
Animales , Ratones , Apoptosis , Ciclo Celular , Proliferación Celular , Fibroblastos , Proteína Hiperexpresada del Nefroblastoma
15.
Front Endocrinol (Lausanne) ; 11: 597549, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33304321

RESUMEN

Objectives: An adequate development of the placenta includes trophoblast differentiation with the processes of trophoblast migration, invasion, cellular senescence and apoptosis which are all crucial to establishing a successful pregnancy. Altered placental development and function lead to placental diseases such as preeclampsia (PE) which is mainly characterized by insufficient trophoblast invasion and abnormally invasive placenta (AIP) disorders (Placenta accreta, increta, or percreta) which are characterized by excessive trophoblast invasion. Both of them will cause maternal and fetal morbidity/mortality. However, the etiology of these diseases is still unclear. Our previous study has shown that the matricellular protein nephroblastoma overexpressed (NOV, CCN3) induces G0/G1 cell cycle arrest, drives trophoblast cells into senescence and activates FAK and Akt kinases resulting in reduced cell proliferation and enhanced migration capability of the human trophoblast cell line SGHPL-5. The present study focuses on whether CCN3 can alter cell cycle-regulated pathways associated with trophoblast senescence and invasion activity in pathological versus gestational age-matched control placentas. Methods: Cell cycle regulator proteins were investigated by immunoblotting and qPCR. For localization of CCN3, p16, p21, and Cyclin D1 proteins, co-immunohistochemistry was performed. Results: In early-onset PE placentas, CCN3 was expressed at a significantly lower level compared to gestational age-matched controls. The decrease of CCN3 level is associated with an increase in p53, Cyclin E1 and pRb protein expression, whereas the level of cleaved Notch-1, p21, Cyclin D1, pFAK, pAKT, and pmTOR protein decreased. In term AIP placentas, the expression of CCN3 was significantly increased compared to matched term controls. This increase was correlated to an increase in p53, p16, p21, Cyclin D1, cleaved Notch-1, pFAK, pAkt, and pmTOR whereas pRb was significantly decreased. However, in late PE and early AIP placentas, no significant differences in CCN3, p16, p21, Cyclin D1, p53, and cleaved Notch-1 expression were found when matched to appropriate controls. Conclusions: CCN3 expression levels are correlated to markers of cell cycle arrest oppositely in PE and AIP by activating the FAK/AKT pathway in AIP or down-regulating in PE. This may be one mechanism to explain the different pathological features of placental diseases, PE and AIP.


Asunto(s)
Biomarcadores/metabolismo , Puntos de Control del Ciclo Celular , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Enfermedades Placentarias/patología , Placenta/patología , Preeclampsia/patología , Adulto , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Senescencia Celular , Femenino , Humanos , Placenta/metabolismo , Enfermedades Placentarias/metabolismo , Preeclampsia/metabolismo , Embarazo
16.
J Neuroinflammation ; 17(1): 349, 2020 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-33222687

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is an immune-mediated disease that damages myelin in the central nervous system (CNS). We investigated the profile of CCN3, a known regulator of immune function and a potential mediator of myelin regeneration, in multiple sclerosis in the context of disease state and disease-modifying treatment. METHODS: CCN3 expression was analysed in plasma, immune cells, CSF and brain tissue of MS patient groups and control subjects by ELISA, western blot, qPCR, histology and in situ hybridization. RESULTS: Plasma CCN3 levels were comparable between collective MS cohorts and controls but were significantly higher in progressive versus relapsing-remitting MS and between patients on interferon-ß versus natalizumab. Higher body mass index was associated with higher CCN3 levels in controls as reported previously, but this correlation was absent in MS patients. A significant positive correlation was found between CCN3 levels in matched plasma and CSF of MS patients which was absent in a comparator group of idiopathic intracranial hypertension patients. PBMCs and CD4+ T cells significantly upregulated CCN3 mRNA in MS patients versus controls. In the CNS, CCN3 was detected in neurons, astrocytes and blood vessels. Although overall levels of area immunoreactivity were comparable between non-affected, demyelinated and remyelinated tissue, the profile of expression varied dramatically. CONCLUSIONS: This investigation provides the first comprehensive profile of CCN3 expression in MS and provides rationale to determine if CCN3 contributes to neuroimmunological functions in the CNS.


Asunto(s)
Interferón beta/uso terapéutico , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Natalizumab/uso terapéutico , Proteína Hiperexpresada del Nefroblastoma/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteína Hiperexpresada del Nefroblastoma/genética , Resultado del Tratamiento
17.
Int J Mol Sci ; 21(20)2020 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-33066270

RESUMEN

Aging is a major risk factor of osteoarthritis, which is characterized by the degeneration of articular cartilage. CCN3, a member of the CCN family, is expressed in cartilage and has various physiological functions during chondrocyte development, differentiation, and regeneration. Here, we examine the role of CCN3 in cartilage maintenance. During aging, the expression of Ccn3 mRNA in mouse primary chondrocytes from knee cartilage increased and showed a positive correlation with p21 and p53 mRNA. Increased accumulation of CCN3 protein was confirmed. To analyze the effects of CCN3 in vitro, either primary cultured human articular chondrocytes or rat chondrosarcoma cell line (RCS) were used. Artificial senescence induced by H2O2 caused a dose-dependent increase in Ccn3 gene and CCN3 protein expression, along with enhanced expression of p21 and p53 mRNA and proteins, as well as SA-ß gal activity. Overexpression of CCN3 also enhanced p21 promoter activity via p53. Accordingly, the addition of recombinant CCN3 protein to the culture increased the expression of p21 and p53 mRNAs. We have produced cartilage-specific CCN3-overexpressing transgenic mice, and found degradative changes in knee joints within two months. Inflammatory gene expression was found even in the rib chondrocytes of three-month-old transgenic mice. Similar results were observed in human knee articular chondrocytes from patients at both mRNA and protein levels. These results indicate that CCN3 is a new senescence marker of chondrocytes, and the overexpression of CCN3 in cartilage may in part promote chondrocyte senescence, leading to the degeneration of articular cartilage through the induction of p53 and p21.


Asunto(s)
Cartílago Articular/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Osteoartritis de la Rodilla/metabolismo , Animales , Cartílago Articular/crecimiento & desarrollo , Línea Celular Tumoral , Células Cultivadas , Senescencia Celular , Condrocitos/metabolismo , Condrocitos/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína Hiperexpresada del Nefroblastoma/genética , Osteoartritis de la Rodilla/patología , Ratas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
18.
Proc Natl Acad Sci U S A ; 117(30): 18018-18028, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32651278

RESUMEN

CCN3 is a matricellular protein that promotes oligodendrocyte progenitor cell differentiation and myelination in vitro and ex vivo. CCN3 is therefore a candidate of interest in central nervous system (CNS) myelination and remyelination, and we sought to investigate the expression and role of CCN3 during these processes. We found CCN3 to be expressed predominantly by neurons in distinct areas of the CNS, primarily the cerebral cortex, hippocampus, amygdala, suprachiasmatic nuclei, anterior olfactory nuclei, and spinal cord gray matter. CCN3 was transiently up-regulated following demyelination in the brain of cuprizone-fed mice and spinal cord lesions of mice injected with lysolecithin. However, CCN3-/- mice did not exhibit significantly different numbers of oligodendroglia or differentiated oligodendrocytes in the healthy or remyelinating CNS, compared to WT controls. These results suggest that despite robust and dynamic expression in the CNS, CCN3 is not required for efficient myelination or remyelination in the murine CNS in vivo.


Asunto(s)
Sistema Nervioso Central/metabolismo , Enfermedades Desmielinizantes/etiología , Regulación de la Expresión Génica , Proteína Hiperexpresada del Nefroblastoma/genética , Remielinización/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Ratones , Vaina de Mielina/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología
19.
Pigment Cell Melanoma Res ; 33(6): 895-898, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32633087

RESUMEN

Systemic sclerosis (SSc) is a severe disease whose pathophysiology remains partly unknown, combining autoimmune, vascular, and fibrotic features. Recently, we evidenced a link between vasculopathy and pigmentary changes in SSc. CCN3 (NOV) is a matricellular protein implicated in both angiogenesis and pigmentation regulation, in particular melanocyte adhesion to the basal layer. We decided to study CCN3 expression in SSc epidermis. We show that in SSc patients with pigmentary changes compared to patients with normal pigmentation, CCN3 is specifically downregulated in situ in melanocytes and upregulated in keratinocytes. Moreover, the number of melanocytes is significantly decreased in SSc patients with a disease duration of more than 5 years compared to the other patients. Altogether, our findings could provide new insights on the mechanisms of pigmentary changes in SSc patients, as well as treatment adaptation in a personalized manner.


Asunto(s)
Epidermis/patología , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Esclerodermia Sistémica/patología , Pigmentación de la Piel , Femenino , Humanos , Masculino , Melanocitos/metabolismo , Melanocitos/patología , Persona de Mediana Edad
20.
J Immunol Res ; 2020: 3891425, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32455138

RESUMEN

Nephroblastoma overexpressed protein (NOV/CCN3), the early discovered member of the CCN family, has recently been suggested to be involved in a number of inflammatory processes, including wound healing, alveolar epithelial cell inflammation, cancer metastasis, and macrophage foam cell formation. However, the role of CCN3 in rheumatoid arthritis (RA), a classic autoimmune and inflammatory disease, remains elusive. RA is a chronic systemic autoimmune disease that eventually leads to cartilage and bone destruction and joint dysfunction. In this study, we investigated the potential of serum CCN3 as a biomarker for RA. The serum levels of CCN3 were measured by ELISA. The clinical and laboratory parameters were collected from a clinical record system, and disease activity was determined by joint disease activity score 28 (DAS28). Our results showed that the serum levels of CCN3 were significantly increased in RA patients compared to healthy controls. Furthermore, the CCN3 level was positively correlated with DAS28 (CRP), DAS28 (ESR), and the level of anti-CCP Ab, an autoantibody highly specific for RA. Furthermore, CCN3 showed a positive correlation with inflammatory cytokine IL-6, while no significant correlation with TNF-α was observed. These data suggest that CCN3 plays an important role in the development of RA and might be a potential disease activity biomarker for RA.


Asunto(s)
Artritis/inmunología , Biomarcadores/sangre , Inflamación/metabolismo , Interleucina-6/sangre , Proteína Hiperexpresada del Nefroblastoma/sangre , Adulto , Anciano , Anticuerpos Antiproteína Citrulinada/sangre , Artritis/diagnóstico , Proteína C-Reactiva/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...