Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26
Filtrer
1.
Neurosci Res ; 203: 42-50, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38154662

RÉSUMÉ

Numerous evidences showed that human umbilical cord blood (UCB) mononuclear cells were a promising approach for the therapy of ischemic stroke(IS). The effect of stage-specific embryonic antigen 3 (SSEA3)positive subpopulation in UCB was not investigated in IS. In this study, we isolated SSEA3 positive cells from healthy UCB mononuclear cells, which comprised about 7.01% of the total UCB-mononuclear cells. Flow cytometry analysis revealed that SSEA3(+)UCB cells were almost positive for CD44 and CD45, and negative for CD73, CD90 and CD105. The expression of Oct3/4 in SSEA3(+)UCB cells were higher than that in UCB. SSEA3(+)UCB cells sorted by magnetic cell sorting were intravenously injected into the middle cerebral arterial occlusion(MCAO) rat model. Neurological score showed that SSEA3(+)UCB transplantation group exhibited significant improvements in the functional outcome of MCAO rats than UCB transplantation group. Nissl staining and microtubule association protein-2(MAP2) immunofluorescence staining showed that the SSEA3(+)UCB transplantation group decreased neuronal loss. SSEA3(+)UCB transplantation group reduced neuronal apoptosis, inhibited caspase3 expression, and decreased tumor necrosis factor α(TNF-α). These results indicate that SSEA3 positive cells are a novel subpopulation of UCB cells, which exhibit great potential for the treatment of ischemic stroke.


Sujet(s)
Modèles animaux de maladie humaine , Sang foetal , Accident vasculaire cérébral ischémique , Animaux , Humains , Accident vasculaire cérébral ischémique/thérapie , Accident vasculaire cérébral ischémique/métabolisme , Sang foetal/cytologie , Agranulocytes/transplantation , Agranulocytes/métabolisme , Mâle , Rat Sprague-Dawley , Rats , Infarctus du territoire de l'artère cérébrale moyenne/thérapie , Transplantation de cellules souches de sang du cordon/méthodes , Antigènes embryonnaires spécifiques de stade/métabolisme , Encéphalopathie ischémique/thérapie , Apoptose/physiologie
2.
Cell Reprogram ; 23(6): 336-348, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34677101

RÉSUMÉ

Human mesenchymal stem cells (MSCs) can be isolated from various tissues. However, the cytokine profile in different MSC types remains unclear. In this study, MSCs were extracted from adipose, umbilical cord, and placental tissues. The surface marker expression, multilineage differentiation potential, and cytokine secretion of these cells were compared. The isolated MSCs exhibited similar morphology and surface marker expression. However, they differed with regard to their differentiation potential. Adipose-MSCs (A-MSCs) exhibited a higher potential for adipogenesis and osteogenic differentiation compared with umbilical cord-MSCs (UC-MSCs) and placental-MSCs (P-MSCs). The expression levels of 80 cytokines were detected, and the data demonstrated that the three MSC types abundantly secreted insulin-like growth factor-binding protein (IGFBP)-4, IGFBP-3, tissue inhibitor of metalloproteinase (TIMP)-1, TIMP-2, IGFBP-6, monocyte chemoattractant protein-1, and granulocyte colony-stimulating factor. However, the expression levels of vascular endothelial growth factor, tumor necrosis factor alpha, interleukin (IL)-6 receptor, and IL-13 in A-MSCs were higher compared with those of UC-MSCs and P-MSCs. Moreover, the expression levels of intercellular adhesion molecule-1 and growth differentiation factor 15 were lower in A-MSCs. Kyoto Encyclopedia of Genes and Genomes analysis indicated that the "adipocytokine" and the "PI3K/Akt pathways" were enriched in A-MSCs. Taken together, the results demonstrated that MSCs from different sources exhibited differences in the secretion of specific factors. A-MSCs were associated with the expression of several proangiogenic factors and may be an improved source for angiogenesis and tissue regeneration.


Sujet(s)
Cellules souches mésenchymateuses , Ostéogenèse , Différenciation cellulaire , Cellules cultivées , Cytokines , Femelle , Humains , Phosphatidylinositol 3-kinases , Placenta , Grossesse , Cordon ombilical , Facteur de croissance endothéliale vasculaire de type A
3.
Asia Pac J Clin Nutr ; 30(1): 60-66, 2021.
Article de Anglais | MEDLINE | ID: mdl-33787041

RÉSUMÉ

BACKGROUND AND OBJECTIVES: To investigate the efficacy of beclomethasone and aminophylline combined with enteral nutrition in the treatment of elderly patients with chronic obstructive pulmonary disease (COPD) and the associated effects of these drugs on patient nutritional status and immune function. METHODS AND STUDY DESIGN: In total, 115 elderly patients with COPD were included and were randomized into an enteral nutrition (EN) group and a control (CON) group. Aminophylline, in combination with beclomethasone, was administered to the CON group, whereas aminophylline and beclomethasone in combination with EN was administered to the EN group. RESULTS: Patients in the EN group showed significant improvement in partial pressure of carbon dioxide, forced expiratory volume in 1 sec/ expiratory forced vital capacity, and partial pressure of oxygen than those in the CON group. The levels of IgM, IgG, and IgA as well as the number of CD4+/CD8+ and CD4+/CD3+ T cells were higher in the EN group than those in the CON group (p<0.05); the EN group also exhibited higher levels of inflammatory cytokines, such as tumor necrosis factor-α and interleukin (IL)-1ß (p<0.05), and lower levels of IL-6 than the CON group. In addition, patients in the EN group showed a significant increase in serum total protein, albumin, and transferrin levels than those in the CON group (p<0.05). CONCLUSIONS: Elderly patients with COPD showed a marked response to a regimen of beclomethasone, aminophylline, and EN, which significantly improved their immune function and nutritional status.


Sujet(s)
Béclométasone , Broncho-pneumopathie chronique obstructive , Sujet âgé , Aminophylline , Nutrition entérale , Humains , Immunité , État nutritionnel , Broncho-pneumopathie chronique obstructive/traitement médicamenteux
4.
Aging (Albany NY) ; 13(3): 4370-4387, 2021 01 20.
Article de Anglais | MEDLINE | ID: mdl-33495412

RÉSUMÉ

Transplantation of mesenchymal stromal cells (MSCs) improves functional recovery in experimental models of spinal cord injury (SCI), but the mechanism is not fully understood. Activation of procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2), a collagen-modifying enzyme, reportedly follows MSC transplantation in an SCI animal model. We investigated the regulation of PLOD2 expression and its potential contribution to the neuroprotective effects of adipose-derived stromal cells (ADSCs) following mechanical injury to neurons in vitro and SCI in vivo. ADSCs enhanced wound healing in vitro and promoted functional recovery after their implantation near injury sites in a rat SCI model. These effects correlated with upregulation of PLOD2, MAP2, NSE and GAP43, and downregulation of GFAP, which is indicative of improved neuronal survival and axonal regeneration as well as reduced glial scar formation. The neurorestorative effect of ADSCs was weakened after inhibition of PLOD2 expression. ADSCs appeared to induce PLOD2 upregulation via TGF-ß1 secretion, as ADSC-mediated PLOD2 expression, neuronal survival, and functional recovery after SCI were largely prevented by SB431542, a TGF-(1 receptor inhibitor. These findings indicate that ADSCs reduce lesion size and promote functional recovery after SCI mainly through activation of a TGF-ß1/P-Samd3/PLOD2 pathway in spinal cord neurons.


Sujet(s)
Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Neurones/métabolisme , Procollagen-lysine, 2-oxoglutarate 5-dioxygenase/génétique , Récupération fonctionnelle , Protéine Smad-3/génétique , Traumatismes de la moelle épinière/génétique , Facteur de croissance transformant bêta-1/génétique , Adipocytes , Adipogenèse , Animaux , Techniques in vitro , Ostéocytes , Ostéogenèse , Cellules PC12 , Procollagen-lysine, 2-oxoglutarate 5-dioxygenase/métabolisme , Rats , Protéine Smad-3/métabolisme , Traumatismes de la moelle épinière/métabolisme , Traumatismes de la moelle épinière/thérapie , Facteur de croissance transformant bêta-1/métabolisme
5.
World J Pediatr ; 16(2): 193-200, 2020 Apr.
Article de Anglais | MEDLINE | ID: mdl-31535281

RÉSUMÉ

BACKGROUND: Neonatal hypoxia ischemia causes severe brain damage. Stem cell therapy is a promising method for treating neuronal diseases. Clinical translation of human umbilical cord-derived mesenchymal stem cells (UC-MSCs) for the recovery of neurons after hypoxic ischemic encephalopathy (HIE) may represent an effective therapy. METHODS: Primary neurons were exposed to oxygen-glucose deprivation (OGD) and subsequently cocultured with UC-MSCs. Apoptosis was examined by Annexin V-FITC-PI. Genes related to apoptosis were detected using RT-PCR and western-blot analyses. Using an in vivo model, HIE was induced in postnatal day 7 mice, and UC-MSCs were transplanted via the intraventricular route. UC-MSC migration was investigated by immunofluorescence, and lesion volumes were measured by TTC staining. Apoptosis in injured brain cells was detected by the TUNEL assay. RT-PCR and ELISA were used to detect the expression of inflammatory factors in cells and animal tissues. RESULTS: Flow cytometry analysis revealed that apoptosis in injured neurons was inhibited by UC-MSCs. The RT-PCR and western blot results indicated that coculture inhibited the expression of proapoptotic genes and upregulated expression of antiapoptotic genes. In the animal model, transplanted UC-MSCs migrated toward the cerebral lesion site and decreased the lesion extent in HIE. TUNEL staining showed that the MSC group exhibited significantly reduced numbers of TUNEL-positive cells. RT-PCR and ELISA showed that UC-MSCs inhibited the upregulation of TNF-α and IL-1ß in response to hypoxic ischemic injury. CONCLUSION: These results indicate that UC-MSCs exert neuroprotective effects against hypoxic ischemic injury by inhibiting apoptosis, and the mechanism appears to be through alleviating the inflammatory response.


Sujet(s)
Apoptose/physiologie , Hypoxie-ischémie du cerveau/étiologie , Cellules souches mésenchymateuses/physiologie , Neuroprotection , Animaux , Animaux nouveau-nés , Cellules cultivées , Femelle , Mâle , Souris , Rats , Rat Wistar , Cordon ombilical/cytologie
6.
Cancer Gene Ther ; 27(6): 473-485, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-31308482

RÉSUMÉ

DNA methylation has an important role in Helicobacter pylori (H. pylori)-induced gastric cancer (GC) processes and development. The aim of this study was to search genome-scale epigenetic modifications for studying pathogenesis of H. pylori-induced GC, and to find factors and powerful signature related to survival and prognosis. In this study, we conducted a comprehensive analysis of DNA methylation and gene expression profiles in the Gene Expression Omnibus (GEO), to identified differentially expressed genes (DEGs) and differentially methylated genes (DMGs). Functional enrichment analysis of the screened genes was performed, and a protein-protein interaction network was constructed. The TCGA DNA methylation databases and 55 H. pylori-infected GC cases of GEO RNA sequencing (GSE62254) were utilized for prognostic value validation of hub genes. Finally, a prognosis-related risk signature was identified by a series of bioinformatics analysis for H. pylori-induced GC patients. Totally, 161 DMGs were identified. Pathway analysis showed that all MDEGs mainly associated with Ras signaling pathway, renal cell carcinoma, mitogen-activated protein kinase signaling pathway. Five hub genes including CACNB2, GNB4, GRIN2A, MEF2C, and PREX1 were screened as independent prognostic factors in H. pylori-induced GC patients. Two-gene (CACNB2 and MEF2C) risk signature was constructed for predicting the overall survival of H. pylori-induced GC patients. Our study indicated possible MDEGs and pathways in H. pylori-induced GC by bioinformatics analysis, which may provide novel insights for unraveling pathogenesis of H. pylori-induced GC. Hub genes might serve as aberrantly methylation-based biomarkers for clinical diagnostic and prognostic evaluation of H. pylori-induced GC.


Sujet(s)
Méthylation de l'ADN , Infections à Helicobacter/génétique , Infections à Helicobacter/anatomopathologie , Helicobacter pylori/isolement et purification , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/microbiologie , Différenciation cellulaire/physiologie , Épigenèse génétique , Femelle , Analyse de profil d'expression de gènes , Infections à Helicobacter/microbiologie , Humains , Mâle , Pronostic , Transduction du signal , Tumeurs de l'estomac/anatomopathologie
7.
PLoS One ; 14(11): e0225174, 2019.
Article de Anglais | MEDLINE | ID: mdl-31721797

RÉSUMÉ

In this study, we analyzed the application of four autosomal kits and the sensitivity of the combined paternity index (CPI) cutoff value (CPI≥10000) in parentage testing. First, 1442 real trios and 803 real duos were tested using the Goldeneye 25A kit. The Goldeneye 25A kit covers the autosomal short tandem repeat (STR) loci of the other three kits, so we calculated the CPI value of every case for the four kits. Second, three complex close relative kinship cases were also analyzed to evaluate the application of the CPI cutoff value. The CPI values of all trio cases were higher than 10000 using the four kits; the CPI values of all duo cases were higher than 10000 using the Goldeneye 25A kit; and the CPI values of a portion of the duo cases were lower than 10000 using the other three kits. In the three complex close relative cases, the alleged father or mother was not excluded using 40 autosomal STRs. Adding X chromosome short tandem repeats (X-STR) and samples of biological fathers or mothers, the conclusions were confirmed. The four kits were adequate to draw conclusions in the trio cases; the Goldeneye 25A Kit was adequate to draw conclusions in the duo cases; and the other three kits were not sufficient for a portion of the duo cases. The CPI cutoff value was sensitive for real trio and duo cases. In complex close relative kinship cases, high CPI values may result in false conclusions.


Sujet(s)
Locus génétiques , Répétitions microsatellites , Paternité , Femelle , Génétique légale/méthodes , Humains , Mâle
8.
Mol Cell Biochem ; 450(1-2): 97-103, 2019 Jan.
Article de Anglais | MEDLINE | ID: mdl-29948506

RÉSUMÉ

Aryl hydrocarbon-receptor nuclear translocator (ARNT2) is a member of the bHLH PAS (basic helix-loop-helix Period/ARNT/Single-minded) family of transcription factors. Recently, some studies indicate that ARNT2 is associated with the occurrence and development of carcinoma. However, its roles in gastric cancer (GC) remain unclear. In the present study, we found that ARNT2 expression level is lower in GC tissues compared with adjacent non-tumor tissues, and negatively correlated with depth of invasion of the tumor, differentiated degree, and poor survival of GC patients. Overexpression of ARNT2 inhibits cell proliferation. Furthermore, AKT pathway contributed to ARNT2 -mediated PC proliferation. Taken together, our results provide the first evidence that high expression of ARNT2 inhibited proliferation of GC cells and affected tumor aggressiveness in GC patients.


Sujet(s)
Adénocarcinome/secondaire , Translocateur nucléaire du récepteur des hydrocarbures aromatiques/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Prolifération cellulaire , Tumeurs de l'estomac/anatomopathologie , Adénocarcinome/métabolisme , Femelle , Études de suivi , Humains , Métastase lymphatique , Mâle , Adulte d'âge moyen , Pronostic , Tumeurs de l'estomac/métabolisme , Cellules cancéreuses en culture
9.
J Cell Physiol ; 234(5): 7128-7140, 2019 05.
Article de Anglais | MEDLINE | ID: mdl-30370523

RÉSUMÉ

Gastric cancer (GC) is a lethal disease, and among its variety of etiological factors, Helicobacter pylori (H. pylori) infection is the strongest risk factor. However, the genetic and molecular mechanisms underlying H. pylori-related GC need further elucidation. We investigated the competing endogenous RNA (ceRNA) network differences between H. pylori (+) and H. pylori (-) GC. The long noncoding RNA (lncRNA), microRNA (miRNA), and messenger RNA (mRNA) expression data from 32 adjacent noncancerous samples and 18 H. pylori (+) and 141 H. pylori (-) stomach adenocarcinoma samples were downloaded from the TCGA database. After construction of lncRNA-miRNA-mRNA ceRNA networks of H. pylori (+) and H. pylori (-) GC, Panther and Kobas databases were used to analyze the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Finally, survival analysis was used to discover the key genes. In H. pylori (+) GC, we identified a total of 1,419 lncRNAs, 82 miRNAs, and 2,501 mRNAs with differentially expressed profiles. In H. pylori (-) GC, 2,225 lncRNAs, 130 miRNAs, and 3,146 mRNAs were differentially expressed. Furthermore, three unique pathways (cytokine-cytokine receptor interaction, HIF-1 signaling pathway, and Wnt signaling pathway) were enriched in H. pylori (+) GC. According to the overall survival analysis, three lncRNAs (AP002478.1, LINC00111, and LINC00313) and two mRNAs (MYB and COL1A1) functioned as prognostic biomarkers for patients with H. pylori (+) GC. In conclusion, our study has identified the differences in ceRNA regulatory networks between H. pylori (+) and H. pylori (-) GC and provides a rich candidate reservoir for future studies.


Sujet(s)
Adénocarcinome/génétique , Marqueurs biologiques tumoraux/génétique , Réseaux de régulation génique , Infections à Helicobacter/microbiologie , Helicobacter pylori/pathogénicité , ARN long non codant/génétique , ARN tumoral/génétique , Tumeurs de l'estomac/génétique , Adénocarcinome/microbiologie , Adénocarcinome/mortalité , Collagène de type I/génétique , Chaine alpha-1 du collagène de type I , Bases de données génétiques , Régulation de l'expression des gènes tumoraux , Infections à Helicobacter/complications , Interactions hôte-pathogène , Humains , Protéines proto-oncogènes c-myb/génétique , ARN messager/génétique , Tumeurs de l'estomac/microbiologie , Tumeurs de l'estomac/mortalité
10.
J Cell Biochem ; 120(5): 8447-8456, 2019 May.
Article de Anglais | MEDLINE | ID: mdl-30485509

RÉSUMÉ

Gastric cancer (GC) is one of the most common cancers. Resistance to 5-fluorouracil (5-Fu)-based chemotherapy is a major cause of treatment failure followed by the poor prognosis of patients. In GC, it was reported that human differentiated embryonic chondrocyte-expressed gene 2 (DEC2), suppressed tumor proliferation and metastasis, but the effect of DEC2 on chemosensitivity of GC cells was unknown. In our study, we found that DEC2 can obviously increase the sensibility of GC cells to 5-Fu by promoting 5-Fu-induced apoptosis. DEC2 overexpression is significantly associated with decreased phosphorylation of STAT5A (P-STAT5A). More importantly, negative correlations between DEC2 with P-STAT5A expression were observed in tissue sections from GC patients. GC patients with low expression levels of DEC2 and high expression levels of P-STAT5A showed a poor prognosis. Furthermore, enhanced chemosensitivity mediated by DEC2 can be reversed by STAT5A which confer GC cells resistance to apoptosis induced by 5-Fu. Together, our results suggest that through inhibiting activation of STAT5A, DEC2 enhances 5-Fu-induced apoptosis and suppression of proliferation in GC cells. These findings will provide new insight for identifying potential targets that can be used to sensitize GC cells to chemotherapy.

11.
Oncotarget ; 9(8): 8120-8132, 2018 Jan 30.
Article de Anglais | MEDLINE | ID: mdl-29487720

RÉSUMÉ

The value of targeting VEGFR (vascular endothelial growth factor receptor) drugs has demonstrated encouraging anti-cancer activity in advanced solid tumors within current clinical trials. This study aimed to serve as the first systemic review to assess their safety and efficacy according to biochemical characteristics of targeting VEGFR drugs in gastric cancer. We analyzed eight clinical trials on targeting VEGFR drugs in gastric cancer. Results showed that targeting VEGFR drugs significantly improved overall survival (OS) [Hazard Ratio (HR) 0.69, 95% confidence interval (CI) (0.55, 0.83), P < 0.001], progression free survival (PFS) [HR 0.50, 95% CI (0.34, 0.66), P < 0.001], disease control rate (DCR) [Odds Ratio (OR) 3.83, 95% CI (2.39, 6.15), P < 0.001] and significantly decreased the progressive disease rate(PDR)[OR 0.45, 95% CI (0.34, 0.59), P < 0.001], but not objective response rate (ORR) [OR 1.46, 95% CI (0.93, 2.29), P = 0.098]. Further subgroup revealed that VEGFR antibody (VEGFR-Ab) drugs were superior to VEGFR tyrosine kinase inhibitor (VEGFR-TKI) drugs in terms of the OS, PFS and PDR. To determine the toxic effect of targeting VEGFR drugs, the relative risk of adverse events (grade ≥ 3) of special interest(AESIs) were estimated. Most of these were predictable and manageable. Furthermore, less AESIs were observed in the VEGFR-Ab than the VEGFR-TKI drugs. In conclusion, VEGFR drugs were effective targeted therapy in advanced or metastatic gastric cancer, and its toxicity is within a controllable range. VEGFR-Ab drugs were more effective than VEGFR-TKI drugs in terms of the OS, PFS and PDR of gastric cancer patients with little toxicity.

12.
J Genet Genomics ; 44(7): 355-362, 2017 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-28750889

RÉSUMÉ

Cigarette smoking is the top environmental risk factor for lung cancer. Nicotine, the addictive component of cigarettes, induces lung cancer cell proliferation, invasion and migration via the activation of nicotinic acetylcholine receptors (nAChRs). Genome-wide association studies (GWAS) show that CHRNA5 gene encoding α5-nAChR is especially relevant to lung cancer. However, the mechanism of this subunit in lung cancer is not clear. In the present study, we demonstrate that the expression of α5-nAChR is correlated with phosphorylated STAT3 (pSTAT3) expression, smoking history and lower survival of non-small cell lung cancer (NSCLC) samples. Nicotine increased the levels of α5-nAChR mRNA and protein in NSCLC cell lines and activated the JAK2/STAT3 signaling cascade. Nicotine-induced activation of JAK2/STAT3 signaling was inhibited by the silencing of α5-nAChR. Characterization of the CHRNA5 promoter revealed four STAT3-response elements. ChIP assays confirmed that the CHRNA5 promoter contains STAT3 binding sites. By silencing STAT3 expression, nicotine-induced upregulation of α5-nAChR was suppressed. Downregulation of α5-nAChR and/or STAT3 expression inhibited nicotine-induced lung cancer cell proliferation. These results suggest that there is a feedback loop between α5-nAChR and STAT3 that contributes to the nicotine-induced tumor cell proliferation, which indicates that α5-nAChR is an important therapeutic target involved in tobacco-associated lung carcinogenesis.


Sujet(s)
Tumeurs du poumon/anatomopathologie , Nicotine/pharmacologie , Récepteurs nicotiniques/métabolisme , Facteur de transcription STAT-3/métabolisme , Carcinogenèse/effets des médicaments et des substances chimiques , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mâle , Adulte d'âge moyen , Régions promotrices (génétique)/génétique , Récepteurs nicotiniques/génétique , Transduction du signal/effets des médicaments et des substances chimiques
13.
PLoS One ; 11(2): e0149120, 2016.
Article de Anglais | MEDLINE | ID: mdl-26909550

RÉSUMÉ

Gastric cancer incidence demonstrates a strong etiologic association with smoking. Nicotine, the major component in tobacco, is a survival agonist that inhibits apoptosis induced by certain chemotherapeutic agents, but the precise mechanisms involved remain largely unknown. Recently studies have indicated that α5-nicotinic acetylcholine receptor (α5-nAChR) is highly associated with lung cancer risk and nicotine dependence. Nevertheless, no information has been available about whether nicotine also affects proliferation of human gastric cancer cells through regulation of α5-nAChR. To evaluate the hypothesis that α5-nAChR may play a role in gastric cancer, we investigated its expression in gastric cancer tissues and cell lines. The expression of α5-nAChR increased in gastric cancer tissue compared with para-carcinoma tissues. In view of the results, we proceeded to investigate whether nicotine inhibits cisplatin-induced apoptosis via regulating α5-nAChR in gastric cancer cell. The results showed that nicotine significantly promoted cell proliferation in a dose and time-dependent manner through α5-nAChR activation in human gastric cells. Furthermore, nicotine inhibited apoptosis induced by cisplatin. Silence of α5-nAChR ablated the protective effects of nicotine. However, when co-administrating LY294002, an inhibitor of PI3K/AKT pathway, an increased apoptosis was observed. This effect correlated with the induction of Bcl-2, Bax, Survivin and Caspase-3 by nicotine in gastric cell lines. These results suggest that exposure to nicotine might negatively impact the apoptotic potential of chemotherapeutic drugs and that α5-nAChR/AKT signaling plays a key role in the anti-apoptotic activity of nicotine induced by cisplatin.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Cisplatine , Nicotine , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs nicotiniques/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs de l'estomac , Lignée cellulaire tumorale , Cisplatine/antagonistes et inhibiteurs , Cisplatine/pharmacologie , Femelle , Humains , Mâle , Nicotine/antagonistes et inhibiteurs , Nicotine/pharmacologie , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie
14.
Oncol Rep ; 34(1): 227-34, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-25997700

RÉSUMÉ

Signal transducer and activator of transcription (STAT) 5, including STAT 5a and STAT 5b, was reported to play important roles in the malignant biological behaviors of tumors. However, their roles in gastric cancer, especially for STAT 5b remain unknown. This study aimed to detect the expression of STAT 5b in gastric cancer cells and analyze the role and possible mechanism of STAT 5b in the chemo-sensitivity of gastric cancer cells to gefitinib. A total of 69 patients with gastric carcinomas were analyzed for the expression of STAT 5b in carcinomas and para-carcinomas by immunohistochemistry. Cultured MGC-803 and MKN-45 cells were exposed to gefitinib and/or STAT 5b siRNA. Mitochondrial proteins including Bcl-2, Bax, caspase-3 and caspase-9 were extracted using special kits for detecting mitochondrial pathway-related apoptosis proteins. The results showed that STAT 5b expression was significantly increased in gastric carcinomas compared with para-carcinomas, with a positive rate of 49/69 in carcinomas and 27/69 in para-carcinomas (P=0.001). Gefitinib exposure reduced the relative viabilities of MGC-803 and MKN-45 cells in a concentration- and time-dependent manner, and cell apoptosis increased significantly (P<0.05) with gefitinib treatment (4 mM, 24 h). STAT 5b expression was significantly downregulated by treatment with gefitinib (4 mM, 24 h). Interference of STAT 5b expression by siRNA targeting enhanced the chemo-sensitivity of gastric cancer cells to gefitinib by promoting mitochondrial pathway-mediated apoptosis. Bax, caspase-3 and caspase-9 expression were upregulated, and Bcl-2 expression was downregulated in the combined treatment group (gefitinib+siRNA) compared with the gefitinib (4 mM, 24 h) only group in the MGC-803 and MKN-45 cells (P<0.05). Overall, STAT 5b was upregulated in gastric carcinomas compared with para-carcinomas. Interference of STAT 5b expression by siRNA targeting enhanced the chemo-sensitivity of gastric cancer cells to gefitinib by promoting mitochondrial pathway-mediated cell apoptosis. These findings may be useful for developing new approaches for the treatment of gastric cancer.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Quinazolines/administration et posologie , Facteur de transcription STAT-5/biosynthèse , Tumeurs de l'estomac/traitement médicamenteux , Sujet âgé , Sujet âgé de 80 ans ou plus , Caspase-3/génétique , Caspase-9/génétique , Femelle , Géfitinib , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mâle , Adulte d'âge moyen , Mitochondries/métabolisme , Protéines proto-oncogènes c-bcl-2/génétique , Facteur de transcription STAT-5/génétique , Transduction du signal , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Protéine Bax/génétique
15.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 22(5): 1408-14, 2014 Oct.
Article de Chinois | MEDLINE | ID: mdl-25338598

RÉSUMÉ

This study was aimed to investigate the change of cell phenotype and the expression of hematopoiesis associated cytokines in umbilical cord mesenchymal stem cells (UC-MSC) in three-dimensional (3-D) system. MSC were isolated from umbilical cord, and then cultured in 2-D and 3-D system respectively. The phenotype of MSC was detected by flow cytometry; the angiogenic capability of MSC cultured in 2-D and 3-D syitem was assessed using in vitro capillary formation assay. The cytokine expression of MSC in two kinds of culture conditions was measured by real-time PCR. The results showed that MSC were successfully isolated from umbilical cord. Flow cytometry showed that the percentage of CD31, CD133 and CD271 expressed in endothelial cells, endothelial progenitor cells and primitive mesenchymal stem cells increased significantly in 3-D culture conditions, as compared to 2-D system. Capillary formation assay showed that the angiogenic capability of UC-MSC was greatly enhanced. Quantitative PCR showed that the expression of ß-actin was upregulated in 3-D system. The expression of some cytokines associated with hematopoiesis, such as G-CSF, LIF, SCF, IL-1α, IL-1ß, IL-3, IL-7 and IL-11, increased, especially for LIF, IL-3, IL-7. The expression of IL-10 associated with immune regulation also increased. The expression of SDF-1, IL-6 slightly decreased, but without significant difference. It is concluded that expression of CD31, CD133 and CD271 increases in 3-D system, the angiogenic capability of UC-MSC enhances and the expression of hematopoiesis-associated cytokines in UC-MSC increases in 3-D system.


Sujet(s)
Cytokines/biosynthèse , Cellules souches mésenchymateuses/métabolisme , Cordon ombilical/métabolisme , Actines , Cellules cultivées , Cytométrie en flux , Hématopoïèse , Humains , Phénotype , Réaction de polymérisation en chaine en temps réel
16.
Zhonghua Xue Ye Xue Za Zhi ; 35(10): 891-6, 2014 Oct.
Article de Chinois | MEDLINE | ID: mdl-25339324

RÉSUMÉ

OBJECTIVE: To investigate the potential immunomodulatory properties of fetal bone marrow derived mesenchymal stem cells (FBM- MSCs). METHODS: Mononuclear cells from the bone marrow of second trimester (14-22 wks) fetus were isolated and cultured for the derivation of MSCs. The derived FBM-MSC cells were characterized via morphology, immunophenotyping and the adipogenic and osteogenic differentiation assays. The immunomodulatory properties of FBM-MSC on lymphocytes were evaluated through the co- culture assay with PHA activated adult peripheral blood mononuclear cells (PBMCs). RESULTS: Derived FBM-MSCs were CD29⁺, CD44⁺, CD49e⁺, CD73⁺, CD90⁺, CD105⁺ and CD31⁻ , CD34⁻ , CD45⁻ , HLA-DR⁻ and can be differentiated into adipocytes and osteocytes. When co-cultured with PHA-activated PBMCs, FBM-MSCs inhibited the proliferation of lymphocytes up to 96% and down-regulated the secretion of inflammatory cytokines such as IFN-γ and TNF-α up to 90.9% and 58.4% respectively. When compared with FBM-MSCs cultured alone, the expression of MSCs derived immunomodulatory cytokines, such as IDO, TSG-6 and TGF-ß, was up-regulated significantly in the co-culture system. CONCLUSION: MSC derived from fetal bone marrow demonstrated immunosuppressive effects on adult PBMCs in vitro. MSC-derived cytokines like IDO, TSG-6 and TGF-ß may be critical for FBM-MSCs mediated immunosuppressive function.


Sujet(s)
Cellules de la moelle osseuse/immunologie , Prolifération cellulaire , Tolérance immunitaire , Cellules souches mésenchymateuses/immunologie , Adulte , Moelle osseuse , Cellules de la moelle osseuse/cytologie , Différenciation cellulaire , Cellules cultivées , Techniques de coculture , Cytokines , Cellules souches hématopoïétiques , Humains , Immunophénotypage , Techniques in vitro , Agranulocytes , Lymphocytes , Cellules souches mésenchymateuses/cytologie , Ostéogenèse
17.
Toxicol Appl Pharmacol ; 278(2): 172-9, 2014 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-24793809

RÉSUMÉ

By binding to nicotinic acetylcholine receptors (nAChRs), nicotine induces the proliferation and apoptosis of non-small cell lung cancer (NSCLC). Previous studies have indicated that α5-nAChR is highly associated with lung cancer risk and nicotine dependence. However, the mechanisms through which α5-nAChRs may influence lung carcinogenesis are far from clear. In the present study, we investigated the roles of α5-nAChR in the nicotine-induced expression of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF). Immunohistochemistry was used to detect the expression of α5-nAChR and HIF-1α in 60 specimens of lung cancer and para-carcinoma tissue. The correlations between the expression levels of α5-nAChR and HIF-1α and other clinicopathological data were analyzed. In a cell line that highly expressed α5-nAChR, the loss of α5-nAChR function by siRNA was used to study whether α5-nAChR is involved in the nicotine-induced expression of HIF-1α and VEGF through the activation of the ERK1/2 and PI3K/Akt signaling pathways. Cell growth was detected using the cell counting kit-8 (CCK-8). α5-nAChR (78.3%) and HIF-1α (88.3%) were both overexpressed in NSCLC, and their expression levels were found to be correlated with each other (P<0.05). In the A549 cell line, α5-nAChR and HIF-1α were found to be expressed under normal conditions, and their expression levels were significantly increased in response to nicotine treatment. The silencing of α5-nAChR significantly inhibited the nicotine-induced cell proliferation compared with the control group and attenuated the nicotine-induced upregulation of HIF-1α and VEGF, and these effects required the cooperation of the ERK1/2 and PI3K/Akt signaling pathways. These results show that the α5-nAChR/HIF-1α/VEGF axis is involved in nicotine-induced tumor cell proliferation, which suggests that α5-nAChR may serve as a potential anticancer target in nicotine-associated lung cancer.


Sujet(s)
Carcinome pulmonaire non à petites cellules/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/biosynthèse , Tumeurs du poumon/métabolisme , Nicotine/toxicité , Récepteurs nicotiniques/physiologie , Facteur de croissance endothéliale vasculaire de type A/biosynthèse , Adulte , Sujet âgé , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Régulation négative/physiologie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Tumeurs du poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Récepteurs nicotiniques/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique
18.
Int Orthop ; 37(7): 1399-404, 2013 Jul.
Article de Anglais | MEDLINE | ID: mdl-23645083

RÉSUMÉ

PURPOSE: Hedgehog signalling plays an important role during the development of tissues and organs, including bone and limb. Dexamethasone (DEX), a synthetic and widely used glucocorticoid, affects osteogenesis of bone marrow mesenchymal stem cells (MSCs), while the signalling pathway by which DEX affects osteoblast differentiation remains obscure. This study aimed to investigate expressions of hedgehog signalling molecules Shh, Ihh and Gli1 during DEX-induced osteogenesis of rat MSCs in vitro. METHODS: DEX promoted osteoblast differentiation of MSCs at 10(-8) mol/L from seven days to 21 days, demonstrated by enhancing alkaline phosphatase (ALP) activity and osteoblast-associated marker type I collagen expression during osteoblastic differentiation. Gene and protein expressions of hedgehog signalling molecules, Shh, Ihh and Gli1 were tested by RT-PCR and western blot analysis during osteoblast differentiation. RESULTS: Shh expression was increased compared to the control while Ihh and Gli1 expressions were decreased on both mRNA and protein level during DEX-induced osteoblast differentiation of MSCs from seven days to 21 days. Altogether, these data demonstrate that DEX can enhance Shh expression via a Gli1-independent mechanism during osteoblast differentiation of MSCs. CONCLUSIONS: These results indicate that different patterns of hedgehog signalling are involved in DEX-induced osteogenesis and these findings provide insights into the mechanistic link between glucocorticoid-induced osteogenesis and hedgehog signalling pathway.


Sujet(s)
Dexaméthasone/pharmacologie , Glucocorticoïdes/pharmacologie , Protéines Hedgehog/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Ostéogenèse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Phosphatase alcaline/métabolisme , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/physiologie , Cellules cultivées , Techniques in vitro , Facteurs de transcription Krüppel-like/métabolisme , Mâle , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Ostéoblastes/cytologie , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Ostéogenèse/physiologie , Rats , Rat Wistar , Transduction du signal/physiologie , Protéine à doigt de zinc GLI1
19.
Diagn Pathol ; 8: 37, 2013 Feb 27.
Article de Anglais | MEDLINE | ID: mdl-23445622

RÉSUMÉ

BACKGROUND: Gastric cancer is a leading causes of cancer-related deaths ,but the underlying molecular mechanisms of its progression are largely unknown. Differentiated embryonic chondrocyte-expressed gene 1 (DEC1), is an important transcription factor involved in the progression of tumors and has recently been identified to be strongly inducible by hypoxia. Little is known about the contribution of DEC1 to the intracellular hypoxia and proliferation signaling events in gastric cancer. METHODS: Immunohistochemistry was used to detect the expression of DEC1, hypoxia-inducible factor 1(HIF-1α) and Ki67 in 173 human gastric cancer samples and adjacent non-tumor tissues samples. The relationship between DEC1, HIF-1α and Ki67 was evaluated. RESULTS: DEC1 protein was persistently expressed in the nucleus and cytoplasm of gastric cancer tissue. The protein expression of DEC1 and HIF-1α in tumour tissues was 83.8% and 54.3%, respectively, and was significantly higher than that in adjacent normal tissues (83.8% vs 23.7%, P < 0.001; 54.3% vs 12.7%, P < 0.001). The expression of DEC1 and HIF-1α was associated with poor histological differentiation. (P < 0. 01). Furthermore, DEC1 level was positively correlated with HIF-1α (P < 0. 01, r=0.290) and Ki67 expression (P < 0. 01, r=0.249). CONCLUSION: The upregulation of DEC1 may play an important role in hypoxia regulation and cell proliferation in gastric cancer. The relevant molecular mechanism requires further investigation.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/analyse , Protéines à homéodomaine/analyse , Sous-unité alpha du facteur-1 induit par l'hypoxie/analyse , Antigène KI-67/analyse , Tumeurs de l'estomac/composition chimique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Différenciation cellulaire , Hypoxie cellulaire , Noyau de la cellule/composition chimique , Prolifération cellulaire , Cytoplasme/composition chimique , Femelle , Humains , Immunohistochimie , Mâle , Adulte d'âge moyen , Tumeurs de l'estomac/anatomopathologie
20.
PLoS One ; 8(1): e54774, 2013.
Article de Anglais | MEDLINE | ID: mdl-23382965

RÉSUMÉ

Alpha-fetoprotein (AFP)-producing gastric cancer (AFPGC), represented by the production of AFP, has a more aggressive behavior than common gastric cancer. The underlying mechanisms are not well understood. Arsenic trioxide (As(2)O(3)) is used clinically to treat acute promyelocytic leukemia(APL) and has activity in vitro against several solid tumor cell lines, with induction of apoptosis and inhibition of proliferation the prime effects. Signal transducer and activator of transcription 3 (STAT3) has an important role in tumorigenesis of various primary cancers and cancer cell by upregulating cell-survival and downregulating tumor suppressor proteins. Here, we found decreased expression of AFP and STAT3 after induction of apoptosis by As(2)O(3) in the AFPGC FU97 cells. Also, the level of the STAT3 target oncogene Bcl-2 was decreased with As(2)O(3), and that of the tumor suppressor Bax was increased. Furthermore, STAT3 expression and depth of invasion and lymph node metastasis were associated. Survival of patients with gastric cancer was lower with AFP and STAT3 double overexpression than with overexpression of either alone. Downregulation of AFP and STAT3 expression plays an important role in As(2)O(3)-induced apoptosis of AFPGC cells, which suggests a new mechanism of As(2)O(3)-induced cell apoptosis. As(2)O(3) may be a possible agent for AFPGC treatment.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Composés de l'arsenic/pharmacologie , Oxydes/pharmacologie , Facteur de transcription STAT-3/métabolisme , Tumeurs de l'estomac/métabolisme , Alphafoetoprotéines/métabolisme , Adulte , Sujet âgé , Trioxyde d'arsenic , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Milieux de culture conditionnés/métabolisme , Femelle , Expression des gènes , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mâle , Adulte d'âge moyen , Stadification tumorale , Pronostic , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/mortalité , Tumeurs de l'estomac/anatomopathologie , Alphafoetoprotéines/génétique , Protéine Bax/génétique , Protéine Bax/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE