Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Innate Immun ; 25(4): 217-223, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30943822

RESUMO

The aim of this study was to evaluate the effect of the miR-301a/PTEN pathway in cervical cancer. miR-301a and PTEN expression were measured by quantitative real-time PCR (qRT-PCR) in tissues samples and HeLa cells. PTEN protein level was determined by Western blotting. Dual reporter luciferase assay was performed to validate PTEN as a direct target of miR-301a. The gain- and loss-of function assay was performed by miR-301a overexpression and silencing. Cell proliferation was monitored by cell counting Kit-8 (CCK-8). Cell apoptosis was quantitated by flow cytometry. SPSS was used to analyze the significant difference in the treatments. miR-301a demonstrated a significantly higher expression in cervical carcinoma tissues compared with the paired non-carcinoma tissues ( n = 12), while PTEN expression was found to be significantly lower in cervical carcinoma tissues than their paired non-carcinoma tissues ( n = 12). In addition, PTEN was identified as the direct target of miR-301a. Moreover, overexpression of miR-301a significantly promoted HeLa cells proliferation and anti-apoptosis which had a reverse pattern after PTEN overexpression. Our results confirm PTEN as a direct target of miR-301a in HeLa cells and suggest that miR-301a/PTEN pathway contributes to the development and progression of cervical cancer.


Assuntos
MicroRNAs/genética , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias do Colo do Útero/genética , Apoptose , Carcinogênese , Proliferação de Células , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/genética , RNA Interferente Pequeno/genética , Transdução de Sinais
2.
Exp Gerontol ; 120: 88-94, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30876950

RESUMO

Mouse models have been widely used in the research of human diseases. Aging, just as cancer, is influenced by the interaction of various genetic and environmental factors. Currently, aging could be induced by many mechanism, including telomere dysfunction, oxidase stress, DNA damage and epigenetic changes. Many of these genetic pathways are also shared by aging and cancer. The mouse models generated to study these pathways might manifest either aging or cancer phenotypes, sometimes both, which in deed has worked as a good model system in understanding the correlation between aging and cancer. Here, we reviewed these mouse models that were generated to model aging or cancer. These mouse models might help us put those related pathways in context and discover essential interactions in cancer and aging regulation.


Assuntos
Envelhecimento , Neoplasias/etiologia , Telômero/fisiologia , Animais , Dano ao DNA , Modelos Animais de Doenças , Inflamação/complicações , Camundongos , Progéria/etiologia , Telomerase/genética , Proteína 2 de Ligação a Repetições Teloméricas/fisiologia
3.
Am J Transl Res ; 10(1): 212-223, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29423006

RESUMO

To investigate the clinical efficacy and safety of umbilical cord mesenchymal stem cell (UCMSC) transplantation for treating multiple sclerosis (MS), the patients with MS were recruited and treated with UCMSC. The procedure of preparing UCMSC was in accordance with the standards formulated by the International Society for Cell Biology. Cell surface markers, multiple differentiation potential and safety of UCMSC for transplantation were detected. The number of cells in each infusion was 1 to 2×106 cells/kg. Patients were recruited in accordance with the standards of the International Mesenchymal Stem Cells Transplantation Study Group. After treatment, the clinical therapeutic effects including symptoms, vital signs, clinical attacks, magnetic resonance imaging (MRI), neurological function scores and adverse reactions such as fever, dizziness, and vascular irritation were monitored and evaluated. In addition, the regulatory effects of UCMSC on immune system of patients were also assessed. The results showed that the patients' symptoms were improved after UCMSC transplantation. No clinical attacks occurred during transplantation. MRI revealed a reduced number of foci and Expanded Disability Status Scale scores were decreased. Some of patients had adverse reactions after transplantation. These adverse effects were not serious and lasted short duration, thus no intervention was conducted and let it be eliminated by itself. The mRNA expression of CD86, IL-2, CTLA-4, and HLADRB1 in peripheral blood was significantly decreased after UCMSC transplantation (P < 0.05). Based on our present studies, UCMSCs would be considered as a safe and alternative option for treatment of MS.

4.
Mol Carcinog ; 57(2): 147-158, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28949402

RESUMO

The mutation p53N236S (p53S) has been identified as one of the recurrent mutations in human cancers by TCGA database. Our in vitro data revealed the oncogenic gain of function of p53S. To understand the function of p53S in vivo, we generated the p53S knock-in mouse. The p53S/S mice manifested highly invasive lymphomas and metastatic sarcomas with dramatically increased double minute chromosomes. The survival curve, the incidence of tumors and the tumor spectrum of p53S/S mice is very similar to the p53R172H mouse model. The p53S/+ mice showed delayed onset of tumorigenesis and a high metastasis rate (40%) and low loss of heterozygosity rate (2/16). The activation of CDKN2A pathway in p53S/S MEF and tumors, and the accumulation of p19ARF protein in tumor tissues suggested p19ARF might contribute to the accumulation of mutant p53S protein in the tumor and promote tumorigenesis. The high expression of p19ARF correlated with mutant p53 accumulation and tumor progression, suggesting a dual role of p19ARF in tumor promotion or suppression that might depend on the p53 mutation status in tumor cells. The oncogenic gain of function of this recurrent mutation p53S prompts the reconsideration of p53 mutations function that occurs at a low frequency.


Assuntos
Carcinogênese/genética , Cromossomos/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Mutação/genética , Oncogenes/genética , Proteína Supressora de Tumor p53/genética , Animais , Modelos Animais de Doenças , Progressão da Doença , Linfoma/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sarcoma/genética
5.
PLoS One ; 12(10): e0186904, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29073178

RESUMO

New drug leads for the treatment of inflammation are urgently needed. Marine molluscs are widely used as traditional medicines for the treatment of inflammation. Here we report the positive effects of a hypobranchial gland (HBG) extract and the dominant bioactive compound 6-bromoisatin from the Muricidae mollusc Dicathais orbita, for reducing lipopolysaccharide (LPS) induced acute lung inflammation in a mouse model. Both 6-bromoisatin and the HBG extract suppressed the inflammatory response in mice that were pre-treated by oral gavage at 48, 24 and 1 h prior to LPS infusion. The inflammatory antagonists were tested at concentrations of 0.5 mg/g and 0.1 mg/g HBG extract and 0.1 mg/g and 0.05 mg/g 6-bromoisatin in carrier oil and all treatments reduced inflammation as indicated by a significant suppression of inflammatory markers present in bronchoalveolar lavage fluid (BALF), in comparison to LPS induced positive control mice administered the carrier oil alone (p < 0.0001). Tumour necrosis factor-alpha (TNFα) and interleukin-1 beta (IL-1ß) levels, in addition to total protein concentration were all significantly reduced in BALF from mice treated with the extract or 6-bromoisatin. Furthermore, all treatment groups showed significant reductions in neutrophil sequestration and preservation of the lung tissue architecture compared to the positive control (p < 0.0001). The combined results from this study and our previous in vitro studies indicate that 6-bromoisatin in the HGB extracts inhibit the activation of inflammatory signalling pathway. The results from this study further confirm that the HBG extract from Muricidae molluscs and 6-bromoisatin are bioavailable and effective in vivo, thus have potential for development as natural therapeutic agents for inflammation.


Assuntos
Lesão Pulmonar Aguda/fisiopatologia , Bromo/química , Modelos Animais de Doenças , Indóis/uso terapêutico , Inflamação/tratamento farmacológico , Biologia Marinha , Moluscos/química , Animais , Cromatografia Líquida , Feminino , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL
6.
Eur Heart J ; 38(19): 1498-1508, 2017 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-28329361

RESUMO

AIMS: Arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D) is characterized by fibrofatty infiltration of the myocardium and ventricular arrhythmias that may lead to sudden cardiac death. It has been observed that male patients develop the disease earlier and present with more severe phenotypes as compared to females. Thus, we hypothesized that serum levels of sex hormones may contribute to major arrhythmic cardiovascular events (MACE) in patients with ARVC/D. METHODS AND RESULTS: The serum levels of five sex hormones, sex hormone-binding globulin, high sensitivity troponin T, pro-brain natriuretic peptide, cholesterol, triglycerides, insulin, and glucose were measured in 54 ARVC/D patients (72% male). Twenty-six patients (48%) experienced MACE. Total and free testosterone levels were significantly increased in males with MACE as compared to males with a favourable outcome, whereas estradiol was significantly lower in females with MACE as compared to females with a favourable outcome. Increased testosterone levels remained independently associated with MACE in males after adjusting for age, body mass index, Task Force criteria, ventricular function, and desmosomal mutation status. Furthermore, an induced pluripotent stem cell-derived ARVC/D cardiomyocyte model was used to investigate the effects of sex hormones. In this model, testosterone worsened and estradiol improved ARVC/D-related pathologies such as cardiomyocyte apoptosis and lipogenesis, strongly supporting our clinical findings. CONCLUSIONS: Elevated serum testosterone levels in males and decreased estradiol levels in females are independently associated with MACE in ARVC/D, and directly influence disease pathology. Therefore, determining the levels of sex hormones may be useful for risk stratification and may open a new window for preventive interventions.


Assuntos
Displasia Arritmogênica Ventricular Direita/etiologia , Hormônios Esteroides Gonadais/metabolismo , Caracteres Sexuais , Adulto , Análise de Variância , Displasia Arritmogênica Ventricular Direita/sangue , Biomarcadores/metabolismo , Morte Súbita Cardíaca/etiologia , Desmossomos/genética , Estradiol/metabolismo , Exercício Físico/fisiologia , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Miócitos Cardíacos/fisiologia , Prognóstico , Testosterona/metabolismo
7.
Tumour Biol ; 37(8): 10317-27, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26842926

RESUMO

Unlike heterogeneous tumor cells, cancer-associated fibroblasts (CAF) are genetically more stable which serve as a reliable target for tumor immunotherapy. Fibroblast activation protein (FAP) which is restrictively expressed in tumor cells and CAF in vivo and plays a prominent role in tumor initiation, progression, and metastasis can function as a tumor rejection antigen. In the current study, we have constructed artificial FAP(+) stromal cells which mimicked the FAP(+) CAF in vivo. We immunized a breast cancer mouse model with FAP(+) stromal cells to perform immunotherapy against FAP(+) cells in the tumor microenvironment. By forced expression of FAP, we have obtained FAP(+) stromal cells whose phenotype was CD11b(+)/CD34(+)/Sca-1(+)/FSP-1(+)/MHC class I(+). Interestingly, proliferation capacity of the fibroblasts was significantly enhanced by FAP. In the breast cancer-bearing mouse model, vaccination with FAP(+) stromal cells has significantly inhibited the growth of allograft tumor and reduced lung metastasis indeed. Depletion of T cell assays has suggested that both CD4(+) and CD8(+) T cells were involved in the tumor cytotoxic immune response. Furthermore, tumor tissue from FAP-immunized mice revealed that targeting FAP(+) CAF has induced apoptosis and decreased collagen type I and CD31 expression in the tumor microenvironment. These results implicated that immunization with FAP(+) stromal cells led to the disruption of the tumor microenvironment. Our study may provide a novel strategy for immunotherapy of a broad range of cancer.


Assuntos
Neoplasias da Mama/patologia , Fibroblastos/imunologia , Gelatinases/imunologia , Imunoterapia/métodos , Proteínas de Membrana/imunologia , Serina Endopeptidases/imunologia , Microambiente Tumoral/imunologia , Animais , Western Blotting , Neoplasias da Mama/imunologia , Modelos Animais de Doenças , Endopeptidases , Feminino , Imunofluorescência , Imunização , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
8.
Eur J Obstet Gynecol Reprod Biol ; 195: 210-213, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26595307

RESUMO

OBJECTIVE: A number of studies have evaluated the association of p53 expression with the cervical cancer clinical outcome, but the results were inconsistent. STUDY DESIGN: Electronic search of PubMed was conducted to select studies. Studies which quantitatively evaluated the relationship between p53 expression and survival of the patients with cervical cancer were chosen, and hazard ratio (HR) with 95% confidence interval (CI) was used to assess the association. RESULTS: The final meta-analysis contained 11 published studies including 1105 patients. Combined HRs suggested that p53 overexpression has an unfavorable impact on overall survival(OS) [HR=1.672, 95% confidence interval (CI): 1.144-2.444, P=0.008], and Caucasian cervical cancer patients OS[HR=1.942, 95% CI: 1.162-3.245, P=0.011]. CONCLUSIONS: p53 expression indicates a poor prognosis for Caucasian people with cervical cancer.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/metabolismo , População Branca , Carcinoma de Células Escamosas/mortalidade , Feminino , Humanos , Prognóstico , Modelos de Riscos Proporcionais , Taxa de Sobrevida , Neoplasias do Colo do Útero/mortalidade
9.
Circ J ; 79(7): 1402-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25971409

RESUMO

Cellular reprogramming of somatic cells to patient-specific induced pluripotent stem cells (iPSCs) enables in-vitro modeling of human cardiac disorders for pathogenic and therapeutic investigations. However, using iPSC-derived cardiomyocytes (iPSC-CMs) to model an adult-onset heart disease remains challenging because of the uncertainty regarding the ability of relatively immature iPSC-CMs to fully recapitulate adult disease phenotypes. Arrhythmogenic right ventricular dysplasia (ARVD) is an inherited cardiomyopathy characterized by pathological fibrofatty infiltration and cardiomyocyte (CM) loss predominantly in the right ventricle (RV), leading to heart failure and lethal arrhythmias. Over 50% of affected individuals have desmosome gene mutations, most commonly inPKP2encoding plakophilin-2. Using Yamanaka's pluripotent factors, we generated iPSC lines from ARVD patients withPKP2mutations. We first developed a method to induce metabolic maturation of iPSC-CMs and showed that induction of adult-like metabolic energetics from an embryonic/glycolytic state is essential to model an adult-onset cardiac disease using patient-specific iPSCs. Furthermore, we showed that coactivation of normal peroxisome proliferator-activated receptor (PPAR)-α and abnormal PPARγ pathways in ARVD iPSC-CMs resulted in exaggerated CM lipogenesis, CM apoptosis, Na(+)channel downregulation and defective intracellular calcium handling, recapitulating the pathological signatures of ARVD. Using this model, we revealed novel pathogenic insights that metabolic derangement in an adult-like metabolic milieu underlies ARVD pathologies, enabling us to propose novel disease-modifying therapeutic strategies.


Assuntos
Displasia Arritmogênica Ventricular Direita/metabolismo , Displasia Arritmogênica Ventricular Direita/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Modelos Cardiovasculares , Adulto , Displasia Arritmogênica Ventricular Direita/genética , Diferenciação Celular , Células Cultivadas , Reprogramação Celular , Desmossomos/genética , Humanos , Mutação , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , PPAR alfa/metabolismo , PPAR gama/metabolismo , Placofilinas/genética
10.
Sci Rep ; 5: 9571, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25826780

RESUMO

Studies have proven that IL-2 and IL-15 showed contrasting roles during CIK cells preparation. By employing microarray, we analyzed miRNA expression profiles of PBMC, CIKIL-2 and CIKIL-15. Advanced bioinformatic analyses were performed to explore the key miRNAs which may regulate cell proliferation and anti-tumor activity of CIK. We identified 261 differentially expressed miRNAs (DEMs) between PBMC and CIKIL-2, and 249 DEMs between PBMC and CIKIL-15. MiR-143-3p/miR-145-5p was miRNA cluster which may positively regulate cell proliferation. In contrast, miR-340-5p/miR-340-3p cluster may negatively regulate cell proliferation via induction apoptosis, which may cause decreased cell proliferation capacity of CIKIL-2. MiRNA-target interaction analysis indicated that 10 co-downregulated miRNAs may synergistically turn on the expression of a pool of tumor cytotoxic genes in CIK cells. The DEMs between CIKIL-2 and CIKIL-15 may contribute to enhanced tumor cytotoxic capacity of CIKIL-2. Importantly, we found that repressed miR-193a-5p may regulate the expressions of inhibitory receptor KLRD1. The results of the validation assay have shown that KLRD1 were upregulated in CIK cells. Our findings have provided new insights into mechanisms of CIK cells production and tumor cytotoxic function, and shed light on their safety for clinical trial.


Assuntos
Complexo CD3/metabolismo , Antígeno CD56/metabolismo , Células Matadoras Induzidas por Citocinas/metabolismo , Perfilação da Expressão Gênica , MicroRNAs/genética , Transcriptoma , Mapeamento Cromossômico , Análise por Conglomerados , Células Matadoras Induzidas por Citocinas/imunologia , Citotoxicidade Imunológica/genética , Regulação da Expressão Gênica , Ontologia Genética , Redes Reguladoras de Genes , Humanos , Imunofenotipagem , Leucócitos Mononucleares/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Interferência de RNA , RNA Mensageiro/genética , Reprodutibilidade dos Testes , Transdução de Sinais
11.
BMC Med Genomics ; 7: 49, 2014 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-25108500

RESUMO

BACKGROUND: Cytokine-induced killer (CIK) cells are an emerging approach of cancer treatment. Our previous study have shown that CIK cells stimulated with combination of IL-2 and IL-15 displayed improved proliferation capacity and tumor cytotoxicity. However, the mechanisms of CIK cell proliferation and acquisition of cytolytic function against tumor induced by IL-2 and IL-15 have not been well elucidated yet. METHODS: CIK(IL-2) and CIK(IL-15) were generated from peripheral blood mononuclear cells primed with IFN-γ, and stimulated with IL-2 and IL-15 in combination with OKT3 respectively. RNA-seq was performed to identify differentially expressed genes, and gene ontology and pathways based analysis were used to identify the distinct roles of IL-2 and IL-15 in CIK preparation. RESULTS: The results indicated that CIKIL-15 showed improved cell proliferation capacity compared to CIK(IL-2). However, CIK(IL-2) has exhibited greater tumor cytotoxic effect than CIKIL-15. Employing deep sequencing, we sequenced mRNA transcripts in CIK(IL-2) and CIK(IL-15). A total of 374 differentially expressed genes (DEGs) were identified including 175 up-regulated genes in CIK(IL-15) and 199 up-regulated genes in CIK(IL-2)). Among DEGs in CIK(IL-15), Wnt signaling and cell adhesion were significant GO terms and pathways which related with their functions. In CIK(IL-2, type I interferon signaling and cytokine-cytokine receptor interaction were significant GO terms and pathways. We found that the up-regulation of Wnt 4 and PDGFD may contribute to enhanced cell proliferation capacity of CIK(IL-15), while inhibitory signal from interaction between CTLA4 and CD80 may be responsible for the weak proliferation capacity of CIK(IL-2). Moreover, up-regulated expressions of CD40LG and IRF7 may make for improved tumor cytolytic function of CIK(IL-2) through type I interferon signaling. CONCLUSIONS: Through our findings, we have preliminarily elucidated the cells proliferation and acquisition of tumor cytotoxicity mechanism of CIK(IL-15) and CIK(IL-2). Better understanding of these mechanisms will help to generate novel CIK cells with greater proliferation potential and improved tumor cytolytic function.


Assuntos
Células Matadoras Induzidas por Citocinas/citologia , Células Matadoras Induzidas por Citocinas/efeitos dos fármacos , Perfilação da Expressão Gênica , Interleucina-15/farmacologia , Interleucina-2/farmacologia , Neoplasias/genética , Neoplasias/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Matadoras Induzidas por Citocinas/metabolismo , Ontologia Genética , Redes Reguladoras de Genes/efeitos dos fármacos , Humanos , Análise de Sequência de RNA
12.
Hypertension ; 63(6): 1260-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24614212

RESUMO

Pulmonary arterial hypertension (PAH) is a devastating cardiopulmonary disorder with significant morbidity and mortality in patients with various lung and heart diseases. PAH is characterized by vascular obstruction which leads to a sustained increased pulmonary vascular resistance, vascular remodeling, and right ventricular hypertrophy and failure. Limited PAH therapies indicate that novel approaches are urgently needed for the treatment of PAH. Nuclear factor-κB (NF-κB) has been shown to play an important role in different cardiac pathologies; however, the role of NF-κB remains limited in the setting of PAH. Here, we investigated whether NF-κB inhibition in the lungs using Club (Clara) cell-10 promoter driving IκBα mutant had any effect in monocrotaline (MCT)-induced PAH mouse model. Our data revealed that MCT-induced PAH and right ventricular hypertrophy were associated with NF-κB activation, inflammatory response, and altered expression of bone morphogenetic protein receptor 2, inhibitor of differentiation, and Notch-3 signaling molecules in wild-type mice; and all these alterations were prevented in IκBα mutant mice treated with MCT. Moreover, endothelial cell apoptosis and endothelial-to-mesenchymal transition occurred in the lungs of MCT-treated wild-type mice and were restored in IκBα mutant+MCT mice, indicating an association with NF-κB signaling. In lung microvascular endothelial cells, IκBα (AA) mutant plasmid restored the decreased bone morphogenetic protein receptor 2 protein level and reversed the endothelial-to-mesenchymal transition process induced by transforming growth factor-ß1. We conclude that NF-κB regulates bone morphogenetic protein receptor 2-inhibitor of differentiation-Notch-3 axis genes and the subsequent endothelial cell apoptosis and endothelial-to-mesenchymal transition events in the lungs, providing new mechanistic information about MCT-induced PAH and right ventricular hypertrophy.


Assuntos
Hipertensão Pulmonar/metabolismo , Proteínas I-kappa B/metabolismo , Pulmão/metabolismo , NF-kappa B/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Núcleo Celular/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Endoteliais/metabolismo , Expressão Gênica , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/genética , Hipertrofia Ventricular Direita/genética , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/fisiopatologia , Proteínas I-kappa B/genética , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Monocrotalina , Mutação , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Interferência de RNA , Receptor Notch3 , Receptores Notch/genética , Receptores Notch/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição RelA/metabolismo
13.
Tumour Biol ; 35(3): 1997-2007, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24104501

RESUMO

Generation of cytokine-induced killer (CIK) cells is an emerging approach in adoptive donor lymphocyte infusion for patients with a wide range of tumors. However, our previous in vitro studies have shown that the killing efficacy of CIK cells against lung cancer was lower than other tumor cells, while the underlying mechanisms are not clear. We explored the feasibility to improve CIK cells mediated cytotoxicity against lung cancer. Interleukin (IL)-15 is a pleiotropic cytokine that stimulates cytolytic activity and cytokine secretion of NK cells, which may enhance the cytotoxic activity of CIK cells. In this study, we intended to stimulate the CIK cells by IL-2 in combination with IL-15 in cell expansion to achieve enhanced cytotoxicity against lung cancer cells. The different phenotypes of IL-2 or combination of IL-2 and IL-15 stimulated cytokine-induced killer cells were determined, and the improved cytotoxicity of IL-2 and IL-15 induced CIK cells against lung adenocarcinoma were evaluated both in vitro and in vivo. CIK cells stimulated with both IL-2 and IL-15 has shown greater proliferative potential than CIK cells treated with IL-2 alone. IL-15 induction also has driven the expansion of CD3+CD56+ subset and significantly enhanced cytotoxicity against tumor cells. Further analysis has demonstrated that CIKIL-2&IL-15 injected mice models have shown significant tumor regression and lower expression level of CyclinD1 in tumor tissue. This study has provided preclinical evidences that CIKIL-2&IL-15 with enhanced cytotoxicity may offer alternative treatment option for patients with lung cancer.


Assuntos
Adenocarcinoma/imunologia , Células Matadoras Induzidas por Citocinas/imunologia , Imunoterapia Adotiva/métodos , Interleucina-15/imunologia , Interleucina-2/imunologia , Neoplasias Pulmonares/imunologia , Adenocarcinoma/terapia , Adenocarcinoma de Pulmão , Animais , Apoptose/imunologia , Técnicas de Cultura de Células/métodos , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Endogâmicos BALB C , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Mol Cell Biochem ; 387(1-2): 135-41, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24178239

RESUMO

Angiotensin II(Ang II)-stimulated cardiomyocytes hypertrophy and apoptosis are associated with nuclear factor-κB (NF-κB) activation. NF-κB, a redox-sensitive transcription factor, contributes a critical role in cell death, but, Ang II-stimulated NF-κB-mediated cardiomyocytes apoptosis remains less understood. Recently, microRNAs (miRNAs) have been shown to be critical regulators in various cardiac remodeling processes; however, NF-κB-mediated miRNA's role in cardiomyocytes apoptosis remains undetermined. The miR-30b has been implicated in diverse cardiac remodeling; but, NF-κB-mediated miR-30b modulation in Ang II-induced cardiomyocytes death is currently unknown. In the present study, neonatal cardiomyocytes were pretreated with SN50, a selective cell permeable peptide inhibitor of NF-κB, or transfected with miR-30b mimetic and inhibitors separately, and then challenged with Ang II. The target gene, Bcl-2, and NF-κB transcriptional activity were analyzed. Our results demonstrated that NF-κB positively regulated miR-30b expression in Ang II-induced cardiomyocytes apoptosis, and Bcl-2 was a direct target for miR-30b. NF-κB further regulated the expression of Bcl-2 in the above setting. Furthermore, Ang II-induced cardiomyocytes apoptosis rescued by inhibiting either NF-κB or miR-30b provided an important role in cardiomyocytes cell death. We evaluated a critical role of NF-κB-mediated miR-30b modulation in Ang II-stimulated cardiomyocytes targeting Bcl-2. Our data may provide a new insight of miR-30b's role in myocardial infarction or ischemia.


Assuntos
Apoptose , MicroRNAs/metabolismo , Miócitos Cardíacos/fisiologia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Regiões 3' não Traduzidas , Angiotensina II/fisiologia , Animais , Sequência de Bases , Células HEK293 , Humanos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Interferência de RNA , Ratos , Transcrição Gênica , Ativação Transcricional
15.
Cell Adh Migr ; 7(5): 404-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24192520

RESUMO

There is currently great interest in the use of mesenchymal stem cells as a therapy for multiple sclerosis with potential to both ameliorate inflammatory processes as well as improve regeneration and repair. Although most clinical studies have used autologous bone marrow-derived mesenchymal stem cells, other sources such as allogeneic umbilical cord-derived cells may provide a more accessible and practical supply of cells for transplantation. In this case report we present the treatment of aggressive multiple sclerosis with multiple allogenic human umbilical cord-derived mesenchymal stem cell and autologous bone marrow-derived mesenchymal stem cells over a 4 y period. The treatments were tolerated well with no significant adverse events. Clinical and radiological disease appeared to be suppressed following the treatments and support the expansion of mesenchymal stem cell transplantation into clinical trials as a potential novel therapy for patients with aggressive multiple sclerosis.


Assuntos
Transplante de Células-Tronco Mesenquimais/efeitos adversos , Células-Tronco Mesenquimais/citologia , Esclerose Múltipla Recidivante-Remitente/terapia , Adulto , Células da Medula Óssea/citologia , Humanos , Masculino , Esclerose Múltipla Recidivante-Remitente/metabolismo , Esclerose Múltipla Recidivante-Remitente/patologia , Regeneração , Cordão Umbilical/citologia
16.
Exp Biol Med (Maywood) ; 237(6): 709-19, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22728706

RESUMO

Umbilical cord mesenchymal stem cells (UC-MSCs) have several advantages for clinical therapy: the material is easily obtainable, the donation procedure is painless and there is low risk of viral contamination. UC-MSCs play important roles in tissue regeneration, tissue damage repair, autoimmune disease and graft-versus-host disease. In this study, we investigated the normal mRNA expression profile of UC-MSCs, and analyzed the candidate proteins responsible for the signaling pathway that may affect the differentiation characteristics of UC-MSCs. UC-MSCs were isolated by mincing UC samples into fragments and placing them in growth medium in a six-well plate. The immunophenotype characteristics and multilineage differentiation potential of the UC-MSCs were measured by flow cytometry and immunohistochemical assays. In addition, the pathway-focused gene expression profile of UC-MSCs was compared with those of normal or tumorous cells by realtime quantitative polymerase chain reaction. We successfully isolated and cultured UC-MSCs and analyzed the appropriate surface markers and their capacity for osteogenic, adipogenic and neural differentiation. In total, 168 genes focusing on signal pathways were examined. We found that the expression levels of some genes were much higher or lower than those of control cells, either normal or tumorous. UC-MSCs exhibit a unique mRNA expression profile of pathway-focused genes, especially some stemness genes, which warrants further investigation.


Assuntos
Diferenciação Celular/fisiologia , Sangue Fetal/citologia , Perfilação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Células Cultivadas , Feminino , Sangue Fetal/fisiologia , Humanos , Imunofenotipagem , Células-Tronco Mesenquimais/fisiologia , Gravidez , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA