Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cytotherapy ; 24(4): 421-427, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35086778

RESUMO

BACKGROUND AIMS: To explore the long-term safety and benefit of umbilical cord mesenchymal stromal cell (MSCs) plus autologous bone marrow mononuclear cell (aBM-MNC) stem cell transplantation (SCT) without immunotherapy in established type 1 diabetes (T1D). METHODS: In the primary completion of this trial (ClinicalTrials.gov identifier: NCT01374854), the authors randomized patients (n = 21 per group) to either SCT or standard care (control) and previously reported effects on insulin secretion. The authors report about the incidence of chronic diabetes complications (primary endpoint) after 8 years of follow-up. The authors also report on secondary endpoints, safety, islet function and metabolic control. RESULTS: Data were obtained from 14 of 21 patients in the SCT group and 15 of 21 patients in the control group who completed follow-up. At 8 years, the incidence of peripheral neuropathy was 7.1% (one of 14) in the SCT group versus 46.7% (seven of 15) in the control group (P = 0.017). The incidence of diabetic nephropathy was 7.1% (one of 14) in the SCT group versus 40.0% (six of 15) in the control group (P = 0.039). The incidence of retinopathy was 7.1% (one of 14) in the SCT group versus 33.3% (five of 15) in the control group (P = 0.081). Two patients (two of 14, 14.3%) in the SCT group and 11 patients (11 of 15, 73.3%) in the control group developed at least one complication (P = 0.001). One and six patients in the SCT group and control group, respectively, had at least two complications (P = 0.039). No malignancies were reported in the treated group. CONCLUSIONS: Co-transplantation of umbilical cord MSCs and aBM-MNCs in patients with established T1D was associated with reduced incidence of chronic diabetes complications.


Assuntos
Complicações do Diabetes , Diabetes Mellitus Tipo 1 , Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Medula Óssea , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/terapia , Seguimentos , Humanos , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Projetos Piloto , Cordão Umbilical
2.
Int J Hyperthermia ; 38(1): 1304-1312, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34468276

RESUMO

BACKGROUND: Several studies have reported the combination of intracavity or cervical lymph node hyperthermia with chemoradiotherapy (CRT) to improve clinical outcomes in nasopharyngeal carcinoma (NPC), but the combination with whole-body hyperthermia (WBH) for treating NPC is unexplored. We aimed to assess the efficacy of the combination of radiotherapy, chemotherapy and WBH in patients with locoregionally advanced NPC. METHODS: Between July 2008 and November 2012, 239 newly diagnosed NPC patients were enrolled in a pre-propensity score-matched cohort, including 193 patients who received CRT (CRT group) and 46 who underwent CRT with WBH (HCRT group). The feasibility and clinical outcomes of both groups were evaluated and toxicities assessed. Survival rates were assessed using the Kaplan-Meier method, log-rank test and Cox regression. RESULTS: Following propensity score matching, 46 patients from each group were included. The 5-year overall survival (OS) rates were 65.2% in the CRT group and 80.3% in the HCRT group (p=.027). In contrast, the other survival outcomes at 5 years were similar between the groups: locoregional recurrence-free survival (LRRFS), 74.7% vs. 87.6% (p=.152); distant metastasis-free survival (DMFS), 67.4% vs. 77.9% (p=.125); and progression-free survival (PFS), 53.1% vs. 69.2% (p=.115). In the multivariate analyses, the only two independent predictors of OS were clinical stage and HCRT. CONCLUSIONS: These results suggest that WBH, when combined with CRT, can improve the OS of patients with advanced NPC.


Assuntos
Neoplasias Nasofaríngeas , Radioterapia de Intensidade Modulada , Quimiorradioterapia , Humanos , Hipertermia , Carcinoma Nasofaríngeo/tratamento farmacológico , Carcinoma Nasofaríngeo/radioterapia , Neoplasias Nasofaríngeas/tratamento farmacológico , Neoplasias Nasofaríngeas/radioterapia , Pontuação de Propensão , Estudos Retrospectivos
3.
Ren Fail ; 43(1): 520-529, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33719820

RESUMO

BACKGROUND: Kidneys obtained from deceased donors increase the incidence of delayed graft function (DGF) after renal transplantation. Here we investigated the influence of the risk factors of donors with DGF, and developed a donor risk scoring system for DGF prediction. METHODS: This retrospective study was conducted in 1807 deceased kidney donors and 3599 recipients who received donor kidneys via transplants in 29 centers in China. We quantified DGF associations with donor clinical characteristics. A donor risk scoring system was developed and validated using an independent sample set. RESULTS: The incidence of DGF from donors was 19.0%. Six of the donor characteristics analyzed, i.e., age, cause of death, history of hypertension, terminal serum creatinine, persistence of hypotension, and cardiopulmonary resuscitation (CPR) time were risk factors for DGF. A 49-point scoring system of donor risk was established for DGF prediction and exhibited a superior degree of discrimination. External validation of DGF prediction revealed area under the receiver-operating characteristic (AUC) curves of 0.7552. CONCLUSIONS: Our study determined the deceased donor risk factors related to DGF after renal transplantation pertinent to the Chinese cohort. The scoring system developed here had superior diagnostic significance and consistency and can be used by clinicians to make evidence-based decisions on the quality of kidneys from deceased donors and guide renal transplantation therapy.


Assuntos
Função Retardada do Enxerto/etiologia , Transplante de Rim/efeitos adversos , Doadores de Tecidos/estatística & dados numéricos , Adulto , Morte Encefálica , China , Isquemia Fria/efeitos adversos , Creatinina/análise , Função Retardada do Enxerto/terapia , Feminino , Sobrevivência de Enxerto , Humanos , Incidência , Rim/fisiopatologia , Masculino , Pessoa de Meia-Idade , Curva ROC , Diálise Renal/estatística & dados numéricos , Estudos Retrospectivos , Fatores de Risco , Transplante Homólogo , Transplantes/fisiopatologia
4.
J Cell Physiol ; 234(11): 21060-21075, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31020657

RESUMO

MicroRNAs have emerged as key regulators involved in a variety of biological processes. Previous studies have demonstrated that miR-192/215 participated in progression of Crohn's disease and colorectal cancer. However, their concrete relationships and regulation networks in diseases remain unclear. Here, we used bioinformatics methods to expound miR-192/215-5p macrocontrol regulatory networks shared by two diseases. For data mining and figure generation, several miRNA prediction tools, Human miRNA tissue atlas, FunRich, miRcancer, MalaCards, STRING, GEPIA, cBioPortal, GEO databases, Pathvisio, Graphpad Prism 6 software, etc . are extensively applied. miR-192/215-5p were specially distributed in colon tissues and enriched biological pathways were closely associated with human cancers. Emerging role of miR-192/215-5p and their common pathways in Crohn's disease and colorectal cancer was also analyzed. Based on results derived from multiple approaches, we identified the biological functions of miR-192/215-5p as a tumor suppressor and link Crohn's disease and colorectal cancer by targeting triglyceride synthesis and extracellular matrix remodeling pathways.


Assuntos
Neoplasias Colorretais/genética , Doença de Crohn/genética , MicroRNAs/genética , Neoplasias Colorretais/metabolismo , Biologia Computacional , Doença de Crohn/complicações , Doença de Crohn/metabolismo , Matriz Extracelular/metabolismo , Genes Supressores de Tumor , Humanos , Redes e Vias Metabólicas/genética , Triglicerídeos/biossíntese
5.
Biochem Biophys Res Commun ; 501(1): 212-219, 2018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29715459

RESUMO

We had previously demonstrated that increased expression of ErbB3 is required for ErbB2-mediated paclitaxel resistance in breast cancer cells. In the present study, we have explored the possible role of mesenchymal stem cells (MSCs) in regulating the paclitaxel-sensitivity of ErbB2/ErbB3-coexpressing breast cancer cells. We show that human umbilical cord-derived MSCs express significantly higher level of neuregulin-1 as compared with ErbB2/ErbB3-coexpressing breast cancer cells themselves. Coculture or treatment with conditioned medium of MSCs not only decreases the anti-proliferation effect of paclitaxel on ErbB2/ErbB3-coexpressing breast cancer cells, but also significantly inhibits paclitaxel-induced apoptosis. We further demonstrate that this MSCs-drived paclitaxel resistance in ErbB2/ErbB3-coexpressing breast cancer cells could be attributed to upregulation of Survivin via paracrine effect of NRG-1/ErbB3/PI-3K/Akt signaling, as either specific knockdown expression of ErbB3, or blocking of downstream PI-3K/Akt signaling, or specific inhibition of Survivin can completely reverse this effect. Moreover, targeted knockdown of NRG-1 expression in MSCs abrogates theirs effect on paclitaxel sensitivity of ErbB2/ErbB3-coexpressing breast cancer cells. Taken together, our study indicate that paracrine of NRG-1 by MSCs induces paclitaxel resistance in ErbB2/ErbB3-coexpressing breast cancer cells through PI-3K/Akt signaling-dependent upregulation of Survivin. Our findings suggest that simultaneously targeting mesenchymal stem cells in tumor microenvironment may be a novel strategy to overcome paclitaxel resistance in patients with ErbB2/ErbB3-coexpressing breast cancer.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Neuregulina-1/metabolismo , Paclitaxel/farmacologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Genes erbB-2 , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Neuregulina-1/antagonistas & inibidores , Neuregulina-1/genética , Comunicação Parácrina , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Survivina
6.
Med Sci Monit ; 24: 8250-8263, 2018 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-30444862

RESUMO

BACKGROUND A variety of treatment strategies have been developed for clear cell kidney carcinoma (KIRC); however, there is still a need for effective therapeutic targets and prognostic molecular biomarkers. Given that long noncoding RNAs (lncRNAs) has been emerging as an important regulator in tumorigenesis, we explored potential functional lncRNAs in KIRC by comprehensively analyzing the lncRNA-miRNA-mRNA regulatory network with bioinformatics processing tools. MATERIAL AND METHODS RNA-seq/miRNA-seq data of KIRC in The Cancer Genome Atlas (TCGA) were obtained and analyzed. The "edgeR" package in R software was used to identify differentially expressed lncRNAs (DElncRNAs, differentially expressed long noncoding RNAs), miRNAs (DEmiRNAs, differentially expressed micro RNAs), and mRNAs (DEmRNAs, differentially expressed messenger RNAs) in KIRC and normal samples. A global triple network was conducted based on the competing endogenous RNA (ceRNA) theory, and survival analysis was conducted by "survival" package in R software. RESULTS A total of 4246 DElncRNAs, 179 DEmiRNAs, and 5758 DEmRNAs were identified, among which a subset of them (321 lncRNAs, 26 miRNAs, and 1068 mRNAs) were found to constitute a global ceRNA network in KIRC. Four lncRNAs (ENTPD3-AS1, FGD5-AS1, LIFR-AS1, and UBAC2-AS1) were revealed to be potential therapeutic targets as well as prognostic biomarkers of KIRC by our extensive functional analysis. CONCLUSIONS We reported here the identification of functional lncRNAs in KIRC via a TCGA data-based bioinformatics analysis. We believe that this study might contribute to improving the comprehension of the lncRNA-mediated ceRNA regulatory mechanisms in the tumorigenesis of KIRC. Meanwhile, our results suggested that 4 lncRNAs might act as potential therapeutic targets or candidate prognostic biomarkers in KIRC.


Assuntos
Carcinoma de Células Renais/genética , Neoplasias Renais/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Mensageiro/genética , Biomarcadores Tumorais , Biologia Computacional/métodos , Bases de Dados Genéticas , Redes Reguladoras de Genes , Humanos , Estimativa de Kaplan-Meier , Prognóstico , Análise de Sobrevida
7.
Clin Lab ; 62(3): 409-16, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27156331

RESUMO

BACKGROUND: Cyclin-dependent kinase inhibitor 1A (CDKN1A) and Cyclin D1 (CCND1) play essential roles in the regulation of cell cycle progression and are closely associated with human cancer. CDKN1A and CCND1 single nucleotide polymorphisms (SNPs) have been demonstrated to influence the prognosis in humans with different cancers. However, their roles in the prognosis of patients with resected gastric adenocarcinoma (RGA) remain to be determined. METHODS: Genotypes of CDKN1A rs1059234 and CCND1 rs603965 SNPs were performed in 235 tissue samples from RGA. The association of the genotypes of these two SNPs with the prognosis in the patients with RGA was analyzed by X2 test, multivariate Cox regression analyses, and Kaplan Meier curves. RESULTS: During the 50 months of median follow-up time, the overall recurrence and survival rate in the whole group was 57.4% and 46.8%, respectively. Whereas, recurrence and survival rate in patients with CC genotype of rs1059234 located in 3'UTR of CDKN1A were 78.0% and 27.1% (p = 0.004; p = 0.006). Multivariate analyses further confirmed that the CC genotype was significantly related with both increased recurrence and death risk (HR 3.33, 95% CI 1.95-5.70; p = 1.07 x 10⁻5, and HR 3.45, 95% CI 1.95-6.10; p = 2.03 x 10⁻5). No significant difference among CCND1 rs603965 SNP with the prognosis was determined. CONCLUSIONS: rs1059234 of CDKN1A is closely associated with the prognosis in patients with RGA.


Assuntos
Adenocarcinoma/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Polimorfismo de Nucleotídeo Único , Neoplasias Gástricas/genética , Adenocarcinoma/mortalidade , Adulto , Idoso , Feminino , Genes bcl-1 , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Neoplasias Gástricas/mortalidade
8.
Cytotherapy ; 17(4): 454-7, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25559146

RESUMO

BACKGROUND AIMS: Traditional bone marrow (BM) collection is inadequate for separation of abundant mononuclear cells (MNCs). We aimed to investigate the effects of preoperative exercise on BM collection in patients with type 2 diabetes mellitus (T2DM). METHODS: Sixty patients with T2DM were randomly assigned to either a control group or an exercise group (n = 30 each). The patients in the exercise group exercised before the collection. All patients underwent routine surgical care. The collected BM volume, operation duration, collecting speed, puncture times and pain scores were recorded. BM samples were tested before and after MNCs separation for CD34+ flow cytometry and whole blood cell count. RESULTS: The collected BM volumes were significantly larger and collection speed was faster in the exercise group (379.77 ± 4.93 mL and 1.40 ± 0.14 mL/s) than those in the control group (356.67 ± 15.36 mL and 0.89 ± 0.16 mL/s, P = 0.00 for both). Puncture times were significantly less and pain scores were lower in the exercise group (2.07 ± 0.25 and 2.67 ± 1.56) than those in the control group (2.50 ± 0.63 and 3.43 ± 1.76, P = 0.00 and 0.02, respectively). CD34+ cells and whole blood cell count variables were comparable in the 2 groups. CONCLUSIONS: Preoperative exercise facilitates BM collection by increasing collected volume, improving collecting speed, relieving patients' pains and ensuring MNC quality.


Assuntos
Células da Medula Óssea/citologia , Separação Celular/métodos , Terapia Baseada em Transplante de Células e Tecidos , Diabetes Mellitus Tipo 2/fisiopatologia , Exercício Físico/fisiologia , Monócitos/citologia , Adulto , Idoso , Diabetes Mellitus Tipo 2/terapia , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/transplante , Cuidados Pré-Operatórios
9.
J BUON ; 20(2): 505-13, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26011343

RESUMO

PURPOSE: To observe the efficacy and side effects of adjuvant dendritic cells' (DCs) vaccine combined with cytokine-induced killer cell (CIK) therapy after renal cell carcinoma (RCC) surgery (RCCS). METHODS: DCs vaccine and CIK that loaded the autologous tumor cell lysate were prepared in vitro. Four hundred and ten RCC patients were recruited, and the study group was given DCs-CIK immunotherapy, while the control group was given IFN-α therapy. RESULTS: Disease progression (recurrence, metastasis or death) showed significant differences between the two groups in clinical stage I and II patients, as well as in highly and moderately differentiated disease (p<0.05), while there was no significant difference between the two groups in patients with poorly differentiated disease (p>0.05). The 3- and 5-year overall survival rates of the DCs-CIK group (96% and 96%, respectively) exhibited significant difference compared to the IFN-α group (83% and 74%, respectively (p<0.01). Progression-free survival (PFS) between the two groups was significantly different (p<0.01). Tumor stage and DCs-CIK treatment were independent factors concerning prognosis of RCC (p<0.05). There was no severe toxicity observed in the DCs-CIK treatment group. CONCLUSIONS: Adjuvant post-RCCS DCs-CIK treatment prolonged PFS and reduced mortality, showing better overall activity compared to interferon treatment.


Assuntos
Vacinas Anticâncer/imunologia , Carcinoma de Células Renais/terapia , Células Matadoras Induzidas por Citocinas/imunologia , Células Dendríticas/imunologia , Neoplasias Renais/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Vacinas Anticâncer/efeitos adversos , Carcinoma de Células Renais/mortalidade , Terapia Combinada , Progressão da Doença , Feminino , Humanos , Interferon-alfa/efeitos adversos , Interferon-alfa/uso terapêutico , Neoplasias Renais/mortalidade , Masculino , Pessoa de Meia-Idade
10.
Mol Cancer ; 13: 18, 2014 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-24479681

RESUMO

BACKGROUND: Interleukin-1ß (IL-1ß) has been implicated in the progression of gastric adenocarcinoma (GA); however, the molecular mechanisms of action of IL-1ß in GA are poorly characterized. P38 and JNK are the major MAPK family members that regulate IL-1ß signaling pathways. Here, we investigated the role of both p38 and JNK in IL-1ß-induced GA cell migration, invasion and metastatic potential. METHODS: The effects of IL-1ß-induced p38 and JNK activation in GA cells were determined using in vitro Transwell migration and invasion assays of MKN-45 and AGS cells, or an in vivo metastasis assay in nude mice. The IL-1ß-induced p38 signaling pathway was further characterized in GA cells. Activation of the IL-1ß/p38 signaling pathway was also assessed in human primary GA tissues by immunohistochemistry. RESULTS: IL-1ß-induced activation of p38 increased GA cell migration and invasion in vitro and promoted the metastatic potential of GA cells in vivo; these effects were attenuated by p38 siRNA or the p38 inhibitor SB202190. MMP2 or MMP9 siRNAs and the MMP2/9 inhibitor BiPS also inhibited IL-1ß-induced GA cell migration and invasion in vitro. IL-1ß-induced p38 activation significantly increased MMP2 and MMP9 mRNA and protein expression and activity. Luciferase reporter assays demonstrated that the activator protein-1 (AP-1) and the AP-1 binding sites of the MMP9 promoter (-670/MMP9) were activated by IL-1ß-induced p38 activation. Phospho-p38 was significantly upregulated in human GA tissues (compared to matched non-neoplastic tissues), and significantly associated with lymph node metastasis, and invasion beyond the serosa. Expression of phospho-p38 significantly correlated with IL-1ß, MMP2, MMP9, and c-fos expression in both human GA tissues and GA cell metastases in the lungs of nude mice. IL-1ß was also capable of activating JNK in GA cells, but activation of JNK was not associated with GA cell migration and invasion. Therefore, IL-1ß-induced the migration and invasion in GA cells were regulated by p38, but not by JNK. CONCLUSIONS: IL-1ß-induced p38 activation and the IL-1ß/p38/AP-1(c-fos)/MMP2 & MMP9 pathway play an important role in metastasis in GA; this pathway may provide a novel therapeutic target for GA.


Assuntos
Adenocarcinoma/metabolismo , Interleucina-1beta/metabolismo , Transdução de Sinais/fisiologia , Neoplasias Gástricas/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Ativação Enzimática/fisiologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Nus , Microscopia Confocal , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/patologia , Fator de Transcrição AP-1/metabolismo , Transfecção , Regulação para Cima
11.
Biochem Biophys Res Commun ; 453(3): 405-10, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25268319

RESUMO

The direct conversion of one cell type to another without an intermediate pluripotent stage is required for regenerative therapies. The ventral pancreas and liver share a common developmental origin. Recent studies have shown that hepatocytes could be induced to transdifferentiate into insulin-producing cells. In this paper, we showed a new strategy to achieve the direct conversion of human hepatocytes into surrogate ß cells. Hepatocytes were transfected with microRNA-302 (miR-302) mimic and Pdx1, Ngn3 and MafA expressed plasmids, followed by a chemical-defined culture system for maturation of insulin-secreting cells. Co-transfection of miR-302 mimic increased the transcription of pancreatic development-related genes (Sox17, Foxa2, and endogenous Pdx1). Furthermore, at the end of this treatment, hepatocytes became insulin expressed cells that released the hormone in response to a physiological glucose change in vitro. This work shows that miR-302 participation may facilitates the conversion of adult hepatocytes into pancreatic islets-like cells.


Assuntos
Diferenciação Celular/fisiologia , Hepatócitos/citologia , Ilhotas Pancreáticas/citologia , MicroRNAs/fisiologia , Sequência de Bases , Células Cultivadas , Meios de Cultura , Primers do DNA , Humanos , MicroRNAs/genética , Reação em Cadeia da Polimerase
12.
Cytotherapy ; 16(2): 258-65, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24290656

RESUMO

BACKGROUND AIMS: The use of bone marrow mononuclear cells (BM-MNCs) has achieved great outcomes in clinical practice. We aim to evaluate the efficacy and safety of autologous BM-MNC infusion and hyperbaric oxygen therapy (HOT) in type 2 diabetes mellitus. METHODS: This single-center, randomized, open-label, controlled clinical trial with a factorial design included two phases. The patients received standard medical therapy in the run-in phase; in the treatment phase, patients with glycated hemoglobin of 7.5-9.5% were randomly assigned into four groups and underwent BM-MNC infusion along with HOT (BM-MNC+HOT group), BM-MNC infusion (BM-MNC group), HOT (HOT group) and standard medical therapy (control group), respectively. The area under the curve of C-peptide was recorded as a primary end point. Our research is registered at ClinicalTrials.gov (NCT00767260). RESULTS: A total of 80 patients completed the follow-up. At 12 months after treatment, the area under the curve of C-peptide (ng/mL per 180 min) of the BM-MNC+HOT group and the BM-MNC group were significantly improved (34.0% and 43.8% from the baseline, respectively). The changes were both significant compared with that in the control group, but no remarkable change was observed in the HOT group. Treatment-related adverse events were mild, including transient abdominal pain (n = 5) and punctual hemorrhage (n = 3). CONCLUSIONS: BM-MNC infusion for type 2 diabetes mellitus improves islet function and metabolic control, with mild adverse effects. HOT does not synergize with BM-MNC infusion.


Assuntos
Células da Medula Óssea/metabolismo , Transplante de Células , Diabetes Mellitus Tipo 2/terapia , Oxigenoterapia Hiperbárica , Células Secretoras de Insulina/metabolismo , Leucócitos Mononucleares/transplante , Idoso , Células da Medula Óssea/patologia , Células Cultivadas , Terapia Combinada , Feminino , Seguimentos , Humanos , Células Secretoras de Insulina/patologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
13.
Cytotherapy ; 16(4): 471-84, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24119646

RESUMO

BACKGROUND AIMS: Minimal change nephrotic syndrome is the most frequent cause of nephrotic syndrome in childhood. Current treatment regimes, which include glucocorticoid hormones and immunosuppressive therapy, are effective and have fast response. However, because of the side effects, long treatment course, poor patient compliance and relapse, novel approaches for the disease are highly desired. METHODS: The adriamycin-induced nephrotic rat model was established. Rats were allocated to a model group, a prednisone group or mesenchymal stromal cell (MSC) group. Clinical parameters in each treatment group were determined at 2 weeks, 4 weeks and 8 weeks. The messenger RNA (mRNA) levels of synaptopodin, p21 and monocyte chemoattractant protein-1 were determined through the use of quantitative real-time-polymerase chain reaction. Protein levels were determined by means of Western blot or enzyme-linked immunosorbent assay. Podocytes were isolated and apoptotic rate after adriamycin with or without MSC treatment was analyzed by means of flow cytometry. RESULTS: MSC intervention improved renal function as assessed by urinary protein, blood creatinine and triglyceride levels. MSC intervention reduced adriamycin-induced renal tissue damage visualized by immunohistochemistry and light and electron microscopic analysis and reduced adriamycin-induced podocyte apoptosis. After MSC intervention, mRNA and protein levels of synaptopodin and p21 in renal cortex were significantly increased. MSCs also restored synaptopodin mRNA and protein expression in isolated podocytes. In addition, monocyte chemoattractant protein-1 mRNA in renal cortex and protein level in serum of the MSC treatment group were significantly decreased compared with that in the adriamycin-induced nephropathy model group. CONCLUSIONS: Our data indicate that MSCs could protect rats from adriamycin-induced minimal change nephrotic syndrome, and the protective effects of MSCs are mediated through multiple actions.


Assuntos
Rim/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais , Nefrose Lipoide/patologia , Nefrose Lipoide/terapia , Animais , Quimiocina CCL2/biossíntese , Doxorrubicina/toxicidade , Regulação da Expressão Gênica , Humanos , Rim/patologia , Células-Tronco Mesenquimais/citologia , Proteínas dos Microfilamentos/biossíntese , Nefrose Lipoide/induzido quimicamente , Prednisona/administração & dosagem , RNA Mensageiro/biossíntese , Ratos , Proteínas rho de Ligação ao GTP/biossíntese
14.
J BUON ; 19(2): 512-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24965414

RESUMO

PURPOSE: This study aimed to investigate the expression of p53 and Ki67 genes in renal cell carcinoma (RCC) and its possible clinical value. METHODS: A retrospective analysis of clinical data from 1239 patients with RCC was performed to explore the relationship between the expression of Ki67 and p53 proteins and tumor stage, grade and prognosis. RESULTS: p53 expression was not significantly correlated with TNM stage and Fuhrman grade (p>0.05); Ki67 expression was significantly correlated with TNM stage and Fuhrman grade (p<0.05). Kaplan-Meier and log-rank survival rate results showed that the prognosis of Ki67 and p53 double-positive group was significantly inferior to the single-positive and negative group (p<0.001). In the multivariate Cox risk regression analysis model, TNM stage, relative risk/RR=3.196, p<0.001), Fuhrman grade (RR=3.196, p<0.001) and Ki67 and p53 double-positive [Ki67 (+) p53 (+) , RR=3.196, p<0.001] were significantly correlated with tumor prognosis, and independent predictors of the patient disease-free survival (DFS). CONCLUSION: The combined detection of p53 and Ki67 expressions, which are superior to single marker, could be used to improve significantly the accuracy of prognosis of RCC patients.


Assuntos
Carcinoma de Células Renais/patologia , Antígeno Ki-67/análise , Neoplasias Renais/patologia , Proteína Supressora de Tumor p53/análise , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Renais/mortalidade , Feminino , Humanos , Neoplasias Renais/mortalidade , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Modelos de Riscos Proporcionais , Estudos Retrospectivos
15.
BMC Cell Biol ; 14: 18, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-23510080

RESUMO

BACKGROUND: The objective of this study was to evaluate the effect of bone marrow mesenchymal stem cells (BMSCs) on the apoptosis of granulosa cells (GCs) in rats. RESULTS: Cisplatin increased GC apoptosis from 0.59% to 13.04% in the control and cisplatin treatment groups, respectively, which was significantly reduced upon co-culture with BMSCs to 4.84%. Cisplatin treatment increased p21 and bax and decreased c-myc mRNA expression, which was reversed upon co-culture with BMSCs. As compared to young rats, increased apoptosis was observed in the perimenopausal rats (P < 0.001). After 3 months, the apoptosis rate in the BMSC group was significantly lower than that of the control group (P = 0.007). CONCLUSIONS: BMSC therapy may protect against GC apoptosis induced by cisplatin and perimenopause. Further studies are necessary to evaluate therapeutic efficacy of BMSCs.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Cisplatino/administração & dosagem , Técnicas de Cocultura/métodos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células da Granulosa/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Perimenopausa/fisiologia , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Estrogênios/administração & dosagem , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/fisiologia , Células-Tronco Mesenquimais/fisiologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
16.
Am J Physiol Renal Physiol ; 304(4): F440-50, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23235477

RESUMO

C/EBP homologous protein (CHOP) is an important mediator of endoplasmic reticulum (ER) stress-induced cell and organ injury. Here we show that lipopolysaccharide (LPS)-induced acute kidney injury (AKI) is associated with ER stress and elevated CHOP. We postulated that CHOP(-/-) mice would be protected against LPS-induced-AKI. Unexpectedly, while Toll-like receptor 4 (TLR4) expression levels were comparable in kidneys of CHOP(-/-) and wild-type (WT) mice, CHOP(-/-) mice developed more severe AKI after LPS injection. Furthermore, the severe kidney injury in CHOP(-/-) mice was associated with an exaggerated inflammatory response. Serum TNF-α levels were more elevated in LPS-treated CHOP(-/-) mice. There was a 3.5-fold higher amount of renal neutrophil infiltrates in LPS-treated CHOP(-/-) than in WT mice. Additionally, the kidneys of LPS-treated CHOP(-/-) mice had a more prominent increase in NF-κB activation and further upregulation of proinflammatory genes, i.e., c-x-c motif ligand 1 (CXCL-1), macrophage inflammatory protein-2 (MIP-2), and IL-6. Finally, proximal tubules, glomeruli, and podocytes isolated from CHOP(-/-) mice also had an exaggerated proinflammatory response to LPS. Since LPS directly increased CHOP in glomeruli and podocytes of WT mice, together these data suggest that the LPS-induced increase of CHOP in kidneys may inhibit inflammatory response in renal cells and provide protection against AKI.


Assuntos
Injúria Renal Aguda/metabolismo , Inflamação/metabolismo , Sepse/complicações , Fator de Transcrição CHOP/deficiência , Injúria Renal Aguda/microbiologia , Injúria Renal Aguda/patologia , Animais , Células Cultivadas , Estresse do Retículo Endoplasmático/fisiologia , Feminino , Inflamação/microbiologia , Inflamação/patologia , Glomérulos Renais/fisiologia , Lipopolissacarídeos , Macrófagos Peritoneais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Podócitos/fisiologia , Estresse Fisiológico , Fator de Transcrição CHOP/genética
17.
Breast Cancer Res ; 15(5): R101, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24168763

RESUMO

INTRODUCTION: Elevated expression of erbB3 rendered erbB2-overexpressing breast cancer cells resistant to paclitaxel via PI-3 K/Akt-dependent upregulation of Survivin. It is unclear whether an erbB3-targeted therapy may abrogate erbB2-mediated paclitaxel resistance in breast cancer. Here, we study the antitumor activity of an anti-erbB3 antibody MM-121/SAR256212 in combination with paclitaxel against erbB2-overexpressing breast cancer. METHODS: Cell growth assays were used to determine cell viability. Cells undergoing apoptosis were quantified by a specific apoptotic ELISA. Western blot analyses were performed to assess the protein expression and activation. Lentiviral vector containing shRNA was used to specifically knockdown Survivin. Tumor xenografts were established by inoculation of BT474-HR20 cells into nude mice. The tumor-bearing mice were treated with paclitaxel and/or MM-121/SAR256212 to determine whether the antibody (Ab) enhances paclitaxel's antitumor activity. Immunohistochemistry was carried out to study the combinatorial effects on tumor cell proliferation and induction of apoptosis in vivo. RESULTS: MM-121 significantly facilitated paclitaxel-mediated anti-proliferative/anti-survival effects on SKBR3 cells transfected with a control vector or erbB3 cDNA. It specifically downregulated Survivin associated with inactivation of erbB2, erbB3, and Akt. MM-121 enhances paclitaxel-induced poly(ADP-ribose) polymerase (PARP) cleavage, activation of caspase-8 and −3, and apoptosis in both paclitaxel-sensitive and -resistant cells. Specific knockdown of Survivin in the trastuzumab-resistant BT474-HR20 cells dramatically enhanced paclitaxel-induced apoptosis, suggesting that increased Survivin caused a cross-resistance to paclitaxel. Furthermore, the studies using a tumor xenograft model-established from BT474-HR20 cells revealed that either MM-121 (10 mg/kg) or low-dose (7.5 mg/kg) paclitaxel had no effect on tumor growth, their combinations significantly inhibited tumor growth in vivo. Immunohistochemical analysis showed that the combinations of MM-121 and paclitaxel significantly reduced the cells with positive staining for Ki-67 and Survivin, and increased the cells with cleaved caspase-3. CONCLUSIONS: The combinations of MM-121 and paclitaxel not only inhibit tumor cell proliferation, but also promote erbB2-overexpressing breast cancer cells to undergo apoptosis via downregulation of Survivin in vitro and in vivo, suggesting that inactivation of erbB3 with MM-121 enhances paclitaxel-mediated antitumor activity against erbB2-overexpressing breast cancers. Our data supports further exploration of the combinatorial regimens consisting of MM-121 and paclitaxel in breast cancer patients with erbB2-overexpressing tumors, particularly those resistant to paclitaxel.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias da Mama/genética , Paclitaxel/farmacologia , Receptor ErbB-2/genética , Receptor ErbB-3/antagonistas & inibidores , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Camundongos , Receptor ErbB-2/metabolismo , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Survivina , Trastuzumab , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Br Med Bull ; 105: 139-55, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23396319

RESUMO

INTRODUCTION: Antibody is a major cause of allograft injury. However, it has not been routinely tested post-transplant. SOURCES OF DATA: A literature search was performed using PubMed on the topics of 'antibody monitoring', 'autoantibody and allograft dysfunction' and 'prevention and treatment of antibody-mediated rejection (AMR)'. AREAS OF AGREEMENT: Donor-specific antibody (DSA) monitoring not only helps to identify patients at risk of AMR, but also serves as a biomarker to personalize patient's maintenance immunosuppression. Development of autoantibody is a secondary response following primary tissue injury. Some autoantibodies are directly involved in allograft injury, while others only serve as biomarkers of tissue injury. AREAS OF CONTROVERSY: It remains controversial whether DSA-positive patients without symptoms need to be treated. In addition, given the variation in study designs and patient's characteristics, there is discrepancy regarding which treatment regimens provide optimal clinical outcome in preventing/treating AMR. GROWING POINTS: Efficacy of B-cell and/or antibody-targeted therapies in treating or preventing AMR would be better measured by the incorporation of antibody monitoring into current functional and pathological assays. AREAS TIMELY FOR DEVELOPING RESEARCH: Research in B-cell targeted therapies to prevent and treat AMR is rapidly growing, which includes monoclonal antibodies against B-cell markers CD20, CD40, CD19, BlyS, etc. It requires extensive clinical research to determine the best approach to inhibit or delete antibody and how to balance the drug efficacy with safety.


Assuntos
Anticorpos/imunologia , Rejeição de Enxerto/imunologia , Transplante/efeitos adversos , Anticorpos Biespecíficos , Anticorpos Monoclonais Humanizados , Anticorpos Monoclonais Murinos , Autoanticorpos/imunologia , Ácidos Borônicos , Bortezomib , Complemento C4b , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/prevenção & controle , Humanos , Isoanticorpos/imunologia , Fragmentos de Peptídeos , Pirazinas , Rituximab
19.
J Urol ; 189(4): 1427-31, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23123549

RESUMO

PURPOSE: We compared the perioperative and postoperative characteristics of prostate PlasmaKinetic™ enucleation and bipolar transurethral resection for large volume benign prostatic hyperplasia. MATERIALS AND METHODS: In this prospective, randomized, controlled trial 80 patients with benign prostatic hyperplasia and a prostate of larger than 70 ml were randomly assigned to prostate bipolar transurethral resection or PlasmaKinetic enucleation. Operative time, resected adenoma weight, changes in hemoglobin, catheterization time and postoperative hospital stay were recorded and compared. Patients were followed 1, 6, 12, 24, 36, 48 and 60 months after surgery. RESULTS: Greater resected prostate weight (mean ± SD 64.2 ± 19.0 vs 50.6 ± 20.0 gm, p = 0.03), less blood loss (mean 0.87 ± 0.42 vs 1.74 ± 0.63 gm, p <0.01), and shorter catheterization time (mean 35.5 ± 5.8 vs 60.1 ± 5.8 hours, p <0.01) and postoperative hospital stay (mean 3 vs 4 days, [corrected] p <0.01) were recorded in the enucleation group than in the resection group. The postoperative improvement in International Prostate Symptom Score, quality of life, maximal flow rate and post-void residual urine volume was similar in the 2 groups at 1, 6, 12 and 24 months but significantly better in the enucleation group at 36, 48 and 60 months. During the 5-year followup no patient in the enucleation group but 2 in the resection group experienced recurrence. CONCLUSIONS: For large volume benign prostatic hyperplasia PlasmaKinetic enucleation of the prostate is associated with less blood loss, shorter hospital stay and catheterization time than bipolar transurethral resection of the prostate. Moreover, PlasmaKinetic enucleation seems to be superior at long-term followup with fewer reoperations necessary.


Assuntos
Eletrocirurgia , Prostatectomia/métodos , Hiperplasia Prostática/cirurgia , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Tamanho do Órgão , Estudos Prospectivos , Próstata/patologia , Hiperplasia Prostática/patologia , Fatores de Tempo , Ressecção Transuretral da Próstata/métodos
20.
Cytotherapy ; 15(8): 893-904, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23751203

RESUMO

The prospective clinical use of multipotent mesenchymal stromal cells (MSCs) holds enormous promise for the treatment of a large number of degenerative and age-related diseases. In particular, autologous MSCs isolated from bone marrow (BM) are considered safe and have been extensively evaluated in clinical trials. Nevertheless, different efficacies have been reported, depending on the health status and age of the donor. In addition, the biological functions of BM-MSCs from patients with various diseases may be impaired. Furthermore, medical treatments such as long-term chemotherapy and immunomodulatory therapy may damage the BM microenvironment and affect the therapeutic potential of MSCs. Therefore, a number of practical problems must be addressed before autologous BM-MSCs can be widely applied with higher efficiency in patients. As such, this review focuses on various factors that directly influence the biological properties of BM-MSCs, and we discuss the possible mechanisms of these alterations.


Assuntos
Doenças Autoimunes/terapia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Doenças Hematológicas/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Doenças Metabólicas/terapia , Células da Medula Óssea , Diferenciação Celular , Proliferação de Células , Senescência Celular , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Humanos , Transplante Autólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA