Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Signal Transduct Target Ther ; 9(1): 79, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38565886

RESUMEN

Fluoropyrimidine-based combination chemotherapy plus targeted therapy is the standard initial treatment for unresectable metastatic colorectal cancer (mCRC), but the prognosis remains poor. This phase 3 trial (ClinicalTrials.gov: NCT03950154) assessed the efficacy and adverse events (AEs) of the combination of PD-1 blockade-activated DC-CIK (PD1-T) cells with XELOX plus bevacizumab as a first-line therapy in patients with mCRC. A total of 202 participants were enrolled and randomly assigned in a 1:1 ratio to receive either first-line XELOX plus bevacizumab (the control group, n = 102) or the same regimen plus autologous PD1-T cell immunotherapy (the immunotherapy group, n = 100) every 21 days for up to 6 cycles, followed by maintenance treatment with capecitabine and bevacizumab. The main endpoint of the trial was progression-free survival (PFS). The median follow-up was 19.5 months. Median PFS was 14.8 months (95% CI, 11.6-18.0) for the immunotherapy group compared with 9.9 months (8.0-11.8) for the control group (hazard ratio [HR], 0.60 [95% CI, 0.40-0.88]; p = 0.009). Median overall survival (OS) was not reached for the immunotherapy group and 25.6 months (95% CI, 18.3-32.8) for the control group (HR, 0.57 [95% CI, 0.33-0.98]; p = 0.043). Grade 3 or higher AEs occurred in 20.0% of patients in the immunotherapy group and 23.5% in the control groups, with no toxicity-associated deaths reported. The addition of PD1-T cells to first-line XELOX plus bevacizumab demonstrates significant clinical improvement of PFS and OS with well tolerability in patients with previously untreated mCRC.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Oxaloacetatos , Humanos , Bevacizumab/uso terapéutico , Capecitabina/uso terapéutico , Oxaliplatino , Neoplasias Colorrectales/tratamiento farmacológico , Fluorouracilo/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Inmunoterapia
2.
Pancreas ; 51(4): 330-337, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35695761

RESUMEN

OBJECTIVES: Embryonic stem cells (ESCs)-derived pancreatic precursor cells have great potential for pancreas repair. Expression of pancreatic duodenal homeobox 1 (Pdx1) in definitive endoderm (DE) cells is the premise that DE cells differentiate into pancreatic cells. To achieve the required number of Pdx1-expressing DE cells for cell transplantation therapy, a valid model must be established. Using this model, researchers investigated how Pdx1 regulates ESC differentiation into pancreatic cells. METHODS: Tet-On inducible lentiviral vector encoding Pdx1 or mock vector was transduced into mouse ESC (ES-E14TG2a). The mouse ESCs were divided into 3 groups: control (ESC), mock vector (Pdx1 - -ESC), and vector encoding Pdx1 (Pdx1 + -ESC). All groups were separately cocultured with the DE cells sorted by immune beads containing CXCR-4 + (C-X-C chemokine receptor type-4) antibody. Doxycycline induced the expression of Pdx1 on the Pdx1 + -ESC cells. The markers of cell differentiation and Notch pathway were examined. RESULTS: Significantly increased expression levels of Ptf1a, CK19, and amylase on day (d) 3 and d7, Neuro-D1 on d10 and d14, Pax6 and insulin on d14, as well as Notch1, Notch2, Hes1, and Hes5 on d3 and thereafter declined on d14 were observed in Pdx1 + -ESC group. CONCLUSIONS: Pdx1 + -ESC could differentiate into pancreatic-like cells with involvement of the Notch pathway.


Asunto(s)
Endodermo , Proteínas de Homeodominio , Células Madre Embrionarias de Ratones , Páncreas , Transactivadores , Animales , Diferenciación Celular , Endodermo/citología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Páncreas/citología , Receptores Notch/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
3.
J Pharmacol Sci ; 139(4): 266-274, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30871870

RESUMEN

Butyrate is widely accepted as a proliferation inhibitor in colon cancer but less thoroughly characterized in the colonic epithelium of objects with type 2 diabetes mellitus. The present study investigated the regulatory effect of butyrate on proliferation, the related molecule high-mobility group box 1 (HMGB1) and the receptor for advanced glycation end products (RAGE) in the colon of db/db type 2 diabetic model mice and non-cancerous NCM460 colon cells. Proliferation and the expression of HMGB1 and RAGE were increased and could be partially reversed by butyrate treatment in the colon of db/db mice, which were consistent in NCM460 cells under a high glucose state. In NCM460 cells, under the normal glucose state, proliferation increased by overexpression of HMGB1. Under a high glucose state, increased expression of HMGB1 was accompanied with a release from cell nuclei into the cytoplasm and extracellular matrix. Down-regulation of HMGB1 could lower the expression of RAGE and attenuate the abnormally increased proliferation. And overexpression of HMGB1 reversed the suppressing effect of butyrate on abnormally increased proliferation. Conclusively, butyrate suppressed the abnormally increased proliferation in colonic epithelial cells under diabetic state by targeting HMGB1.


Asunto(s)
Butiratos/farmacología , Proliferación Celular/efectos de los fármacos , Colon/citología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Células Epiteliales/fisiología , Expresión Génica/efectos de los fármacos , Proteína HMGB1/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Proteína HMGB1/genética , Masculino , Ratones Endogámicos , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo
4.
Exp Cell Res ; 368(1): 101-110, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29689277

RESUMEN

BACKGROUND: Intestinal barrier dysfunctions are related to dysbacteriosis and chronic gut inflammation in type 2 diabetes. Although there is emerging evidence that the chronic gut inflammatory response is stimulated by nucleotide-binding oligomerization domain-like receptors (NLRs), the relationship and precise mechanism between NLRC3 and the colonic epithelial barrier remains largely elusive. METHODS: We investigated the function and mechanism of NLRC3 in the colonic tissues of diabetic mice and colonic epithelial cell lines. The regulatory mechanism between NLRC3, butyrate and tight junctions was elucidated via a transepithelial electrical resistance measurement, transmission electron microscopy, RNA interference and western blotting. RESULTS: In this study, we found that NLRC3 expression was decreased in the colonic tissues of diabetic mice. NLRC3 over-expression ameliorated colonic epithelial barrier integrity and up-regulated tight junction proteins in colonic epithelial cells. Knockdown of TRAF6 diminished NLRC3-induced ZO-1/occludin expression. In addition, we demonstrated that butyrate could stimulate NLRC3 expression in both diabetic mice and colonic epithelial cells. GPR43 on colonic epithelial cells is involved in the activation of NLRC3 induced by butyrate. CONCLUSION: Our findings demonstrated that NLCR3 could ameliorate colonic epithelial barrier integrity in diabetes mellitus in a TRAF6-dependent manner, and NLCR3 was stimulated by butyrate via binding GPR43 on colonic epithelial cells.


Asunto(s)
Butiratos/farmacología , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptores de Superficie Celular/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Ratones Transgénicos , Sustancias Protectoras/farmacología , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Uniones Estrechas/metabolismo
5.
Stem Cell Res Ther ; 8(1): 60, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28279198

RESUMEN

BACKGROUND: Distinctive structures called crypts harbor intestinal epithelial stem cells (IESCs) which generate progenitor and terminally differentiated cells in the intestinal epithelium. Mammalian IESCs and their daughter cells require the participation of DNA methylation and the transcription factor Sox9 for proliferation and differentiation. However, the association between Sox9 and DNA methylation in this process remains elusive. METHODS: The DNA methylation of small intestinal epithelial crypts in db/db mice was detected via combining methylated DNA immunoprecipitation with microarray hybridization. DNA methylation of Sox9 promoter in crypts and IESCs was validated using bisulfite sequence analysis. The target sequence of the transcription factor Sox9 in IESCs was investigated via chromatin immunoprecipitation (ChIP) combined with deep sequencing (ChIP-seq). RESULTS: Increased Sox9 expression is accompanied by the loss of methylation in its promoter in IESCs. Sox9 targets the enhancers of the Wnt signaling pathway-related genes. Sox9 predominantly acts as a transcriptional activator at proximal enhancers of Wnt4, Tab2, Sox4, and Fzd8, but also functions as a potential transcriptional inhibitor at a distant enhancer of Cdk1. Lack of Sox9 transcriptional activation in specific repressors of the Wnt signaling pathway leads to the loss of intrinsic inhibitory action and ultimately produces overactivation of this pathway in db/db mice. CONCLUSIONS: Our study sheds light on the connections among DNA methylation, transcription factor modulation, and Wnt signaling in IESCs in the diabetic state. Hypomethylation in the Sox9 promoter is correlated to increased Sox9 expression in db/db IESCs. Although there is increased expression of Sox9 in db/db IESCs, the loss of Sox9 transcriptional activation in specific repressors of the Wnt signaling pathway might result in abnormalities in this pathway.


Asunto(s)
Metilación de ADN/genética , Diabetes Mellitus/terapia , Factor de Transcripción SOX9/genética , Trasplante de Células Madre , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Regulación del Desarrollo de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos NOD , Regiones Promotoras Genéticas , Células Madre/metabolismo , Activación Transcripcional/genética , Vía de Señalización Wnt/genética
6.
Cell Tissue Res ; 366(2): 341-352, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27299781

RESUMEN

Diabetes mellitus (DM) is a group of metabolic diseases characterised by insulin deficiency/resistance and hyperglycaemia. We previously reported the presence of an impaired tight junction and decreased expression of occludin (Ocln) and zonula occludens-1 (ZO-1) in the intestinal epithelial cells (IECs) of type 1 DM mice, but the exact mechanism remains unclear. In this study, we investigated the role of microRNAs (miRNAs) in impairing the tight junction in IECs of DM mice. Using an integrated comparative miRNA microarray, miR-429 was found to be up-regulated in IECs of type 1 DM mice. Then, miR-429 was confirmed to directly target the 3'-UTR of Ocln, although it did not target ZO-1. Moreover, miR-429 down-regulated the Ocln expression in IEC-6 cells in vitro. Finally, exogenous agomiRNA-429 was shown to down-regulate Ocln and induce intestinal barrier dysfunction in normal mice, while exogenous antagomiRNA-429 up-regulated Ocln in vivo and improved intestinal barrier function in DM mice. In conclusion, increased miR-429 could down-regulate the expression of Ocln by targeting the Ocln 3'-UTR, which impaired intestinal barrier function in DM mice.


Asunto(s)
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Regulación hacia Abajo , Intestinos/patología , MicroARNs/metabolismo , Ocludina/genética , Regiones no Traducidas 3'/genética , Animales , Antagomirs/metabolismo , Secuencia de Bases , Sitios de Unión , Permeabilidad de la Membrana Celular , Regulación hacia Abajo/genética , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ocludina/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas
7.
Mol Cell Biochem ; 413(1-2): 165-78, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26724951

RESUMEN

In previous studies, we have reported the abnormal proliferation and differentiation of intestinal epithelial cells (IECs) in diabetes mellitus (DM) mice. The insulin receptor (IR) and its downstream mitogen-activated protein kinase kinase (MAPKK also known as MEK)/extracellular-regulated protein kinase (ERK) pathway is a classic pathway associated with cell proliferation and differentiation. The purpose of the present study is to investigate the role of the MEK/ERK pathway in abnormal proliferation and differentiation of IECs in DM mice. DM mouse models were induced by intraperitoneal injection of streptozotocin. The expression levels of the IR and its isoforms in IECs of DM mice and in IEC-6 cells were investigated. To ensure that the downstream pathways were monitored, QPCR and Western blotting were performed to detect the expression levels of MEK1/2, ERK1/2, PI3K, and Akt. Moreover, siRNA for IR-A and U0126, a specific inhibitor of MEK, were used to further investigate the relationship between the IR/MEK/ERK pathway and abnormal proliferation and differentiation of IECs in DM mice. In DM mice, excessive proliferation, disturbed differentiation, and a high ratio of IR-A/IR-B were detected in IECs. The expression levels of MEK1, MEK2, and ERK1/2 and their phosphorylated proteins in DM mice were significantly higher than those in the control group (P < 0.05), which could be offset by using siRNA for IR-A. The abnormal proliferation and differentiation of IECs in DM mice were normalized after the in vivo administration of U0126. The abnormal proliferation and differentiation of IECs in DM mice are associated with high IR-A/IR-B ratio and increased IR/MEK/ERK pathway activity.


Asunto(s)
Diabetes Mellitus Experimental/patología , Células Epiteliales/citología , Mucosa Intestinal/citología , Sistema de Señalización de MAP Quinasas , Receptor de Insulina/metabolismo , Animales , Butadienos/administración & dosificación , Butadienos/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Nitrilos/administración & dosificación , Nitrilos/farmacología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor de Insulina/genética , Estreptozocina
8.
Oncotarget ; 7(1): 961-75, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26510906

RESUMEN

Long intergenic noncoding RNAs (lincRNAs) play important roles in regulating the biological functions and underlying molecular mechanisms of colorectal cancer (CRC). Here, we investigated the association of linc-POU3F3 and prognosis in CRC. We demonstrated that linc-POU3F3 was overexpressed in CRC tissues and positively correlated with tumor grade and N stage. Inhibition of linc-POU3F3 resulted in inhibition of cell proliferation and G1 cell cycle arrest, which was mediated by cyclin D1, CDK4, p18, Rb, and phosphorylated Rb. Inhibition of linc-POU3F3 induced apoptosis, and suppressed migration and invasion in LOVO and SW480 cell lines. This inhibition also increased the expressions of epithelial markers and decreased the expressions of mesenchymal markers, thus inhibiting the cancer epithelial-mesenchymal transition. The decreased migration and invasion following linc-POU3F3 knockdown were mediated by an increased BMP signal. Furthermore, autophagy was enhanced by linc-POU3F3 knockdown, suggesting the involvement of autophagy in the induced apoptosis. Collectively, linc-POU3F3 might be crucial in pro-proliferation, anti-apoptosis, and metastasis in LOVO and SW480 cells by regulating the cell cycle, intrinsic apoptosis, BMP signaling and autophagy. Thus, linc-POU3F3 is a potential therapeutic target and novel molecular biomarker for CRC.


Asunto(s)
Apoptosis/genética , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Factores del Dominio POU/genética , ARN Largo no Codificante/genética , Autofagia/genética , Western Blotting , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal/genética , Técnica del Anticuerpo Fluorescente , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Microscopía Electrónica de Transmisión , Factores del Dominio POU/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
9.
Clin Res Hepatol Gastroenterol ; 40(1): 110-23, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26162991

RESUMEN

OBJECTIVE: Recent studies proved that patients with diabetes were at significantly higher risk of developing colorectal cancer. However, the association between diabetes mellitus and the risk of colorectal adenoma remains undefined. Thus we conducted an updated meta-analysis to identify the association between diabetes mellitus and the risk of colorectal neoplasia including adenoma and cancer. METHODS: We conducted a search in databases including Pubmed, Web of Science, EMBASE Databases, Cochrane CENTRAL, Wanfang Data, and CNKI database. Case-control and cohort studies were included. All articles were published before January 2015 and the quality of each study was evaluated by the Newcastle-Ottawa Scale. Odds ratios (ORs) or relative risks (RRs) and its corresponding 95% confidence intervals (CIs) for each study were calculated and summary relative risk estimates with corresponding 95% CIs were generated using the random-effects model. Heterogeneity and publication bias were assessed. RESULTS: Twenty-nine articles including ten case-control studies and nineteen cohort studies were included in this meta-analysis. In a pooled analysis of all studies, diabetes mellitus was associated with increased risk of colorectal neoplasia (RR=1.35, 95% CI=1.28-1.42). The risk increased significantly for both colorectal cancer (RR=1.37, 95% CI=1.30-1.45) and adenoma (RR=1.26, 95% CI=1.11-1.44). Subgroup analyses on study design, gender, geographical region, and type of diabetes mellitus further evidenced these findings. CONCLUSIONS: Diabetes mellitus was associated with an increased risk of colorectal neoplasia. Not only the increased risk of colorectal cancer but also the higher risk of adenoma was identified in patients with diabetes mellitus.


Asunto(s)
Neoplasias Colorrectales/epidemiología , Neoplasias Colorrectales/etiología , Complicaciones de la Diabetes/epidemiología , Humanos , Factores de Riesgo
10.
PLoS One ; 10(7): e0132031, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26135128

RESUMEN

BACKGROUND: The effect of rebamipide on repairing intestinal mucosal damage induced by nonsteroidal anti-inflammatory drugs and its mechanism remain unclear. In this study, we sought to explore the mechanism whereby rebamipide could promote the regeneration of aspirin-induced intestinal mucosal damage. METHODS: BALB/c mice were administered aspirin (200 mg/kg/d) for 5 days to induce acute small intestinal injury (SII). Subsequently, SII mice were treated with rebamipide (320 mg/kg/d) for 5 days. The structure of intestinal barrier was observed with transmission electron microscope, and Zo-1 and occludin expressions were detected. The proliferative index was indicated by the percentage of proliferating cell nuclear antigen positive cells. The prostaglandin E2 (PGE2) levels in the small intestine tissues were measured by an enzyme immunoassay. The mRNA and protein expression levels of cyclooxygenase (COX) and ß-catenin signal were detected in the small intestine using quantitative PCR and Western blot, respectively. RESULTS: COX expression was significantly down-regulated in aspirin induced SII (P < 0.05). In SII mice treated with rebamipide, histopathological findings of aspirin-induced intestinal inflammation were significantly milder and tight junctions between intestinal epithelial cells were improved significantly. The proliferative index increased after rebamipide treatment when compared with that in the control mice. The expressions of COX-2, ß-catenin, and c-myc and the PGE2 concentrations in small intestinal tissues were significantly increased in mice with rebamipide treatments (P < 0.05). CONCLUSION: Rebamipide administration in aspirin-induced SII mice could improve the intestinal barrier structure and promote the regeneration of small intestinal epithelial injury through up-regulating COX-2 expression and the accumulation of ß-catenin.


Asunto(s)
Alanina/análogos & derivados , Antiinflamatorios no Esteroideos/toxicidad , Aspirina/toxicidad , Mucosa Intestinal/efectos de los fármacos , Yeyuno/efectos de los fármacos , Quinolonas/farmacología , Regeneración/efectos de los fármacos , beta Catenina/fisiología , Alanina/farmacología , Animales , Peso Corporal/efectos de los fármacos , División Celular/efectos de los fármacos , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Dinoprostona/biosíntesis , Femenino , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/fisiología , Mucosa Intestinal/ultraestructura , Yeyuno/fisiología , Yeyuno/ultraestructura , Masculino , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos BALB C , Permeabilidad , Proteínas Proto-Oncogénicas c-myc/biosíntesis , ARN Mensajero/biosíntesis , Distribución Aleatoria , Uniones Estrechas/efectos de los fármacos , beta Catenina/biosíntesis , beta Catenina/genética
11.
Cell Biol Int ; 39(9): 995-1006, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25820869

RESUMEN

Treatments for pancreatic injuries have been significantly improved recently, but full recovery of pancreatic function remains difficult. Embryonic stem cells have great potentialities for self-renewal and multiple differentiations. In this study, we explored an approach to induce the differentiation of pancreatic progenitor cells from embryonic stem cells in vitro. Male mouse embryonic stem cells were cultured by the hanging-drop method to form embryoid bodies. The definitive endoderm marked by CXCR4 in embryoid bodies was sorted by magnetic activated cell sorting and subsequently administrated with b-FGF, exendin-4, and cyclopamine to induce the differentiation of putative pancreatic progenitor cells, which was monitored by Pdx1, and Shh expressions. The putative pancreatic progenitor cells were transplanted into female BALB/c mice with pancreatitis induced by L-Arginine. Male donor cells were located by detecting sex-determining region of Y-chromosome DNA. Definitive endoderm cells (CXCR4(+) cells) were sorted from 5-day embryoid bodies. After 3-day administration with b-FGF, exendin-4, and cyclopamine, Pdx1-high/Shh-low cells were differentiated from CXCR4(+) cells. These cells developed into more amylase-secreted cells in vitro and could specifically reside in the damaged pancreas acinar area in mice with acute pancreatitis to enhance the regeneration. The putative pancreatic progenitor cells (Pdx1-high/Shh-low cells) derived from mouse embryonic stem cells through the administration of b-FGF, exendin-4, and cyclopamine on the CXCR4(+) cells in vitro could improve the regeneration of injured pancreatic acini in vivo.


Asunto(s)
Células Madre Embrionarias/citología , Proteínas Hedgehog/biosíntesis , Proteínas de Homeodominio/biosíntesis , Pancreatitis Aguda Necrotizante/terapia , Receptores CXCR4/biosíntesis , Trasplante de Células Madre/métodos , Transactivadores/biosíntesis , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Separación Celular/métodos , Modelos Animales de Enfermedad , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Células Madre Embrionarias/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C
12.
J Diabetes Complications ; 28(5): 689-97, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24930713

RESUMEN

AIMS: ß-Catenin accumulation promotes proliferation. However, the correlation between proliferation of colorectal epithelium and ß-catenin in type 2 diabetes mellitus (DM) patients remains unclear. METHODS: Colorectal epithelium samples from distal ends of colorectal adenocarcinomas without histological aberrances were divided into two groups: DM patients with type 2 DM for more than 1year (n=27) and non-DM patients without hyperglycemia (n=20). Samples from patients without colorectal epithelial disease or hyperglycemia served as a control group (n=6). Proliferative index was calculated as the percentage of proliferating cell nuclear antigen positive cells. Wnt/ß-catenin signaling was assessed immunohistochemically and phosphorylation of ß-catenin was assessed by immunofluorescence. RESULTS: Compared with the non-DM or control group, the proliferative index and expression of lactate dehydrogenase A and Wnt/ß-catenin signaling were significantly higher in the DM group (all p<0.01). The proliferative index correlated positively with ß-catenin expression (Spearman correlation coefficient=0.55; p<0.01). Reduced phosphorylation at serine 33/37 and increased phosphorylation at serine 675 of ß-catenin were detected in the DM group (all p<0.01). CONCLUSIONS: Enhanced proliferation, accompanied by increased aerobic glycolysis, was detected in colorectal epithelium of patients with diabetes. ß-Catenin accumulation with altered phosphorylation correlated with the proliferative changes.


Asunto(s)
Adenoma , Proliferación Celular , Neoplasias Colorrectales , Diabetes Mellitus Tipo 2 , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , beta Catenina/metabolismo , Adenoma/complicaciones , Adenoma/metabolismo , Adenoma/patología , Adulto , Anciano , Estudios de Casos y Controles , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Femenino , Humanos , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Masculino , Persona de Mediana Edad , Antígeno Nuclear de Célula en Proliferación/metabolismo
13.
J Immunol Res ; 2014: 286170, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24868562

RESUMEN

Regulatory T (Treg) cells are potent suppressors that maintain immune homeostasis. Accumulation of Treg can inhibit effective immune responses in cancer patients, leading to tumor development and progression. Despite direct cytotoxicity, several chemotherapeutic drugs have been reported to deplete Treg cells for better prognosis for cancer patients. Treg cells are a heterogenous population with at least three different subsets, nonsuppressive, resting, and activated Treg cells. However, the characteristics of Treg cell subsets in lung cancer patients and how chemotherapy affects Treg cells remain elusive. In this study, we first analyzed Treg cell subsets in peripheral blood samples from 40 nonsmall cell lung cancer (NSCLC) patients and 20 healthy donors. Treg cells, specifically activated Treg cell subset, significantly increased in patients with NSCLC. Compared to nonsuppressive Treg cells, activated Treg cells expressed higher level of CD39 and predominantly produced inhibitory cytokines. In vitro assay showed that docetaxel reduced all three subsets of Treg cells. More importantly, we found docetaxel-based chemotherapy significantly decreased all three Treg subsets after 4 cycles of treatment in 17 NSCLC patients. Taken together, this study revealed dynamic changes of various Treg cell subsets in NSCLC patients before and after chemotherapy, providing activated Treg cells as a potential target for chemotherapy.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Depleción Linfocítica , Linfocitos T Reguladores/efectos de los fármacos , Taxoides/uso terapéutico , Anciano , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Estudios de Casos y Controles , Linaje de la Célula/inmunología , Docetaxel , Femenino , Humanos , Inmunofenotipificación , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Linfocitos T Reguladores/clasificación , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
14.
Biomed Pharmacother ; 68(5): 551-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24855035

RESUMEN

Cytokine-induced killer (CIK) cells have achieved therapeutic benefit in treatment of solid tumors in clinic. However, some patients show no response after CIK treatment. Animal assays have shown that successful infiltration of CIK cells to the tumor sites could affect the outcome. Chemokines play important roles in lymphocyte trafficking. Understanding the molecular mechanism of chemokines in the process of CIK cell homing is important for further modification of CIK therapy. In this study, we investigated the spectrum of chemokine ligands in the colorectal cancer sites and observed that chemokine ligands CCL20 and CXCL10 were overexpressed in the CRC tumor tissues compared with adjacent tissues. Although the corresponding receptors CCR6 and CXCR3 increased on CIK cells compared with PBMCs, their expression on CIK cells derived from CRC patients had lower levels than healthy donors, which might be a limited factor for autologous-CIK cells trafficking to tumor site. Importantly, stimulation with chemokines CCL20 and CXCL10 promotes the expression levels of CCR6 and CXCR3 on CIK cells, thus augmenting the relative migration of CIK cells in vitro. Our results suggest that modification of surface chemokine receptors may enhance the homing ability of CIK cells for better therapeutic achievements.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células Asesinas Inducidas por Citocinas/metabolismo , Receptores de Quimiocina/metabolismo , Anciano , Movimiento Celular , Quimiocinas/metabolismo , Femenino , Humanos , Ligandos , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/metabolismo
15.
PLoS One ; 9(4): e94208, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24710620

RESUMEN

BACKGROUND: Although the chemopreventive effect of 5-aminosalicylates on patients with ulcerative colitis has been extensively studied, the results remain controversial. This updated review included more recent studies and evaluated the effectiveness of 5-aminosalicylates use on colorectal neoplasia prevention in patients with ulcerative colitis. METHODS: Up to July 2013, we searched Medline, Embase, Web of Science, Cochrane CENTRAL, and SinoMed of China for all relevant observational studies (case-control and cohort) about the effect of 5-aminosalicylates on the risk of colorectal neoplasia among patients with ulcerative colitis. The Newcastle-Ottawa Scale was used to assess the quality of studies. Adjusted odds ratios (ORs) were extracted from each study. A random-effects model was used to generate pooled ORs and 95% confidence intervals (95%CI). Publication bias and heterogeneity were assessed. RESULTS: Seventeen studies containing 1,508 cases of colorectal neoplasia and a total of 20,193 subjects published from 1994 to 2012 were analyzed. 5-aminosalicylates use was associated with a reduced risk of colorectal neoplasia in patients with ulcerative colitis (OR 0.63; 95%CI 0.48-0.84). Pooled OR of a higher average daily dose of 5-aminosalicylates (sulfasalazine ≥ 2.0 g/d, mesalamine ≥ 1.2 g/d) was 0.51 [0.35-0.75]. Pooled OR of 5-aminosalicylates use in patients with extensive ulcerative colitis was 1.00 [0.53-1.89]. CONCLUSION: Our pooled results indicated that 5-aminosalicylates use was associated with a reduced risk of colorectal neoplasia in patients with ulcerative colitis, especially in the cases with a higher average daily dose of 5-aminosalicylates use. However, the chemopreventive benefit of 5-aminosalicylates use in patients with extensive ulcerative colitis was limited.


Asunto(s)
Colitis Ulcerosa/complicaciones , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/prevención & control , Mesalamina/farmacología , Humanos , Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA