Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Exp Cell Res ; 408(2): 112862, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34626585

RESUMEN

Macrophage receptor with collagenous structure (MARCO) is a member of the class A scavenger receptor family which is expressed on the cell surface of macrophages. It is well known that MARCO avidly binds to unopsonized pathogens, leading to its ingestion by macrophages. However, the role of MARCO in the recognition and phagocytosis of tumor cells by macrophages remains poorly understood. In this study, we used lentiviral technology to knockdown and overexpress MARCO and investigated the ability of macrophages to phagocytose tumor cells. Our results showed that MARCO expression was correlated with the ability of macrophages to carry on phagocytosis. MARCO knockdown led to significant decreases in the number of engulfment pseudopodia of macrophages and inhibition of the phagocytosis of tumor cells. On the other hand, MARCO overexpression elevated activity of SYK, PI3K and Rac1 in macrophages, which led to changes in macrophage morphology and enhanced phagocytosis by promoting formation of stress fibers and pseudopodia. By Co-IP analysis we showed that MARCO directly binds to the ß5 integrin of SL4 tumor cells. In summary, these results demonstrated the important role for MARCO in demonstrated tumor cells uptake and clearance by macrophages.


Asunto(s)
Cadenas beta de Integrinas/genética , Neoplasias/genética , Fagocitosis/genética , Receptores Inmunológicos/genética , Receptores Depuradores/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Fosfatidilinositol 3-Quinasas/genética , Quinasa Syk/genética , Proteína de Unión al GTP rac1/genética
2.
Int J Mol Sci ; 19(5)2018 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-29786668

RESUMEN

Mucinous colorectal adenocarcinoma (MCA) patients often a show high risk of malignant potential and a poorer survival rate. Given that the pathological feature and oncobiological characteristics of MCA are correlated with its abundant extracellular mucin2 (MUC2), we paid interest toward investigating the key factor that promotes MUC2 production exposure to highly-activated stem cell factor (SCF)/c-KIT signaling, which we believed to contribute to MCA formation. Long-term azoxymethane and dextran sodium sulfate treatment successfully induced MCA only in wild-type (WT) mice at week 37 and 43, while all c-kit loss-of-function mutant mice (Wadsm/m) developed non-MCA. Significantly, MUC2 and its key transcriptional factor Atonal homologue 1 (Atoh1) were remarkably expressed in MCA mice compared with non-MCA mice. Atoh1 was significantly elevated in colorectal cancer (CRC) cells stimulated by exogenous SCF or overexpressing c-KIT in vitro, while decreased by the blockage of SCF/c-KIT signaling with Imatinib. Furthermore, the maintained Atoh1 protein level was due to the inactive glycogen synthase kinase 3ß (p-GSK3ß) by virtue of the activated SCF/c-KIT-Protein Kinase B (AKT) signaling. Similar results were obtained from the ONCOMINE database and CRC patients. In conclusion, we suggested that SCF/c-KIT signaling promoted MUC2 production and MCA tumorigenesis by maintaining Atoh1 expression. Therefore, targeting the related key molecules might be beneficial for treating MCA patients.


Asunto(s)
Adenocarcinoma Mucinoso/metabolismo , Neoplasias Colorrectales/metabolismo , Mucina 2/metabolismo , Transducción de Señal , Factor de Células Madre/metabolismo , Adenocarcinoma Mucinoso/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Mucina 2/genética , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo
3.
J Cell Mol Med ; 21(9): 1881-1892, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28580775

RESUMEN

Gastrointestinal motility disorders (GMDs) are attributed to loss of interstitial cells of Cajal (ICC), whose survival and function are deeply dependent on the activation of KIT/SCF signalling. Based on the facts that gastrointestinal distention is common in GMD patients and SCF produced by smooth muscle cells (SMCs) is usually decreased before ICC loss, we considered a possible contribution of persistent gastrointestinal distention/stretch to SCF deficiency. In this study, chronic colonic distention mouse model, diabetic gastrointestinal paresis mouse model, cultured mouse colonic SMCs and colon specimens from Hirschsprung's disease patients were used. The results showed that SCF was clearly decreased in distent colon of mice and patients, and microRNA array and real-time PCR indicated a concomitant increase of miR-34c in distent colon. A negative regulation of miR-34c on SCF expression was confirmed by luciferase reporter assays together with knock-down and overexpression of miR-34c in cultured colonic SMCs. Using EMSA and ChIP assays, we further consolidated that in response to persistent stretch, the transcription factor AP-1/c-Jun was highly activated in colonic SMCs and significantly promoted miR-34c transcription by binding to miR-34c promoter. Knock-down or overexpression of AP-1/c-Jun in cultured colonic SMCs leads to down- or up-regulation of miR-34c, respectively. In addition, the activation of AP-1/c-Jun was through ERK1/2 signalling provoked by Ca2+ overload in colonic SMCs that were subject to persistent stretch. In conclusion, our data demonstrated that persistent distention/stretch on colonic SMCs could suppress SCF production probably through Ca2+ -ERK-AP-1-miR-34c deregulation, resulting in ICC loss or impairment and GMD progress.


Asunto(s)
Colon/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Intersticiales de Cajal/patología , MicroARNs/metabolismo , Factor de Células Madre/genética , Factor de Transcripción AP-1/metabolismo , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Activación Enzimática , Humanos , Células Intersticiales de Cajal/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Modelos Biológicos , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Células Madre/metabolismo , Transcripción Genética
4.
Biochem Biophys Res Commun ; 486(4): 1062-1068, 2017 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-28373072

RESUMEN

It was reported that Src-mediated and RTK-dependent accumulation of key transcription factor, ETV4, which played an important role in the migration of embryonic cells and tumor cells, were regulated by their common downstream MAPK molecules. However, the detailed mechanism was not completely clear. In the present study, we revealed that ETV4 protein was significantly enhanced by ERK kinase activation in the colorectal cancer (CRC) patients and mouse models as well as in the CRC cell lines. It was further confirmed that the activation of ERK kinase led to the phosphorylation of ETV4 at Ser73 and the ETV4 phosphorylation could block its binding to COP1, thereby stabilized ETV4 via avoiding its ubiquitination degradation. In addition, this effect was not due to altering an E3 ubiquitin ligase, COP1 amount or p-COP1/COP1 ratio. Our results will help understand the mechanism of ETV4 overexpression in CRC patients and provide a clue to search new therapeutic target to treat the related tumors in clinical practice.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Neoplasias Colorrectales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ubiquitinación , Ubiquitinas/metabolismo , Animales , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Fosforilación , Unión Proteica , Células Tumorales Cultivadas , Proteínas Ubiquitinadas/metabolismo
5.
BMC Cancer ; 15: 969, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26674205

RESUMEN

BACKGROUND: Silence of the tumor suppressor miR-34c is implicated in the development of colorectal cancer (CRC). For the past few years, Resveratrol (Res) has been introduced to oncotherapies alone or with traditional chemotherapeutic drugs. However, the study of molecular mechanism involved in the anti-CRC effect of Res is still ongoing. METHODS: The anti-CRC effect of Res alone or with Oxaliplatin (Oxa) was determined by cell viability assay, soft agar colony formation assay, flow cytometry and real-time cellular analyzer in HT-29 (p53+) and HCT-116 (p53-) CRC cell lines. Expressions of miR-34c and its targets were detected by qPCR and/or western blot. To evaluate the role of miR-34c in anti-CRC effect by Res alone or with Oxa, miR-34c was up or down-regulated by lentiviral mediation or specific inhibitor, respectively. To investigate how miR-34c was increased by Res, the methylation status of miR-34c promoter was detected by MSP. The tumor bearing mouse model was established by subcutaneous injection of HCT-116 cells to assess anti-CRC effect of Res alone or with Oxa in vivo. IL-6 and TNF-α in xenografts were detected by ELISA. RESULTS: Res inhibited cell viability, proliferation, migration and invasion as well as promoted apoptosis both in HT-29 and HCT-116 CRC cells. The anti-CRC effect of Res was partially but specifically through up-regulating miR-34c which further knocked down its target KITLG; and the effect was enhanced in the presence of p53 probably through inactivating PI3K/Akt pathway. Besides, Res sensitized CRC cells to Oxa in a miR-34c dependent manner. The xenograft experiments showed that exposure to Res or Oxa suppressed tumor growth; and the efficacy was evidently augmented by the co-treatment of Res and Oxa. Likewise, miR-34c level was elevated in xenografts of Res-treated mice while the KITLG was decreased. Finally, Res clearly reduced IL-6 in xenografts. CONCLUSION: Res suppressed CRC by specifically activating miR-34c-KITLG in vitro and in vivo; and the effect was strengthened in the presence of p53. Besides, Res exerted a synergistic effect with Oxa in a miR-34c dependent manner. We also suggested that Res-increased miR-34c could interfere IL-6-triggered CRC progression.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/genética , MicroARNs/metabolismo , Estilbenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Células HT29 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/efectos de los fármacos , Compuestos Organoplatinos/farmacología , Oxaliplatino , Reacción en Cadena en Tiempo Real de la Polimerasa , Resveratrol , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Neurosci Res ; 91(7): 978-86, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23633297

RESUMEN

Dopaminergic (DA) neuron therapy has been established as a new clinical tool for treating Parkinson's disease (PD). Prior to cell transplantation, there are two primary issues that must be resolved: one is the appropriate seed cell origin, and the other is the efficient inducing technique. In the present study, human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) were used as the available seed cells, and conditioned medium from human amniotic epithelial cells (ACM) was used as the inducing reagent. Results showed that the proportion of DA neuron-like cells from hUCB-MSCs was significantly increased after cultured in ACM, suggested by the upregulation of DAT, TH, Nurr1, and Pitx3. To identify the process by which ACM induces DA neuron differentiation, we pretreated hUCB-MSCs with k252a, the Trk receptor inhibitor of brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), and found that the proportion of DA neuron-like cells was significantly decreased compared with ACM-treated hUCB-MSCs, suggesting that NGF and BDNF in ACM were involved in the differentiation process. However, we could not rule out the involvement of other unidentified factors in the ACM, because ACM + k252a treatment does not fully block DA neuron-like cell differentiation compared with control. The transplantation of ACM-induced hUCB-MSCs could ameliorate behavioral deficits in PD rats, which may be associated with the survival of engrafted DA neuron-like cells. In conclusion, we propose that hUCB-MSCs are a good source of DA neuron-like cells and that ACM is a potential inducer to obtain DA neuron-like cells from hUCB-MSCs in vitro for an ethical and legal cell therapy for PD.


Asunto(s)
Amnios/citología , Diferenciación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Células Epiteliales/química , Sangre Fetal/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Análisis de Varianza , Animales , Apomorfina , Factor Neurotrófico Derivado del Encéfalo/farmacología , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Ensayo de Inmunoadsorción Enzimática , Feto , Citometría de Flujo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/cirugía , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor trkA/genética , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Conducta Estereotipada/efectos de los fármacos , Conducta Estereotipada/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
7.
J Crohns Colitis ; 16(4): 656-667, 2022 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-34628497

RESUMEN

BACKGROUND AND AIMS: Disruption of the intestinal barrier of the digestive tract is a common pathophysiological change in the elderly, which may partly contribute to gut dysfunction and inflammatory bowel disease [IBD]. This study aimed to discover new interactive epigenetic regulation patterns involved in intestinal barrier dysfunction and colitis in elderly populations. METHODS: Intestinal barrier function and structure were evaluated in naturally ageing mice and elderly people. High-throughput analysis was performed on colonic tissues from humans and mice. The synergistic roles of miR-1-3p and miR-124-3p were identified using microRNA mimic/agomirs. Related genes were examined in biopsies of old IBD patients. RESULTS: A defective mucus barrier was observed before mucosal microstructural damage during ageing. Elevated miR-1-3p expression in the colons of older individuals impaired the mucus barrier by directly targeting T-synthase, similarly to the mechanism of miR-124-3p, which we reported previously. Importantly, the synergistic effect of a half dose of each microRNA supplement on T-synthase and CDK4/6 was stronger than that of a full dose of miR-1-3p or miR-124-3p alone, and mice co-treated with two microRNAs showed greater susceptibility to chemical-induced colitis than mice treated with either microRNA alone. These two microRNAs were up-expressed in old IBD patients. CONCLUSIONS: The slight increases in miR-1-3p and miR-124-3p expression with ageing may be important contributors to the breakdown of intestinal homeostasis by targeting divergent genes in different cells. These data reveal the potential ability of multiple microRNAs to exert synergistic effects to damage the intestinal barrier and promote inflammatory bowel disease development in elderly populations.


Asunto(s)
Envejecimiento , Colitis , Enfermedades Inflamatorias del Intestino , MicroARNs , Anciano , Envejecimiento/genética , Animales , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Epigénesis Genética , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo
8.
Aging Cell ; 19(11): e13252, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33040455

RESUMEN

The risk of colitis and colorectal cancer increases markedly throughout adult life, endangering the health and lives of elderly individuals. Previous studies have proposed that bacterial translocation and infection are the main risk factors for these diseases. Therefore, in the present study, we aimed to identify the underlying mechanism by focusing on the mucus barrier function and mucin-type O-glycosylation. We evaluated alterations in the colon mucus layer in 2-, 16-, and 24-month-old mice and aged humans. Aged colons showed defective intestinal mucosal barrier and changed mucus properties. The miR-124-3p expression level was significantly increased in the aged distal colonic mucosa, which was accompanied by an increase in pathogens and bacterial translocation. Meanwhile, T-synthase, the rate-limiting enzyme in O-glycosylation, displayed an age-related decline in protein expression. Further experiments indicated that miR-124-3p modulated O-glycosylation by directly targeting T-synthase. Moreover, young mice overexpressing miR-124-3p exhibited abnormal glycosylation, early-onset, and more severe colitis. These data suggest that miR-124-3p predisposes to senile colitis by reducing T-synthase, and the miR-124-3p/T-synthase/O-glycans axis plays an essential role in maintaining the physiochemical properties of colonic mucus and intestinal homeostasis.


Asunto(s)
Colitis/metabolismo , Galactosiltransferasas/metabolismo , MicroARNs/metabolismo , Moco/metabolismo , Factores de Edad , Colitis/genética , Colitis/patología , Colon/patología , Femenino , Humanos , Mucosa Intestinal/metabolismo , Masculino
9.
Histol Histopathol ; 34(2): 191-200, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30211433

RESUMEN

The alteration of intestinal mucosal barrier is considered to be the central pathophysiological process in response to gastrointestinal infections, and mucosal microstructural damage is a major factor for enhancing epithelial permeability in persistent bacterial infections. However, the mechanism involved in hyperpermeability in the early stage of acute bacterial infections is not fully understood. In the present study, fluorescein isothiocyanate-dextran across and transepithelial resistance measured in Ussing chambers were used to assess the intestinal paracellular permeability. Mast cell activation was evaluated by western blotting for the presence of tryptase released from mast cells. Serum levels of interleukin-6 were evaluated using enzyme-linked immunosorbent assay. Our results indicated that mast cells played a pivotal role in colonic mucosal hyperpermeability in wild type mice treated with lipopolysaccharide (LPS) for 2 h. And the effect of LPS was mainly dependent on mast cell degranulation, while no change in permeability was observed in the mast cell-deficient mice (Wads⁻/⁻) after LPS administration. No obvious changes of the mucosal structure including histomorphological architecture and expression of intercellular junction proteins were obtained in either wild type or Wads⁻/⁻ mice after LPS stimulation by hematoxylin and eosin staining, immunofluorescence staining and western blot analysis. Furthermore, the self-renewal of intestinal epithelia, detected by using proliferation marker 5'-bromo-2'-deoxyuridine, was not involved in increased permeability. Collectively, activation of mast cells induced by LPS mediated intestinal hyperpermeability in the initial stage, and played a crucial role in barrier dysfunction rather than mucosal microstructural damage in acute enterogenous bacterial infection.


Asunto(s)
Uniones Intercelulares/efectos de los fármacos , Mucosa Intestinal/metabolismo , Lipopolisacáridos/toxicidad , Mastocitos/inmunología , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Colon , Modelos Animales de Enfermedad , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos C57BL , Permeabilidad
10.
Biochem Biophys Res Commun ; 373(1): 151-4, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18555005

RESUMEN

Low expression of the oxidative stress sensor Keap1 is thought to be involved in carcinogenesis. However, the mechanisms responsible for inactivation of the Keap1 gene remain unknown. We investigated Keap1 expression using RT-PCR and found that it was downregulated in lung cancer cell lines and tissues when compared with a normal bronchial epithelial cell line. Treatment with 5-Aza-2'-deoxycytidine restored Keap1 expression in lung cancer cell lines, indicating the silencing mechanism to be promoter methylation. Moreover, we evaluated cytosine methylation in the Keap1 promoter and demonstrated that the P1 region, including 12 CpG sites, was highly methylated in lung cancer cells and tissues, but not in normal cells. Importantly, we found evidence that three specific CpG sites (the 3rd, 6th, and 10th CpGs of P1) might be binding sites for proteins that regulate Keap1 expression. Thus, our results suggest for the first time that Keap1 expression is regulated by an epigenetic mechanism in lung cancer.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pulmonares/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Sitios de Unión , Línea Celular Tumoral , Islas de CpG , Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/antagonistas & inhibidores , Decitabina , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Humanos , Proteína 1 Asociada A ECH Tipo Kelch
11.
J Neuroimmunol ; 197(2): 99-109, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18534691

RESUMEN

One concern in the use of transplantation of non-hematopoietic stem cells from human umbilical cord blood (CB-nHSCs) is the possibility of rejection by the host's immune system. This study shows that both CB-nHSCs and their progenies after passaging, neuronal differentiation or IFN-gamma treatment have no significant effects on proliferation of xenogenic T lymphocytes. CB-nHSCs transplanted into the striatum of SD rat are shown to induce a lower level of CD4 and CD8 expression in the brain and in the peripheral blood and to survive better in the brain than SH-SY5Y cells. The results indicate that both undifferentiated and differentiated CB-nHSCs all have weak immunogenicity.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/fisiología , Sangre Fetal/citología , Expresión Génica/fisiología , Células Madre/fisiología , Análisis de Varianza , Diferenciación Celular/efectos de los fármacos , Línea Celular Transformada , Proliferación Celular , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/trasplante , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Citometría de Flujo/métodos , Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/farmacología , Proteínas de Filamentos Intermediarios/metabolismo , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/fisiología , Proteínas del Tejido Nervioso/metabolismo , Nestina , Neuronas/metabolismo , Trasplante de Células Madre/métodos , Factores de Tiempo , Tubulina (Proteína)/metabolismo
12.
Am J Cancer Res ; 8(6): 1064-1073, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30034943

RESUMEN

Mucinous colorectal adenocarcinoma (MCA) is characterized by a great mount of extracellular mucus fundamentally composed of Mucin2 (MUC2) which is significantly correlated with the high malignancy and strong invasive ability of MCA. However, rare is known about the underlying mechanism of the mucus accumulation in MCA. Our latest study demonstrated that SCF/c-KIT signaling was highly activated in MCA patients and mouse model, which up-regulated MUC2 transcription. In the present study, we paid a special interest in whether and how SCF/c-KIT signaling promoted mucus secretion by using wild-type (WT) C57BL mice and their littermates who harbor mutational c-kit gene (Wadsm/m), clinical colorectal cancer (CRC) samples, as well as human CRC cell lines. Our results clearly showed that the inner mucus layer of colon was thinner and the intracellular mucin residual was more in Wadsm/m mice than those in WT mice by Alcian blue and PAS staining, suggesting that the mucus secretion process was crippled when SCF/c-KIT signaling was hypo-activated. Inhibiting SCF/c-KIT signaling by Imatinib also resulted in weakened mucus secretion in WT mice. Intraperitoneal administration of MANS which competitively inhibits the activity of the vesicular transport protein MARCKS efficiently reduced mucus secretion in colonic goblet cells of WT mice. Significantly, phosphorylated MARCKS (p-MARCKS) was overtly decreased in colonic mucosa of Wadsm/m mice compared with WT mice, indicating that SCF/c-KIT signaling-regulated mucus secretion was probably mediated by MARCKS activation. Similar results were obtained in MCA patients and mouse model. Moreover, SCF/c-KIT signaling was activated or inhibited in HT-29 and LS174T CRC cells, which potently increased or decreased MARCKS activity, respectively. Finally, we found that PKCδ, a known kinase for MARCKS, was activated in WT and MCA mice along with MARCKS. Inhibition or activation of SCF/c-KIT signaling resulted in decreased or increased PKCδ activity respectively in vitro. In conclusion, we demonstrated that SCF/c-KIT signaling can promote the mucus secretion by activating PKCδ-MARCKS, which provided a new insight into understanding the mechanism of mucus secretion of goblet cells and MCA development.

13.
Mol Cancer Res ; 16(3): 508-516, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29330291

RESUMEN

Low expression of the tumor suppressor Kelch-like ECH-associated protein 1 (KEAP1) in non-small cell lung cancer (NSCLC) often results in higher malignant biological behavior and poor prognosis; however, the underlying mechanism remains unclear. The present study demonstrates that overexpression of Keap1 significantly suppresses migration and invasion of three different lung cancer cells (A549, H460, and H1299). Highly expressed Keap1, compared with the control, promotes formation of multiple stress fibers with larger mature focal adhesion complexes in the cytoplasm where only fine focal adhesions were observed in the membrane under control conditions. RhoA activity significantly increased when Keap1 was overexpressed, whereas Myosin 9b expression was reduced but could be rescued by proteasome inhibition. Noticeably, mouse tumor xenografts with Keap1 overexpression were smaller in size and less metastatic relative to the control group. Taken together, these results demonstrate that Keap1 stabilizes F-actin cytoskeleton structures and inhibits focal adhesion turnover, thereby restraining the migration and invasion of NSCLC. Therefore, increasing Keap1 or targeting its downstream molecules might provide potential therapeutic benefits for the treatment of patients with NSCLC.Implications: This study provides mechanistic insight on the metastatic process in NSCLC and suggests that Keap1 and its downstream molecules may be valuable drug targets for NSCLC patients. Mol Cancer Res; 16(3); 508-16. ©2018 AACR.


Asunto(s)
Actinas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Adhesiones Focales/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Neoplasias Pulmonares/metabolismo , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Xenoinjertos , Humanos , Inmunohistoquímica , Proteína 1 Asociada A ECH Tipo Kelch/biosíntesis , Proteína 1 Asociada A ECH Tipo Kelch/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica
14.
Aging (Albany NY) ; 10(12): 3851-3865, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30530917

RESUMEN

Aging is a significant risk factor for gastrointestinal dysmotility, but aging-associated differences between different organs and the exact time to start degenerating have remained obscure. Here we evaluated alterations of interstitial cells of Cajal, enteric neurons and connexin43 expression in the stomach, jejunum and colon in 2-, 12-, 16-, 20- and 24-month-old mice, as well as in aged human colon. Interstitial cells of Cajal, cholinergic and nitrergic neurons within the whole digestive tract were reduced over time, but their loss first appeared in stomach, then in intestine, helping to understand that gastric function was first impaired during aging. The decrease of connexin43 expression occurred before interstitial cells of Cajal and neurons loss, suggesting that connexin43 might be the major target influenced during senescence. Furthermore, changes in expressions of pro-inflammatory cytokines (tumour necrosis factor-α, interleukin-1ß, interleukin-6) and apoptosis-related proteins (B-cell lymphoma-2, caspase-3) which indicated "inflammaging", might contribute to the loss of enteric neurons and interstitial cells of Cajal in aged gastrointestinal tract. Our results provide possible therapeutic time window for beneficial intervention for geriatric patients with gastrointestinal motility disorders.


Asunto(s)
Envejecimiento/fisiología , Conexina 43/metabolismo , Sistema Nervioso Entérico/fisiología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/inervación , Células Intersticiales de Cajal/fisiología , Animales , Conexina 43/genética , Citocinas/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología
15.
Biosci Rep ; 37(5)2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-28931726

RESUMEN

Diabetic gastroparesis is a common complication of diabetes mellitus (DM) that is characterized by decreased serum insulin and insulin-like growth factor-1 (IGF-1). Despite the fact that insulin treatment not glycemic control potently accelerated gastric emptying in type 1 DM patients, the role of insulin/InsR and IGF-1/IGF-1R signaling in diabetic gastroparesis remains incompletely elucidated. In the present study, type 1 DM mice were established and treated with insulin or Voglibose for 8 weeks. The gastric emptying was delayed from DM week 4 when the gastric InsR and IGF-1R were declined. Meanwhile, the gastric choline acetyltransferase (ChAT) was significantly reduced and the myenteric cholinergic neurones and their fibers were significantly diminished. The production of stem cell factor (SCF) was dramatically repressed in the gastric smooth muscles in DM week 6. TWereafter, interstitial cells of Cajal (ICC) were clearly lost and their networks were impaired in DM week 8. Significantly, compared with Voglibose, an 8-week treatment with insulin more efficiently delayed diabetic gastroparesis development by protecting the myenteric cholinergic neurones and ICC. In conclusion, diabetic gastroparesis was an aggressive process due to the successive damages of myenteric cholinergic neurones and ICC by impairing the insulin/InsR and IGF-1/IGF-1R signaling. Insulin therapy in the early stage may delay diabetic gastroparesis.


Asunto(s)
Antígenos CD/genética , Complicaciones de la Diabetes/genética , Gastroparesia/genética , Factor I del Crecimiento Similar a la Insulina/genética , Insulina/administración & dosificación , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Animales , Antígenos CD/metabolismo , Colina O-Acetiltransferasa/genética , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/patología , Complicaciones de la Diabetes/tratamiento farmacológico , Complicaciones de la Diabetes/metabolismo , Complicaciones de la Diabetes/patología , Modelos Animales de Enfermedad , Gastroparesia/tratamiento farmacológico , Gastroparesia/metabolismo , Gastroparesia/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inositol/administración & dosificación , Inositol/análogos & derivados , Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Intersticiales de Cajal/metabolismo , Células Intersticiales de Cajal/patología , Ratones , Ratones Endogámicos NOD/genética , Ratones Endogámicos NOD/metabolismo , Músculo Liso/metabolismo , Músculo Liso/patología , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Factor de Células Madre/genética
16.
J Exp Clin Cancer Res ; 35: 75, 2016 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-27154193

RESUMEN

BACKGROUND: Recent evidence has proven that long noncoding RNAs (lncRNAs) play important roles in cancer biology, while few lncRNAs have been characterized in NSCLC. Here, we characterized a novel lncRNA, SBF2 antisense RNA 1 (SBF2-AS1), in non-small cell lung cancer (NSCLC). METHODS: Quantitative real-time PCR was used to quantify SBF2-AS1 expression in NSCLC tissues and cell lines. The correlation of SBF2-AS1 expression with clinicopathologic features was analyzed in a cohort NSCLC patient. Loss of function and gain of function studies were performed to determine the effects of SBF2-AS1 on proliferation and metastasis of NSCLC cells. RNA immunoprecipitation and chromosome immunoprecipitation assay was performed to confirm the interaction between SBF2-AS1 with protein and chromosome. RESULTS: We confirmed that SBF2-AS1 was significantly upregulated in NSCLC compared with corresponding non-tumor tissues, and a high expression level of SBF2-AS1 was correlated with lymph node metastasis and advanced TNM stage. Using siRNAs specifically targeting SBF2-AS1 and plasmid vector, we successfully silenced and overexpressed SBF2-AS1 in NSCCLC cell lines and investigated its biological function both in vitro and in vivo. After the silencing of SBF2-AS1, the metastasis of NSCLC cells was significantly inhibited, the silencing of SBF2-AS1 decreased the proliferation of NSCLC cells, and the cell cycle was arrested at the G1 phase; while overexpression promoted proliferation ability. Xenograft tumor models revealed that the silencing of SBF2-AS1 inhibited tumor growth in vivo. We speculated that SBF2-AS1 might negatively regulate P21. RNA immunoprecipitation discovered that SBF2-AS2 could bind with a core component of polycomb repressive complex2, SUZ12. Additionally chromatin immunoprecipitation assay demonstrated that, after silencing SBF2-AS1, the enrichment of SUZ12 and trimethylation of histone 3 lysine 27 decreased at the promoter region of P21. CONCLUSIONS: We demonstrated that SBF2-AS1 is upregulated in NSCLC and promotes proliferation of NSCLC tumor cells. SBF2-AS1 may serve as a novel biomarker and potential therapeutic target for NSCLC patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , ARN Largo no Codificante/genética , Regulación hacia Arriba , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Trasplante de Neoplasias
17.
Biosci Rep ; 36(1): e00293, 2015 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-26704889

RESUMEN

Tumour suppressor miR-34c deficiency resulted from hyper-methylation in its promoter is believed to be one of the main causes of colorectal cancer (CRC). Till date, miR-34c has been validated as a direct target of p53; but previous evidence suggested other transcription factor(s) must be involved in miR-34c transcription. In the present study, we in the first place identified a core promoter region (-1118 to -883 bp) of pre-miR-34c which was embedded within a hyper-methylated CpG island. Secondly, E2F1 promoted miR-34c transcription by physical interaction with the miR-34c promoter at site -897 to -889 bp. The transcriptional activating effect of E2F1 on miR-34c was in a p53 independent manner but profoundly promoted in the presence of p53 with exposure to 5-aza-2'-deoxycytidine (DAC). Thirdly, stem cell factor (SCF), a miR-34c target, was specifically reduced upon an introduction of E2F1 which lead to suppression of CRC cell proliferation. The E2F1-suppressed cell proliferation was partially abrogated by additional miR-34c inhibitor, indicating that the anti-proliferation effect of E2F1 was probably through activating miR-34c-SCF axis. Finally, SCF/KIT signalling increased E2F1 production by reducing its proteosomal degradation dependent on PI3K/Akt-GSK3ß pathway. In conclusion, our results suggested the existence of E2F1-miR-34c-SCF negative feedback loop which was interrupted by the hyper-methylation of miR-34c promoter in CRC cells and increased cell proliferation.


Asunto(s)
Proliferación Celular , Neoplasias Colorrectales/metabolismo , Metilación de ADN , ADN de Neoplasias/metabolismo , Factor de Transcripción E2F1/metabolismo , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Neoplásico/metabolismo , Elementos de Respuesta , Transducción de Señal , Factor de Células Madre/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/genética , ADN de Neoplasias/genética , Factor de Transcripción E2F1/genética , Humanos , MicroARNs/genética , Proteínas de Neoplasias/genética , ARN Neoplásico/genética , Factor de Células Madre/genética
18.
Oncotarget ; 6(29): 27037-48, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26356816

RESUMEN

It was reported that the receptor tyrosine kinase (RTK) family often highly expressed in several mucinous carcinomas. In the present study, we established a murine model of colorectal mucinous adenocardinoma (CRMAC) by treating C57 mice [both wild type (WT) and loss-of-function c-kit mutant type (Wads-/-)] with AOM+DSS for 37 weeks and found that c-kit, a member of RTK family, clearly enhanced the tumor cell proliferation by decreasing p53 and increasing cyclin D1 through AKT pathway. Significantly, c-kit strongly promoted tumor cell invasiveness by increasing ETV4, which induced MMP7 expression and epithelial-mesenchymal transition (EMT) via ERK pathway. In vitro up- or down-regulating c-kit activation in human colorectal cancer HCT-116 cells further consolidated these results. In conclusion, our data suggested that the c-kit signaling obviously promoted proliferation and invasion of CRMAC. Therefore, targeting the c-kit signaling and its downstream molecules might provide the potential strategies for treatment of patients suffering from CRMAC in the future.


Asunto(s)
Adenocarcinoma Mucinoso/metabolismo , Neoplasias Colorrectales/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Adenocarcinoma/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Genotipo , Células HCT116 , Humanos , Lentivirus/genética , Metaloproteinasa 7 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
19.
J Biomater Appl ; 29(1): 72-80, 2014 07.
Artículo en Inglés | MEDLINE | ID: mdl-24327349

RESUMEN

PURPOSE: The objective of this study was to investigate the therapeutic potential of poly(lactic-co-glycolic acid) (PLGA) microspheres loaded with recombinant human growth and differentiation factor-5 (rhGDF-5) on the disc degeneration induced by needle puncture in a rat caudal disc model. METHODS: The rhGDF-5-loaded PLGA microspheres were prepared by the water-oil-water double-emulsion solvent evaporation method, and release kinetics was determined over 42 days. Rats that underwent 21-G needle puncture at rat tail discs were injected with rhGDF-5/PLGA microspheres at four weeks after needle injury. At eight weeks after the injection, disc height, glycosaminoglycans content, and DNA content of the discs were evaluated. In addition, gene expression analysis of aggrecan, collagen type I, and collagen type II in the rat nucleus pulposus was measured by real-time polymerase chain reaction. Rat discs were also assessed by histology using hematoxylin and eosin stain. RESULTS: Encapsulation of rhGDF-5 in PLGA microspheres guaranteed a sustained release of active rhGDF-5 for more than 42 days. The injection of GDF-5/PLGA microspheres resulted in a statistically significant restoration of disc height (p < 0.01), improvement of sulfated glycosaminoglycan (p < 0.05), DNA content (p < 0.05), and significantly increased mRNA levels of collagen type II (p < 0.01), and the differentiation index (the ratio of collagen type II to collagen type I, p < 0.01). In addition, rhGDF-5/PLGA microspheres treatment also improved histological changes induced by needle puncture. CONCLUSIONS: The results of this study suggest that injection of rhGDF-5 loaded in PLGA microspheres into rat tail discs may be as a promising therapy strategy to regenerate or repair the degenerative disc.


Asunto(s)
Factor 5 de Diferenciación de Crecimiento/administración & dosificación , Degeneración del Disco Intervertebral/tratamiento farmacológico , Agrecanos/genética , Animales , Materiales Biocompatibles , Colágeno/genética , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Factor 5 de Diferenciación de Crecimiento/farmacocinética , Humanos , Degeneración del Disco Intervertebral/metabolismo , Degeneración del Disco Intervertebral/patología , Ácido Láctico , Masculino , Ensayo de Materiales , Microesferas , Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética
20.
Neurosci Lett ; 539: 86-91, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23403104

RESUMEN

We have reported that human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) are capable of differentiating into dopaminergic (DA) neuron-like cells upon being induced by amniotic epithelial cells (AECs). However, what factor(s) is involved in the differentiation process has not been explored out thoroughly. Because pleiotrophin (PTN) is known to exert important trophic effects on DA neurons, in the present study, we investigated whether PTN is released by AECs and whether it is involved in the differentiation of hUCB-MSCs into DA neuron-like cells. The expression and secretion of PTN by AECs were detected by immunofluorescence, RT-PCR and ELISA. The hUCB-MSCs were isolated and treated with AEC-conditioned medium (ACM) or recombinant human PTN. Compared to the controls, a higher proportion of treated cells differentiated into DA neuron-like cells, indicated by the increased expression of TH and DAT and the increased dopamine content. These results indicate that PTN released by AECs acts as a synergetic factor with other neurotrophic factors and is involved in the differentiation of hUCB-MSCs into DA neuron-like cells. We suggest that ACM, which contains PTN and other neurotrophic factors, could potentially be used as an agent to promote the differentiation of DA neuron-like cells from hUCB-MSCs for cell therapy of Parkinson's disease without creating legal or ethical issues.


Asunto(s)
Amnios/citología , Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Neuronas Dopaminérgicas/citología , Células Epiteliales/citología , Sangre Fetal/citología , Células Madre Mesenquimatosas/citología , Amnios/metabolismo , Proteínas Portadoras/farmacología , Diferenciación Celular , Células Cultivadas , Medios de Cultivo Condicionados , Citocinas/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Sangre Fetal/metabolismo , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA