Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2323: 267-280, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34086287

RESUMEN

Spontaneous tumor regression following bacterial infection has been observed for hundreds of years. These observations along with anecdotal medical findings in 1890s led to the development of Coley's "toxins," consisting of killed Streptococcus pyogenes and Serratia marcescens bacteria, as the first cancer immunotherapy. The use of this approach, however, was not widely accepted at the time especially after the introduction of radiation therapy as a treatment for cancer in the early 1900s. Over the last 30-40 years there has been renewed interest in the use of bacteria to treat human solid tumors. This is based on the observation that various nonpathogenic anaerobic bacteria can infiltrate and replicate within solid tumors when given intravenously. Bacteria tested as potential anticancer agents include the Gram-positive obligate anaerobes Bifidobacterium and Clostridium, as well as the gram-negative facultative anaerobe Salmonella. Recent advances in synthetic biology and clinical success in cancer immunotherapy provide renewed momentum for developing bacteria-based cancer immunotherapy for cancer treatment and should allow greater potential for the development of novel therapeutic approaches for this devastating disease.


Asunto(s)
Terapia Biológica/métodos , Neoplasias/terapia , Interferencia de ARN , Biología Sintética/métodos , Animales , Línea Celular Tumoral , Ensayos Clínicos Fase I como Asunto , Neoplasias del Colon/microbiología , Neoplasias del Colon/terapia , Escherichia coli/genética , Escherichia coli/fisiología , Femenino , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Humanos , Inmunoterapia/métodos , Inmunoterapia/tendencias , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias/microbiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Inducción de Remisión , Salmonella typhimurium/genética , Salmonella typhimurium/fisiología , Especificidad de la Especie , Organismos Libres de Patógenos Específicos , Biología Sintética/tendencias , Ensayos Antitumor por Modelo de Xenoinjerto
2.
PLoS One ; 7(8): e43628, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22928007

RESUMEN

The cancer stem cell (CSC) model posits the presence of a small number of CSCs in the heterogeneous cancer cell population that are ultimately responsible for tumor initiation, as well as cancer recurrence and metastasis. CSCs have been isolated from a variety of human cancers and are able to generate a hierarchical and heterogeneous cancer cell population. CSCs are also resistant to conventional chemo- and radio-therapies. Here we report that ionizing radiation can induce stem cell-like properties in heterogeneous cancer cells. Exposure of non-stem cancer cells to ionizing radiation enhanced spherogenesis, and this was accompanied by upregulation of the pluripotency genes Sox2 and Oct3/4. Knockdown of Sox2 or Oct3/4 inhibited radiation-induced spherogenesis and increased cellular sensitivity to radiation. These data demonstrate that ionizing radiation can activate stemness pathways in heterogeneous cancer cells, resulting in the enrichment of a CSC subpopulation with higher resistance to radiotherapy.


Asunto(s)
Rayos gamma , Células Madre Neoplásicas/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética
3.
PLoS Pathog ; 8(7): e1002833, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22911498

RESUMEN

Infection with genus beta human papillomaviruses (HPV) is implicated in the development of non-melanoma skin cancer. This was first evidenced for HPV5 and 8 in patients with epidermodysplasia verruciformis (EV), a genetic skin disease. So far, it has been unknown how these viruses overcome cutaneous immune control allowing their persistence in lesional epidermis of these patients. Here we demonstrate that Langerhans cells, essential for skin immunosurveillance, are strongly reduced in HPV8-positive lesional epidermis from EV patients. Interestingly, the same lesions were largely devoid of the important Langerhans cells chemoattractant protein CCL20. Applying bioinformatic tools, chromatin immunoprecipitation assays and functional studies we identified the differentiation-associated transcription factor CCAAT/enhancer binding protein ß (C/EBPß) as a critical regulator of CCL20 gene expression in normal human keratinocytes. The physiological relevance of this finding is supported by our in vivo studies showing that the expression patterns of CCL20 and nuclear C/EBPß converge spatially in the most differentiated layers of human epidermis. Our analyses further identified C/EBPß as a novel target of the HPV8 E7 oncoprotein, which co-localizes with C/EBPß in the nucleus, co-precipitates with it and interferes with its binding to the CCL20 promoter in vivo. As a consequence, the HPV8 E7 but not E6 oncoprotein suppressed C/EBPß-inducible and constitutive CCL20 gene expression as well as Langerhans cell migration. In conclusion, our study unraveled a novel molecular mechanism central to cutaneous host defense. Interference of the HPV8 E7 oncoprotein with this regulatory pathway allows the virus to disrupt the immune barrier, a major prerequisite for its epithelial persistence and procarcinogenic activity.


Asunto(s)
Betapapillomavirus/patogenicidad , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Queratinocitos/metabolismo , Células de Langerhans/fisiología , Infecciones por Papillomavirus/inmunología , Neoplasias Cutáneas/virología , Betapapillomavirus/inmunología , Betapapillomavirus/metabolismo , Sitios de Unión , Proteína beta Potenciadora de Unión a CCAAT/genética , Línea Celular Tumoral , Movimiento Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos , Epidermis/metabolismo , Epidermis/virología , Epidermodisplasia Verruciforme/virología , Humanos , Queratinocitos/inmunología , Proteínas Oncogénicas Virales/metabolismo , Regiones Promotoras Genéticas
4.
Cell Cycle ; 11(14): 2691-8, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22732500

RESUMEN

Recent studies indicate that cancer stem cells (CSCs) exist in most hematological and solid tumors. CSCs are characterized by their ability to self-renew and their capacity to differentiate into the multitude of cells that comprise the tumor mass. Moreover, these cells have been shown to be intrinsically resistant to conventional anticancer therapies. Despite their fundamental role in cancer pathogenesis, the cellular origin of CSCs remains highly controversial. The aim of this study was to examine whether heterogeneous cancer cells can acquire stem cell-like properties in response to chemotherapy. We demonstrate that carboplatin can induce the self-renewal (spherogenesis) and pluripotency (Sox2 and Oct3/4 expression) of hepatocellular carcinoma (HCC) cells grown under stem cell culture conditions. Moreover, we show that non-CSC cells, obtained by side population flow cytometric sorting using Hoechst 33342, can acquire stem-like properties after exposure to carboplatin. Finally, we show that knockdown of Sox2 and Oct3/4 gene expression in HCC cells can reduce carboplatin-mediated increases in sphere formation and increase cellular sensitivity to chemotherapy. Taken together, our data indicate that bulk cancer cells may be an important source of CSCs during tumor development, and that targeting Sox2 and/or Oct3/4 may be a promising approach for targeting CSCs in clinical cancer treatment.


Asunto(s)
Antineoplásicos/toxicidad , Carboplatino/toxicidad , Células Madre Neoplásicas/metabolismo , Antineoplásicos/uso terapéutico , Carboplatino/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/antagonistas & inhibidores , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factores de Transcripción SOXB1/antagonistas & inhibidores , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
5.
Oncotarget ; 3(4): 365-70, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22562943

RESUMEN

Recent advances in cancer genomics have opened up unlimited potential for treating cancer by directly targeting culprit genes. However, novel delivery methods are needed in order for this potential to be translated into clinically viable treatments for patients. Magnetic nanoparticle technology offers the potential to achieve selective and efficient delivery of therapeutic genes by using external magnetic fields, and also allows simultaneous imaging to monitor the delivery in vivo. Compared to conventional gene delivery strategies, this technique has been shown to significantly increase gene delivery to human xenograft tumors models, as well as various internal organs (e.g. liver, kidney) and the central nervous system. Magnetic nanoparticle technology, therefore, has the potential to turn the challenge of gene therapy in vivo into a new frontier for cancer treatment.


Asunto(s)
Terapia Genética , Nanopartículas de Magnetita , Neoplasias/terapia , Animales , Técnicas de Transferencia de Gen/tendencias , Humanos , Ratones , Terapia Molecular Dirigida , Neoplasias/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Methods Mol Biol ; 487: 147-60, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19301646

RESUMEN

RNA interference (RNAi) is a potent and specific mechanism for eliminating the mRNA of specific genes. This gene silencing mechanism occurs naturally and is highly conserved from plants to human cells, holding promise for functional genomics and for revolutionizing medicine due to its unlimited potential to treat genetic, epigenetic, and infectious disease. However, efforts to unleash the enormous potential of RNAi have met with significant challenges. Delivery is problematic because short interfering RNAs (siRNA) are negatively charged polymers that inefficiently enter cells and undergo rapid enzymatic degradation in vivo. In addition, the synthesis of siRNAs is expensive for long-term research and therapeutic applications. Recently, we have shown that nonpathogenic bacteria can be engineered to activate RNAi in mammalian cells (TransKingdom RNA interference; tkRNAi). This new approach offers several advantages and has significant implications. First, this method allows the establishment of a long-term stable gene silencing system in the laboratory against genes of interests in vitro and in vivo, and enables high-throughput functional genomics screening in mammalian systems. RNAi libraries can be constructed, stored, reproduced, amplified, and used with the help of E. coli as currently done with gene cloning. Second, this technology provides a clinically compatible way to achieve RNAi for therapeutic applications due to the proven clinical safety ofnonpathogenic bacteria as a gene carrier, tkRNAi also eliminates the siRNA manufacture issue, and may circumvent or mitigate host interferon-like responses since siRNA is produced intracellularly.


Asunto(s)
Neoplasias del Colon/terapia , Técnicas de Silenciamiento del Gen/métodos , Silenciador del Gen , Terapia Genética/métodos , ARN Interferente Pequeño/administración & dosificación , beta Catenina/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Neoplasias del Colon/genética , Escherichia coli/genética , Femenino , Humanos , Técnicas In Vitro , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , ARN Interferente Pequeño/genética , Trasplante Heterólogo , beta Catenina/genética , beta Catenina/metabolismo
7.
Biotechnol Genet Eng Rev ; 25: 113-27, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-21412352

RESUMEN

Since its discovery in 1998 RNA interference (RNAi), a potent and highly selective gene silencing mechanism, has revolutionized the field of biological science. The ability of RNAi to specifically down-regulate the expression of any cellular protein has had a profound impact on the study of gene function in vitro. This property of RNAi also holds great promise for in vivo functional genomics and interventions against a wide spectrum of diseases, especially those with "undruggable" therapeutic targets. Despite the enormous potential of RNAi for medicine, development of in vivo applications has met with significant problems, particularly in terms of delivery. For effective gene silencing to occur, silencing RNA must reach the cytoplasm of the target cell. Consequently, various strategies using chemically modified siRNA, liposomes, nanoparticles and viral vectors are being developed to deliver silencing RNA. These approaches, however, can be expensive and in many cases they lack target cell specificity or clinical compatibility. Recently, we have shown that RNAi can be activated in vitro and in vivo by non-pathogenic bacteria engineered to manufacture and deliver silencing shRNA to target cells. This new approach, termed TransKingdom RNAi (tkRNAi), has several key advantages. First, tkRNAi may provide a viable means to accomplish therapeutic RNAi since non-pathogenic bacteria have a proven safety record in clinical applications. Second, tkRNAi eliminates the cost of siRNA manufacture since silencing shRNA are produced inside bacteria. Moreover, the intracellular mechanism of shRNA release inherent to tkRNAi may circumvent, or mitigate, the activation of host immune responses. Finally, tkRNAi may facilitate high-throughput in vivo functional genomics screening since bacteria-based RNAi libraries can be easily constructed, stored, reproduced and amplified, thereby allowing for the creation of a stable gene silencing system.


Asunto(s)
Bacterias/genética , Técnicas de Transferencia de Gen , Interferencia de ARN , Animales , Ingeniería Genética , Vectores Genéticos , Humanos , Liposomas , Ratones , Neoplasias/terapia , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Virus/genética
8.
J Infect Dis ; 196(1): 95-103, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17538889

RESUMEN

BACKGROUND: Helicobacter pylori infection increases the risk of gastric carcinogenesis. The aim of the present study was to determine whether H. pylori could up-regulate the expression of the epidermal growth factor receptor (EGFR), a critical gene in the carcinogenic process. METHODS: AGS gastric epithelial cells were infected with cag(+) toxigenic or cag(-) nontoxigenic strains of H. pylori or isogenic mutants. EGFR protein expression was determined by Western blotting and immunofluorescence. EGFR mRNA levels were evaluated using real-time polymerase chain reaction. The signaling pathways leading to EGFR up-regulation were examined using the ERK1/2 inhibitor PD98059, the Src inhibitor pp2, the nuclear factor- kappa B inhibitor caffeic acid phenethyl ester, EGFR neutralizing antibodies, and the EGFR kinase inhibitor AG1478. RESULTS: Infection of AGS cells by H. pylori significantly increased EGFR mRNA and protein levels. We found that this effect was limited to cag(+) H. pylori strains and that mutants with a defective type IV secretion system were unable to cause EGFR up-regulation. Increased EGFR expression was found to be dependent on EGFR receptor transactivation, ERK1/2 phosphorylation, and Src activation. CONCLUSION: Infection of gastric epithelial cells by H. pylori triggers an autocrine loop whereby EGFR transactivation leads to the up-regulation of EGFR expression. This, in turn, may contribute to unrestrained epithelial cell proliferation and carcinogenesis.


Asunto(s)
Células Epiteliales/microbiología , Receptores ErbB/metabolismo , Helicobacter pylori/fisiología , Estómago/microbiología , Regulación hacia Arriba , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Western Blotting , Línea Celular Tumoral , Receptores ErbB/genética , Técnica del Anticuerpo Fluorescente , Mucosa Gástrica/metabolismo , Perfilación de la Expresión Génica , Humanos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estómago/citología , Familia-src Quinasas/metabolismo
9.
J Immunol ; 178(12): 8013-21, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17548638

RESUMEN

Previously, we reported that normal colonocytes produce the memory CD4(+) T cell-directed chemokine MIP-3alpha, and that epithelial MIP-3alpha levels are elevated in inflammatory bowel disease. Interestingly, the unique receptor for MIP-3alpha, CCR6, is expressed by a variety of cell types including colonocytes, suggesting that MIP-3alpha may regulate additional biological activities in the intestine. The aim of this study was to determine whether MIP-3alpha can induce intestinal epithelial cell proliferation and to examine the signaling mechanisms that mediate this response. We show that nonstimulated Caco-2 and HT-29 colonic epithelial cells express CCR6, and that stimulation of Caco-2 cells by MIP-3alpha can dose dependently increase cell proliferation as well as activate the epidermal growth factor receptor (EGFR) and ERK1/2 MAPK. MIP-3alpha-mediated ERK1/2 activation in Caco-2 cells appeared to require metalloproteinase-dependent release of the endogenous EGFR ligand amphiregulin and transactivation of the EGFR. Moreover, blockade of amphiregulin bioactivity using a neutralizing polyclonal Ab significantly reduced MIP-3alpha-mediated, but not EGF-mediated Caco-2 cell proliferation. Taken together, our findings indicate that MIP-3alpha can regulate mitogenic signaling in colonic epithelial cells and thus may serve an important homeostatic function in the intestine by regulating tissue turnover and maintenance of the epithelium, in addition to its role in regulating leukocyte recruitment.


Asunto(s)
Quimiocinas CC/metabolismo , Colon/inmunología , Receptores ErbB/genética , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Inflamatorias de Macrófagos/metabolismo , Activación Transcripcional , Anfirregulina , Anticuerpos Monoclonales , Células CACO-2 , Proliferación Celular , Quimiocina CCL20 , Quimiocinas CC/farmacología , Colon/efectos de los fármacos , Familia de Proteínas EGF , Glicoproteínas/antagonistas & inhibidores , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Proteínas Inflamatorias de Macrófagos/farmacología , Metaloproteasas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Receptores CCR6 , Receptores de Quimiocina/metabolismo , Transducción de Señal
10.
Am J Physiol Gastrointest Liver Physiol ; 292(5): G1263-71, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17272517

RESUMEN

A characteristic feature of human inflammatory bowel disease, particularly Crohn's disease, is the presence of activated CD4(+) T cells. Recently, we have shown that colonic epithelial cell production of macrophage inflammatory protein (MIP)-3alpha, a CD4 T cell-directed chemokine, is elevated in inflammatory bowel disease. However, the functional relevance of MIP-3alpha production during intestinal inflammation is poorly understood. The aim of this study was to determine whether MIP-3alpha production is increased during murine 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis and to examine the effect of anti-MIP-3alpha neutralizing monoclonal antibody administration in this model. We found that the administration of TNBS significantly increased colonic MIP-3alpha protein levels in Balb/c mice. Consistent with this, a marked increase in the number of CCR6-bearing lamina propria CD4(+) and CD8(+) T cells was also observed in TNBS-treated animals. Treatment of mice with an anti-MIP-3alpha neutralizing monoclonal antibody significantly reduced TNBS-mediated increases in colonic weight-to-length ratio, mucosal ulceration, histological damage, and myeloperoxidase activity. TNBS-mediated increases in the number of CCR6-bearing lamina propria T cells were also substantially reduced by anti-MIP-3alpha neutralizing monoclonal antibody treatment. Taken together, our findings indicate that blockade of MIP-3alpha bioactivity can significantly reduce TNBS-mediated colonic injury and T cell recruitment, suggesting a role for this chemokine in the pathophysiology of intestinal inflammation.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Quimiocinas CC/inmunología , Colitis/prevención & control , Enfermedades del Colon/prevención & control , Proteínas Inflamatorias de Macrófagos/inmunología , Animales , Recuento de Linfocito CD4 , Antígenos CD8/análisis , Quimiocina CCL20 , Colitis/inmunología , Enfermedades del Colon/inducido químicamente , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ácido Trinitrobencenosulfónico , Regulación hacia Arriba
11.
Pharmacogenomics ; 8(7): 867-71, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18240911

RESUMEN

Cequent Pharmaceuticals, Inc. is a recently established biopharmaceutical company that aims to develop clinically compatible therapies based on RNAi, a potent gene-silencing mechanism discovered in 1998. The company's proprietary technology, transkingdom RNAi (tkRNAi), uses nonpathogenic bacteria to produce and deliver shRNA into target cells to induce RNAi. Our initial focus is on the development of a tkRNAi-based therapy for familial adenatomous polyposis, an inherited form of colon cancer. Cequent's first tkRNAi-based drug for familial adenatomous polyposis, CEQ501, is currently in advanced preclinical testing. As part of its ongoing activities, Cequent plans to develop additional tkRNAi-based products for indications within and outside the GI tract. Our overall goal is to establish tkRNAi as a platform for developing a wide range of RNAi-based therapies.


Asunto(s)
Industria Farmacéutica , Terapia Genética/métodos , Interferencia de ARN , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/terapia , Boston , Humanos
12.
Am J Physiol Gastrointest Liver Physiol ; 289(6): G1075-83, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16099872

RESUMEN

Epithelial neutrophil-activating peptide-78 (ENA-78), a member of the CXC chemokine subfamily, is induced by inflammatory cytokines in human colonic enterocyte cell lines and increased in the colon of patients with inflammatory bowel disease (IBD). Lipopolysaccharide-induced CXC-chemokine (LIX) was recently identified as the murine homolog of ENA-78. Here we show that, similar to ENA-78, inflammatory cytokine stimulation of a murine colonic epithelial cell line, MODE-K, results in increased LIX expression. Consistent with the expression pattern of ENA-78 in IBD, LIX expression is significantly increased in mice with colitis induced by the ingestion of dextran sodium sulfate (DSS). Treating mice with antisense oligonucleotides to LIX via rectal enema delivery before DSS treatment results in colonic enterocyte uptake and a significant reduction in neutrophil infiltration and severity of colitis. These findings indicate that LIX plays an integral role in the pathogenesis of DSS-induced colitis. Similarly, enterocyte-derived CXC chemokines may play a key role in regulating neutrophil recruitment and intestinal injury in IBD. The intracolonic administration of ENA-78 antisense oligonucleotides may be effective in treating distal ulcerative colitis in humans.


Asunto(s)
Quimiocinas CXC/antagonistas & inhibidores , Colitis/tratamiento farmacológico , Oligodesoxirribonucleótidos Antisentido/uso terapéutico , Animales , Línea Celular , Quimiocina CXCL5 , Quimiocinas CXC/biosíntesis , Quimiocinas CXC/genética , Colitis/inducido químicamente , Colitis/patología , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Enterocitos/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C
13.
J Biol Chem ; 279(24): 25179-88, 2004 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-15069060

RESUMEN

Flagellin, a specific ligand for Toll-like receptor 5 (TLR5), is a molecular pattern associated with several bacterial species. Recently, TLR signaling has been intensively studied. However, TLR5-associated signaling in non-transformed colonocytes has not been investigated. Here we studied the expression of cytokines induced by flagellin in non-transformed human colonic NCM460 cells and the signaling mechanisms mediating these responses. Cytokine expression array experiments showed that exposure of the cells to flagellin (100 ng/ml) for 12 h increased the expression of interleukin (IL)-8 and macrophage-inflammatory protein 3alpha (MIP3alpha) in a TLR5-specific manner. Flagellin also activated MAP kinases (ERK1/2, JNK, and p38) and degraded IkappaBalpha. Dominant negative MEK1 (a kinase that activates ERK1/2) blocked flagellin-stimulated IL-8 and MIP3alpha transcriptional activity, while the MEK-specific inhibitors PD98059 and U0126 reduced protein production of these cytokines. Conversely, transfection with a constitutively active MEK1 increased IL-8 and MIP3alpha transcriptional activity in a NFkappaB-independent manner. Furthermore, overexpression of the constitutively active MEK1 induced IL-8 and MIP3alpha protein production. We also demonstrated that C-terminal coiled-coil and TRAF-C domains of TRAF6, unable to mediate NFkappaB activation, are involved in MEK-mediated IL-8 and MIP3alpha expression. Thus, in non-transformed human colonocytes, MEK activation following flagellin/TLR5 engagement is a key modulator for NFkappaB-independent, IL-8 and MIP3alpha expression.


Asunto(s)
Quimiocinas CC/genética , Colon/metabolismo , Regulación de la Expresión Génica , Interleucina-8/genética , Proteínas Inflamatorias de Macrófagos/genética , Glicoproteínas de Membrana/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Receptores de Superficie Celular/fisiología , Quimiocina CCL20 , Colon/citología , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , MAP Quinasa Quinasa 1 , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteínas/fisiología , Transducción de Señal , Factor 6 Asociado a Receptor de TNF , Receptor Toll-Like 5 , Receptores Toll-Like
14.
J Biol Chem ; 278(2): 875-84, 2003 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-12414801

RESUMEN

We have previously shown that colonic epithelial cells are a major site of MIP-3alpha production in human colon and that enterocyte MIP-3alpha protein levels are elevated in inflammatory bowel disease. The aim of this study was to determine the molecular mechanisms regulating MIP-3alpha gene transcription in Caco-2 intestinal epithelial cells. We show that a kappaB element at nucleotides -82 to -93 of the MIP-3alpha promoter binds p50/p65 NF-kappaB heterodimers and is a major regulator of basal and interleukin-1beta (IL-1beta)-mediated gene activation. Scanning mutagenesis of the MIP-3alpha 5'-flanking region also identified two additional binding elements: Site X (nucleotides -63 to -69) and Site Y (nucleotides -143 to -154). Site X (CGCCTTC) bound Sp1 and regulated basal MIP-3alpha gene transcription. Overexpression of Sp1 increased basal luciferase activity, whereas, substitutions in the Sp1 element significantly reduced reporter activity. In contrast, Site Y (AAGCAGGAAGTT) regulated both basal and cytokine-induced gene activation and bound the Ets nuclear factor ESE-1. Substitutions in the Site Y element markedly reduced inducible MIP-3alpha reporter activity. Conversely, overexpression of ESE-1 significantly up-regulated MIP-3alpha luciferase levels. Taken together, our findings demonstrate that co-ordinate activation and binding of ESE-1, Sp1, and NF-kappaB to the MIP-3alpha promoter is required for maximal gene expression by cytokine-stimulated Caco-2 human intestinal epithelial cells.


Asunto(s)
Quimiocinas CC/genética , Proteínas Inflamatorias de Macrófagos/genética , Proteínas Proto-Oncogénicas , Receptores de Quimiocina , Transactivadores/fisiología , Secuencia de Bases , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células CACO-2 , Quimiocina CCL20 , Clonación Molecular , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Interleucina-1/farmacología , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-ets , Receptores CCR6 , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3 , Factores de Transcripción/metabolismo , Transcripción Genética , Activación Transcripcional
15.
Gastroenterology ; 122(4): 1048-57, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11910356

RESUMEN

BACKGROUND & AIMS: Clostridium difficile toxin A causes mitochondrial dysfunction resulting in generation of oxygen radicals and adenosine triphosphate (ATP) depletion. We investigated whether mitochondrial dysfunction is involved in nuclear factor kappaB (NF-kappaB) activation and interleukin (IL)-8 release from toxin A-exposed enterocytes. METHODS: NF-kappaB activation and IL-8 release in response to toxin A were correlated with reactive oxygen intermediate (ROI) generation and ATP production in HT-29 monolayers or HT-29 cells exposed to ethidium bromide (EB) to inhibit mitochondrial function. RESULTS: HT-29 cells exposed to EB showed damaged mitochondria and diminished resting levels of ATP. ROI production in EB-treated cells exposed to toxin A for 30 minutes was significantly reduced. Exposure of wild-type HT-29 cells to toxin A resulted in increased oxygen radical generation and IL-8 production (P < 0.01 vs. control) that was inhibited by antioxidant pretreatment. Degradation of IkappaB was observed within 30 minutes of toxin exposure, before ras homologue (Rho) glucosylation, and was followed by nuclear translocation of NF-kappaB. Toxin A did not increase IL-8 levels in EB-treated cells, whereas IL-8 release in response to IL-1beta was not affected. CONCLUSIONS: Our data support an early role for mitochondria-derived ROIs in stimulation of IL-8 release from colonocytes by toxin A. ROI generation is independent of Rho inactivation and involves nuclear translocation of NF-kappaB before release of IL-8.


Asunto(s)
Toxinas Bacterianas/farmacología , Enterotoxinas/farmacología , Proteínas I-kappa B , Interleucina-8/metabolismo , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Proteínas de Unión al ADN/metabolismo , Glicosilación , Células HT29 , Humanos , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA