Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
1.
Gastroenterol Clin Biol ; 34(12): 653-61, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21051167

RESUMO

The fine-tuning between cell proliferation and differentiation of self-renewing stem cells and pluripotent progenitors in gastric glands and colon epithelial crypts is coordinated by the mechanisms that regulate colon epithelial cell migration and guidance along the crypt axis. This leads to the acquisition of specialized cellular functions and the exfoliation of desquamated senescent and apoptotic epithelial cells at the apical mucosa interface with the gut lumen. Self-renewing stem cells and pluripotent progenitors are involved in the clonal and polyclonal growth of digestive tumors. Several lines of evidence support the existence of a subpopulation of cancer cells with stem cell-like (SCL) phenotypes in solid tumors of breast and digestive system. Consistently, epithelial cancer cell lines in long-term culture are phenotypically and functionally heterogeneous. It is suggested that only a small proportion of transformed cells are clonogenic in vivo and ex vivo to form colonies and to initiate tumor growth in immunodeficient mice. A discrete subpopulation of tumor -initiating SCL cancer cells are highly competent to survive, propagate and spread through the invasive and metastatic cascade. A better understanding of the mechanisms driving the plasticity and pluripotency of stem cells, their derived progenitors and SCL colon cancer initiating cells during tumor progression will open new avenues for the early detection and treatment of local and distant tumors of the digestive tract.


Assuntos
Neoplasias do Colo/etiologia , Neoplasias do Colo/patologia , Células-Tronco , Animais , Humanos , Células-Tronco Neoplásicas , Células-Tronco/fisiologia
2.
Oncogene ; 26(38): 5615-25, 2007 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-17334389

RESUMO

Deleted in colon cancer (DCC) and UNC5 function as netrin dependence receptors by inducing apoptosis in the absence of their ligand and accordingly were recently designated as putative conditional tumor suppressors. Herein, we determined whether netrin-1 and its receptors are implicated in cancer cell invasion and tumor progression. Expression of DCC, UNC5 and adenosine A2B-receptors (A2B-Rs) was investigated by reverse transcription polymerase chain reaction in human colon cancer cells. The impact of DCC restitution and netrin-1 was evaluated on collagen type I invasion, tumor growth and metastasis in nude mice, cancer cell survival and gene expression profiling. Flow cytometry, poly(ADP-ribose)polymerase-1 and caspase-8 activation were used to evaluate the impact of DCC on cell death. Both netrin-1 and A2B-R activation induced the invasive phenotype through the Rho-Rho kinase axis in DCC-deficient human colorectal cancer cells. Restitution of wild-type DCC blocked invasion induced by netrin-1, A2B-R agonist and other agents. Ectopic expression of netrin-1 led to increased growth of human colon tumor xenografts in athymic mice. Conversely, introduction of wt-DCC in kidney MDCKts.src-ggl cells strongly inhibited metastasis in lymph nodes and lungs and increased sensitivity to apoptosis in hypoxia. DNA microarrays revealed that netrin and DCC had common and divergent impacts on gene expression linked to cell cycle, survival, surface signaling and adhesion. Our findings underscore that netrin is a potent invasion and tumor growth-promoting agent and that DCC is a metastasis suppressor gene targeting both proinvasive and survival pathways in a cumulative manner.


Assuntos
Neoplasias/patologia , Fatores de Crescimento Neural/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Western Blotting , Hipóxia Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Proliferação de Células , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Receptor DCC , Regulação Neoplásica da Expressão Gênica , Células HT29 , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Fatores de Crescimento Neural/genética , Netrina-1 , Receptor A2B de Adenosina/genética , Receptor A2B de Adenosina/metabolismo , Receptores de Superfície Celular/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transplante Heterólogo , Proteínas Supressoras de Tumor/genética
3.
Oncogene ; 26(41): 6071-81, 2007 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-17667937

RESUMO

The recently identified subfamily of WNK protein kinases is characterized by a unique sequence variation in the catalytic domain and four related human WNK genes were identified. Here, we describe the cloning and functional analysis of the human family member WNK2. We show that the depletion of endogenous WNK2 expression by RNA interference in human cervical HeLa cancer cells led to the activation of the extracellular signal-regulated kinase (ERK)1/2 mitogen-activated protein kinases but, in contrast to the depletion of WNK1, had no effect on ERK5. Furthermore, expression of a kinase-dead WNK2-K207M mutant also activated ERK1/2 suggesting that WNK2 catalytic activity is required. Depletion of WNK2 expression increased G1/S progression and potentiated the cellular response to low epidermal growth factor concentrations. The molecular mechanism of ERK1/2 activation in WNK2-depleted cells lies downstream of the Raf kinases and involves MEK1 phosphorylation at serine 298 in both HeLa and HT29 colon cancer cells. This modification is linked to the upregulation of MEK1 activity toward ERK1/2. Together, these results provide evidence that WNK2 is involved in the modulation of growth factor-induced cancer cell proliferation through the MEK1/ERK1/2 pathway. The data identify WNK2 as a candidate tumor suppressor gene and suggest a coordinated activity of WNK kinases in the regulation of cell proliferation.


Assuntos
Divisão Celular/efeitos dos fármacos , MAP Quinase Quinase 1/metabolismo , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética , Processamento Alternativo , Clonagem Molecular , Replicação do DNA , Ativação Enzimática , Células HeLa , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases/deficiência , Proteínas Serina-Treonina Quinases/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Oncogene ; 25(50): 6628-36, 2006 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-16715141

RESUMO

TFF1 is overexpressed in inflammatory diseases and human cancers of the digestive and urogenital systems. To examine the transforming potential of TFF1 in human colon epithelial cells, premalignant PC/AA/C1 adenoma cells (PC) derived from a patient with familial adenomatous polyposis (FAP) were transformed by the TFF1 cDNA and used as a model of the adenoma-carcinoma transition. Constitutive expression of TFF1 increased anchorage-independent cell growth in soft agar, and induced or potentiated the growth of colon PC-TFF1 and kidney MDCKts.src-TFF1 tumor xenografts in athymic mice. This resulted in reduction of thapsigargin-induced apoptosis and promotion of collagen type I invasion through several oncogenic pathways. Using the differential display approach to identify TFF1 target genes, we found that the dual specific phosphatases Cdc25A and B implicated in cell cycle transitions are strongly upregulated under active forms in both PC-TFF1 and HCT8/S11-TFF1 colon cancer cells. Accordingly, TFF1 expression is absent in normal human colon crypts but is induced in correlation with Cdc25a and b transcript levels and tumor grade in familial and sporadic colon adenomas and carcinomas. We propose that TFF1 and Cdc25A-B cooperate with other dominant oncogenic pathways to induce the adenoma and adenocarcinoma transitions. Agents that target TFF1/Cdc25 signaling pathways may be useful for treating patients with TFF1-positive solid tumors.


Assuntos
Adenoma/patologia , Carcinoma/patologia , Proteínas de Ciclo Celular/metabolismo , Colo/citologia , Neoplasias do Colo/patologia , Mucosa Intestinal/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Fosfatases cdc25/metabolismo , Adenoma/enzimologia , Adenoma/metabolismo , Animais , Carcinoma/enzimologia , Carcinoma/metabolismo , Adesão Celular/genética , Proliferação de Células , Sobrevivência Celular , Transformação Celular Neoplásica/genética , Neoplasias do Colo/enzimologia , Neoplasias do Colo/metabolismo , Neoplasias Colorretais/metabolismo , Progressão da Doença , Cães , Indução Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica/genética , Fenótipo , Lesões Pré-Cancerosas/patologia , Transfecção , Transplante Heterólogo , Fator Trefoil-1 , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
5.
Poult Sci ; 96(12): 4409-4418, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29053815

RESUMO

In mammals, insulin regulates blood glucose levels and plays a key regulatory role in appetite via the hypothalamus. In contrast, chickens are characterized by atypical glucose homeostasis, with relatively high blood glucose levels, reduced glucose sensitivity of pancreatic beta cells, and large resistance to exogenous insulin. The aim of the present study was to investigate in chickens the effects of 5 h fasting and 5 h insulin immuno-neutralization on hypothalamic mRNA levels of 23 genes associated with food intake, energy balance, and glucose metabolism. We observed that insulin immune-neutralization by administration of anti-porcine insulin guinea pig serum (AI) significantly decreased food intake and increased plasma glucose levels in chickens, while 5 h fasting produced a limited and non-significant reduction in plasma glucose. In addition, 5 h fasting increased levels of NPY, TAS1R1, DIO2, LEPR, GLUT1, GLUT3, GLUT8, and GCK mRNA. In contrast, AI had no impact on the levels of any selected mRNA. Therefore, our results demonstrate that in chickens, food intake inhibition or satiety mechanisms induced by insulin immuno-neutralization do not rely on hypothalamic abundance of the 23 transcripts analyzed. The hypothalamic transcripts that were increased in the fasted group are likely components of a mechanism of adaptation to fasting in chickens.


Assuntos
Galinhas/fisiologia , Ingestão de Alimentos/fisiologia , Jejum/fisiologia , Expressão Gênica , Insulina/metabolismo , Animais , Galinhas/genética , Hipotálamo/metabolismo , Insulina/deficiência , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
6.
Cancer Res ; 48(18): 5079-83, 1988 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-2842044

RESUMO

High-performance liquid chromatography-purified 125I-vasoactive intestinal peptide (VIP) bound to T-47D human breast cancer cells in a specific, saturable, and reversible manner. Scatchard plots were compatible with the presence of one class of VIP receptors with high affinity (Kd = 4.5 X 10(-10) M VIP, and Bmax = 293 fmol/mg protein). The neuropeptide and its natural analogues inhibited the binding of 125I-VIP and stimulated cyclic AMP (cAMP) generation in T-47D cells 96-fold (EC50 = 7 X 10(-10) M VIP), in the following order of potency: VIP greater than helodermin greater than human peptide with N-terminal histidine and C-terminal methionine greater than human pancreatic growth hormone-releasing factor greater than human secretin. In contrast, 125I-VIP binding was not displaced by pancreatic glucagon, human oxyntomodulin, truncated glucagon-like peptide-1, glucagon-like peptide-2, the somatostatin analogue SMS 201-995, gastric inhibitory peptide, and a series of steroid hormones or peptides unrelated to VIP. VIP also increased cAMP generation in seven other human breast cancer cell lines: H4-66B, HSL 53, HSL 78, MCF 7, MDA-MB231, T-47D2, and ZR75-1. Adenylate cyclase activity rose from 72.2 +/- 14 to 1069 +/- 66 pmol cAMP/min mg protein after the addition of 10(-7) M VIP to T-47D plasma membranes. In agreement with our pharmacological results and the Scatchard analysis of the binding data, sodium dodecyl sulfate-polyacrylamide gel electrophoresis of the solubilized receptor in the T-47D membranes permitted identification of one autoradiographic band with a molecular weight of 69,000. The sensitivity of the Mr 69,000 binding site to GTP and low doses of VIP implies that in T-47D cells, this component constitutes the membrane domain involved in the functional regulation of adenylate cyclase by VIP receptors. Our results indicate a role for the VIP receptor-cAMP system in human breast cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo , Adenilil Ciclases/metabolismo , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Reagentes de Ligações Cruzadas/farmacologia , AMP Cíclico/metabolismo , Eletroforese em Gel de Poliacrilamida , Feminino , Guanosina Trifosfato/metabolismo , Humanos , Peso Molecular , Receptores de Peptídeo Intestinal Vasoativo , Succinimidas/farmacologia , Peptídeo Intestinal Vasoativo/metabolismo
7.
Cancer Res ; 50(20): 6716-22, 1990 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-2145064

RESUMO

Normal and E1A-immortalized rat fetal intestinal epithelial SLC-11 cells were compared for the characteristics of the 35S-labeled proteoglycans isolated from their cell-associated and secreted fractions. In comparison with control cells in primary culture, immortalized SLC-11 cells: (a) secreted larger amounts of radiolabeled proteoglycans; (b) contained larger amounts of membrane-intercalated proteoglycans, analyzed by hydrophobic affinity chromatography on octyl-Sepharose; (c) produced cell-associated and secreted proteoglycans of smaller hydrodynamic size, assessed by measurement of Kav values; (d) contained a higher percentage of heparan sulfate in the cell-associated proteoglycans, determined by differential susceptibility of glycosaminoglycans to specific glycosaminoglycan lyases; (e) displayed heparan sulfate and chondroitin sulfate with a shorter chain length; and (f) synthesized glycosaminoglycans with a lower degree of sulfation, determined by ion-exchange chromatography. Taken together, these results demonstrate that in E1A-immortalized intestinal epithelial SLC-11 cells, the expression of proteoglycans alters considerably at an early stage of oncogenic transformation.


Assuntos
Transformação Celular Neoplásica/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Oncogênicas Virais/genética , Proteoglicanas/análise , Proteínas Precoces de Adenovirus , Animais , Células Cultivadas , Epitélio/metabolismo , Epitélio/ultraestrutura , Feto , Glicosaminoglicanos/análise , Intestinos/ultraestrutura , Proteoglicanas/química , Proto-Oncogenes , Ratos , Ratos Endogâmicos , Solubilidade
8.
Cancer Res ; 46(9): 4406-13, 1986 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-3015387

RESUMO

Vasoactive intestinal peptide (VIP) receptors are widely distributed in different tissues or carcinoma cells originating from entoderm and have been shown to regulate the growth of colonic adenocarcinoma cells through the action of cyclic AMP (cAMP). After exposure of cultured HT-29 human colonic carcinoma cells to 10(-8) M VIP, the cAMP-mediated signals in response to a new challenge with this neuropeptide were strongly attenuated as a function of time (half-life, less than 3 min) and VIP concentrations (half-maximal desensitization, 4 X 10(-9) M VIP). Desensitization is receptor mediated as indicated by: (a) the pharmacological specificity of the desensitization (VIP greater than secretin); (b) the considerable decrease of the potentiative action of VIP on forskolin-induced cAMP generation; and (c) the close temporal relationship between VIP receptor desensitization and the disappearance of the VIP binding sites from the cell surface. Desensitization is reversible upon the removal of VIP. Recovery of functional VIP receptors is insensitive to cycloheximide treatment, is critically dependent upon temperature, and in optimal conditions (37 degrees C) does not exceed 75 and 55% of the binding of 125I-VIP monoiodinated on tyrosine residue and VIP-induced cAMP production, respectively. The characteristics of the desensitization and internalization/recycling of the VIP receptors in carcinoma cells in culture are consistent with the transient action of this neurotransmitter and underline the biological significance of these processes. The study of drugs and natural agents interfering with membrane regulation of VIP receptor density and activity may be of considerable importance in intestinal cell tumor biology.


Assuntos
Carcinoma/metabolismo , Neoplasias do Colo/metabolismo , Receptores de Superfície Celular/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo , Compartimento Celular , Células Cultivadas , Colforsina/farmacologia , AMP Cíclico/biossíntese , Cicloeximida/farmacologia , Relação Dose-Resposta a Droga , Humanos , Receptores de Peptídeo Intestinal Vasoativo , Secretina/metabolismo , Temperatura , Fatores de Tempo
9.
Cancer Res ; 60(3): 672-8, 2000 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-10676652

RESUMO

Bile acids were shown previously to inhibit proliferation and to induce monocytic differentiation in HL60 human acute promyelocytic leukemia cells (A. Zimber et al., Int. J. Cancer, 59: 71-77, 1994). In this report, we hypothesized that bile acids may exert a positive cooperativity with two known inducers of leukemic cell differentiation, all-trans retinoic acid and 1,25(OH)2-vitamin D3. Our results provide evidence that bile acids induced the monocytic differentiation of HL60 and THP-1 human leukemia cells exposed to ineffective concentrations of these inducers. The protein kinase C (PKC) inhibitors H-7 (10 and 20 microM) and staurosporine (5 and 20 nM) modulated the effects of bile acids on HL60 cell differentiation. Most interestingly, bile acids are shown herein to down-regulate the expression of the serine protease myeloblastin gene involved in the differentiation of myeloid hematopoietic cells. In agreement with the recent identification of nuclear receptors for bile acids, our data suggest that functional interactions between nuclear bile acid signaling pathways, PKC, and nuclear receptors for retinoic acid and vitamin D3 are involved in the down-regulation of the myeloblastin gene and the induction of cell differentiation in human leukemic cells.


Assuntos
Ácidos e Sais Biliares/farmacologia , Calcitriol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Serina Endopeptidases/genética , Tretinoína/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Regulação para Baixo , Células HL-60 , Humanos , Monócitos/citologia , Mieloblastina , Proteína Quinase C/antagonistas & inibidores , Acetato de Tetradecanoilforbol/farmacologia
10.
Cancer Res ; 57(9): 1776-84, 1997 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9135022

RESUMO

Carcinoembryonic antigen gene family member 2 (CGM2), a member of the carcinoembryonic antigen (CEA) family, is expressed in normal colon and rectum but is down-regulated in colorectal adenocarcinomas. In situ hybridization studies demonstrate that CGM2 expression is limited to epithelial cells in the upper third of the crypts. Two other CEA family members, biliary glycoprotein (BGP) and nonspecific cross-reacting antigen (NCA), are similarly expressed, whereas CEA transcripts were found down to the base of the crypts but were less predominant in the upper region. Only low CGM2 and BGP mRNA levels were seen in colorectal tumors. CEA mRNA was expressed at an equivalent level in normal epithelia and in tumor cells, whereas NCA transcript levels were upregulated in tumor cells. Monoclonal antibodies that recognize the CGM2 protein reveal its presence on the apical membranes of epithelial cells in the upper third of the crypts but its absence from colorectal tumors, which do express the CEA and NCA-50/90 proteins. The newly cloned CGM2 3'-untranslated region was used to probe RNAs from adenomas, colorectal tumors at different stages of progression, and liver metastases of colorectal adenocarcinomas. This showed that CGM2 is already down-regulated in adenomas when compared to normal mucosae. The CGM2 expression pattern along with its sequence homology to BGP suggests a similar tumor suppressor function for CGM2.


Assuntos
Adenocarcinoma/metabolismo , Antígenos de Neoplasias , Moléculas de Adesão Celular/genética , Neoplasias Colorretais/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Anticorpos Monoclonais , Antígenos CD , Sequência de Bases , Antígeno Carcinoembrionário/genética , Moléculas de Adesão Celular/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Regulação para Baixo , Proteínas Ligadas por GPI , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/genética , Humanos , Imuno-Histoquímica , Hibridização In Situ , Mucosa Intestinal/metabolismo , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Neoplásico/genética , Transcrição Gênica
11.
Cancer Res ; 53(12): 2762-70, 1993 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-8504416

RESUMO

The proteins encoded by ras and src protooncogenes are frequently activated in a constitutive state in human colorectal cancers. To investigate the mechanism(s) whereby oncogenic p21ras and pp60c-src contribute to malignant transformation of intestine, human colonic Caco-2 cells transfected with an activated (Val 12) human Ha-ras gene (Caco-2-T cells) or Py-MT oncogene, a constitutive activator of pp60c-src tyrosine kinase activity (Caco-2-MT cells), were analyzed for tumorigenicity, protein kinase C (PKC) isoform expression, and PKC activity. As compared with control vector Caco-2-H cells, Caco-2-T and Caco-2-MT cells displayed: (a) an enhanced tumorigenicity in nude mice; (b) a 4-fold increase in the level of PKC-alpha mRNA which was not due to enhanced mRNA stability and was mediated through a PKC-independent pathway since it persisted after PKC depletion; (c) increased PKC-alpha immunoreactive protein content (3-fold), total PKC catalytic activity (3.5-fold), and total cell number of [3H]phorbol-12,13-dibutyrate binding sites (4-fold); and (d) a 1.7-fold higher membrane-bound/total PKC activity ratio together with 1.8- and 1.5-fold increases in [3H]arachidonate- and [3H]myristate-labeled diacylglycerol levels. In conclusion, the tumorigenic progression induced by oncogenic p21ras or the Py-MT/pp60c-src complex in Caco-2 cells is associated with increased PKC-alpha gene transcription and PKC-alpha expression as well as with constitutive PKC activation. These results provide the first evidence that the PKC-alpha gene is a target for the signaling pathways of oncogenically activated p21ras and pp60c-src in human colonic cells. They raise the possibility that PKC-alpha is an effector of these oncoproteins for activation of Caco-2 cell tumorigenic potential.


Assuntos
Adenocarcinoma/enzimologia , Neoplasias do Colo/enzimologia , Genes Virais/fisiologia , Genes ras/fisiologia , Genes src/fisiologia , Isoenzimas/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Proteína Quinase C/metabolismo , Adenocarcinoma/genética , Animais , Membrana Celular/enzimologia , Neoplasias do Colo/genética , Citosol/enzimologia , Genes Virais/genética , Genes ras/genética , Genes src/genética , Glicerol/metabolismo , Meia-Vida , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteína Oncogênica p21(ras)/genética , Proteína Quinase C/genética , RNA Mensageiro/metabolismo , Transfecção , Células Tumorais Cultivadas
12.
Cancer Res ; 42(4): 1541-8, 1982 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-6277484

RESUMO

A human gastric adenocarcinoma cell line, HGT-1, was established in vitro from the primary tumor of a 60-year-old patient. Histological examination of the tumor revealed a poorly differentiated adenocarcinoma. Primary tumor cells were cloned in soft agarose and gave rise to tumor colonies. The procedures enabling us to form a continuous cell line from the agarose colonies are described. The cultured cells grew as monolayers of closely apposed polygonal cells with a population-doubling time of 19.48 +/- 1.20 (S.E.) hr during exponential growth at passage 59. They had an epithelial morphology. Ultrastructural studies revealed the presence of microvilli and tight junctions. The HGT-1 cell line is tumorigenic in nude mice and has a hyperdiploid karyotype with a modal number of 57 chromosomes. It exhibits numerous marker chromosomes. These human gastric epithelial cells do not secrete mucus or carcinoembryonic antigen. They exhibit functional histamine H2-receptors mediating cellular cyclic adenosine 3':5'-monophosphate production and adenylate cyclase activation. In conclusion, the use of a soft-agarose clonogenic assay permitted us to develop a cancer cell line without the problems of fibroblastic cell contamination. The existence of histamine H2-receptors on gastric HGT-1 cells stresses the importance of this line as a model for studies of regulatory mechanisms involved in gastric secretion.


Assuntos
Receptores Histamínicos H2/análise , Receptores Histamínicos/análise , Neoplasias Gástricas/patologia , Adenilil Ciclases/análise , Animais , Linhagem Celular , Cromossomos , AMP Cíclico/biossíntese , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Transplante de Neoplasias , Neoplasias Gástricas/genética
13.
Cancer Res ; 56(8): 1823-7, 1996 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-8620499

RESUMO

Five somatostatin receptor subtypes (sst1 to sst5) have been cloned. We demonstrated previously that sst2 and sst5 mediate the antiproliferative effect of the somatostatin analogues octreotide and vapreotide. Using reverse transcription-PCR, we investigated gene expression of the five receptors in 47 human normal and cancerous tissues or cell lines from pancreatic and colorectal origin. mRNAs of somatostatin receptor subtypes were detected in 98% of samples, with more than two mRNA subtypes being expressed in 55% of cases. sst1, sst4, and sst5 were heterogeneously expressed in both normal and cancerous tissues; sst3 was rarely or not expressed. sst2 was present in normal pancreatic tissues but was absent in exocrine pancreatic carcinomas and their metastases. sst2 mRNAs were detected in normal colon, sporadic polyadenomas, and 50% of Dukes' stage B and 20% of Dukes' stage C carcinomas but were undetectable in Dukes' stage D carcinomas, hepatic metastases, and adenomas from familial adenomatous polyposis. The loss of sst2 expression could represent a growth advantage in these tumors and provide an explanation for the lack of therapeutic effect of somatostatin analogues in such adenocarcinomas. A subtyping of somatostatin receptors should be carried out before considering a somatostatin analogue treatment in patients with colorectal or pancreatic cancer.


Assuntos
Neoplasias Colorretais/metabolismo , Expressão Gênica , Intestino Grosso/metabolismo , Pâncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Somatostatina/biossíntese , Receptores de Somatostatina/genética , Linhagem Celular , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/cirurgia , Humanos , Estadiamento de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Valores de Referência , Células Tumorais Cultivadas
14.
Oncogene ; 18(48): 6835-9, 1999 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-10597294

RESUMO

Rac1 is a member of the Ras superfamily of small GTPases involved in signal transduction pathways that induce the formation of lamellipodia, stimulate cell proliferation and activate the JNK/SAPK protein kinase cascade. Here we describe that amplification by RT-PCR of the entire Rac1 coding sequence from a series of human adult and fetal tissues revealed beside the expected Rac1 cDNA, a variant product which contained additional 57 nucleotides between codons 75 and 76. This variant resulted in an in-frame insertion of 19 new amino acids immediately behind the switch II region, including two potential threonine phosphorylation sites for casein kinase II and protein kinase C. Primers designed within and downstream of the inserted nucleotide sequence allowed isolation of a genomic clone with intronic consensus sequences demonstrating that the insertion corresponds to a novel, yet undescribed exon 3b. This Rac1 splice variant, designated Rac1b, was predominantly identified in skin and epithelial tissues from the intestinal tract. Most notably, the expression of rac1b versus rac1 was found to be elevated in colorectal tumors at various stages of neoplastic progression, as compared to their respective adjacent tissues. We suggest that the 19 amino acid-insertion following the switch II region may create a novel effector binding site in rac1b, and thus participate in signaling pathways related to the normal or neoplastic growth of the intestinal mucosa.


Assuntos
Neoplasias Colorretais/genética , Neuropeptídeos/genética , Splicing de RNA , Proteínas rac de Ligação ao GTP/genética , Adulto , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , DNA de Neoplasias , Humanos , Dados de Sequência Molecular , Neuropeptídeos/química , Fosforilação , Homologia de Sequência de Aminoácidos , Proteínas rac de Ligação ao GTP/química , Proteínas rac1 de Ligação ao GTP
15.
Oncogene ; 14(13): 1589-600, 1997 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-9129150

RESUMO

The products of ras and src proto-oncogenes are frequently activated in a constitutive state in human colorectal cancer. In this study we attempted to establish whether the tumorigenic progression induced by oncogenic activation of p21ras or pp60c-src in human colonic cells is associated with alterations of the activity and expression of nuclear factor kappaB (NF-kappaB), a transcription factor suspected to participate in the development of cancer. To this end, we used Caco-2 cells made highly tumorigenic by transfection with an activated Val-12 human Ha-ras gene or with the polyoma middle T (PyMT) oncogene, a constitutive activator of pp60c-src tyrosine kinase activity. Compared with control vector-transfected Caco-2 cells, both oncogene-transfected cell lines exhibited: (i) decreased constitutive NF-kappaB DNA-binding activity and NF-kappaB-mediated reporter gene expression, without alteration of their response to TNF-alpha for activation of these parameters; (ii) reduced NF-kappaB cytosolic stores along with a decreased p65 expression due, at least in part, to destabilization of p65 mRNA; (iii) a decrease in adhesion to extracellular matrix component-coated substrata which was partially corrected when stimulating NF-kappaB transcriptional activity with TNF-alpha. These results indicate that the tumorigenic progression induced by oncogenic p21ras or PyMT/pp60c-src in human colonic Caco-2 cells is associated with a down-regulation of p65 expression and NF-kappaB activity which could be responsible for the reduced adhesive properties of these cells after oncogene transfection.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Genes ras , NF-kappa B/genética , NF-kappa B/metabolismo , Oncogenes , Células CACO-2 , Adesão Celular , Citosol/metabolismo , DNA de Neoplasias/metabolismo , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Proteína Quinase C/metabolismo , Fator de Transcrição RelA , Transcrição Gênica , Transfecção
16.
Oncogene ; 17(7): 877-87, 1998 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-9780004

RESUMO

We previously reported that ras and polyoma middle T (PyMT), a constitutive activator of the src protooncogene product, up-regulated Caco-2 cell proliferation along with protein kinase C (PKC) alpha expression and PKC activity. We aimed to investigate whether oncogene-induced up-regulation of Caco-2 cell proliferation involved stimulation of the autocrine IGF-II/IGF-I receptor (IGFIR) loop described in these cells and if so, to analyse the role of overexpressed and activated PKC. Compared with control vector transfected Caco-2 cells, ras- and PyMT-transfected cells exhibited increased expression of the 6.0 and 4.8 kb IGF-II transcripts. This was due to increased activity of the P3 and P4 promoters of the IGF-II gene which correlated with increased expression and DNA-binding activity of Sp1, a transcription factor interacting with several specific sites in P3 and P4 promoters. Oncogene-transfected cells displayed enhanced autocrine IGF-II production, which was fully responsible for the oncogene-induced increase in their proliferation since this increase was blunted by anti-human IGF-II and IGF1R (alphaIR3) antibodies. PKC mediated oncogene activation of the IGF-II gene presumably through action on Sp1 since (i) PKC activation by phorbol 12-myristate 13-acetate increased Sp1 expression, P3 and P4 activity and IGF-II mRNA in control but not in oncogene-transfected cells; and (ii) PKC inhibition by the PKC inhibitor Gö6976 reduced Sp1, P3 and P4 activity and IGF-II mRNA in all three cell lines. This is the first evidence that ras- and PyMT/src oncogenes up-regulate Caco-2 cell proliferation through a PKC-mediated pathway which stimulates IGF-II gene transcription and thereby increases autocrine IGF-II production. The mechanisms underlying IGF-II gene activation by PKC most probably involve action on Sp1.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Divisão Celular/genética , Regulação Neoplásica da Expressão Gênica , Genes ras , Fator de Crescimento Insulin-Like II/genética , Oncogenes , Proteína Quinase C/metabolismo , Transcrição Gênica , Anticorpos Monoclonais/farmacologia , Antígenos Transformantes de Poliomavirus/biossíntese , Células CACO-2 , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/biossíntese , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Fator de Crescimento Insulin-Like II/biossíntese , Fator de Crescimento Insulin-Like II/fisiologia , Receptor IGF Tipo 1/fisiologia , Transdução de Sinais , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Ativação Transcricional , Transfecção
17.
Oncogene ; 18(1): 87-92, 1999 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-9926923

RESUMO

Downregulation of the colon tumour-suppressor homeobox gene Cdx-2 by oncogenic ras Constitutive activation of the ras proto-oncogene is a frequent and early event in colon cancers, but the downstream nuclear targets are not fully understood. The Cdx-1 and Cdx-2 homeobox genes play crucial roles in intestinal cell proliferation and differentiation. In addition, Cdx-2 is a colonic tumour-suppressor gene, whereas Cdx-1 has oncogenic potential. Here, we show that constitutive activation of ras alters Cdx-1 and Cdx-2 expression in human colonic Caco-2 and HT-29 cells that harbour a normal ras proto-oncogene. Oncogenic ras downregulates Cdx-2 through activation of the PKC pathway and a decline in activity of the Cdx-2 promoter AP-1 site. This decline results from a PKC-dependent decrease in the relative expression of c-Jun, an activator of Cdx-2 transcription, compared to c-Fos, an inhibitor of Cdx-2. Unlike Cdx-2, Cdx-1 is upregulated by oncogenic ras and this effect is mediated by activation of the MEK1 pathway. These results indicate that oncogenic ras activation has opposite effects on Cdx-1 and Cdx-2 expression through distinct signalling pathways and they provide the first evidence for a functional link between ras activation and the downregulation of the Cdx-2 tumour-suppressor gene in colon cancer cells.


Assuntos
Proteínas Aviárias , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Genes ras , Proteínas de Homeodomínio/genética , Fator de Transcrição CDX2 , Células CACO-2 , Neoplasias do Colo , Citoplasma , Células HT29 , Humanos , Isoenzimas/metabolismo , Regiões Promotoras Genéticas , Proteína Quinase C/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-jun/genética , Elementos de Resposta , Transdução de Sinais , Transativadores , Fator de Transcrição AP-1/metabolismo
18.
Oncogene ; 18(26): 3878-85, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10445851

RESUMO

The TEL/PDGFR beta (T/P) fusion protein isolated from patients bearing a t(5;12) translocation is transforming when expressed in haematopoietic cells. To examine the signal transduction events activated by this protein, we measured the effect of T/P on activation of the c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) in mouse bone marrow-derived Ba/F3 cells. Significant increase in the activity of JNK/SAPK1 was observed in transient transfection as well as in Ba/F3 cells stably expressing T/P. This activation was abrogated when the T/P-expressing cells were treated with a specific inhibitor of the PDGFR beta tyrosine kinase, indicating that the activity of the PDGFR beta part of the fusion protein was involved in JNK/SAPK activation. Expression of a dominant negative mutant of mitogen-activated protein kinase kinase 4 (MKK4), a direct activator of JNK/SAPK, prevented T/P-induced JNK/SAPK activation. In addition, inhibition of phosphoinositide-3 OH kinase (PI-3 kinase), a promoting survival factor, potentiated the effect of T/P on JNK/SAPK activation. Interestingly, expression of T/P was shown to initiate an apoptotic response that was enhanced by treatment of cells with the PI-3 kinase inhibitor LY294002, suggesting that T/P mediated cell death through activation of JNK/SAPK signalling pathway. Consistent with this hypothesis, expression of the dominant negative mutant of MKK4 decreased T/P-mediated apoptosis, while a dominant-negative mutant of PI-3 kinase enhances cell death. These findings indicate that activation of JNK/SAPK by T/P is related to apoptosis rather than cell proliferation and transformation.


Assuntos
Apoptose/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno , Proteínas de Fusão Oncogênica/farmacologia , Transdução de Sinais/fisiologia , Animais , Apoptose/genética , Linhagem Celular , Cromonas/farmacologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Morfolinas/farmacologia , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Proteínas de Fusão Oncogênica/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/fisiologia , Transfecção
19.
Oncogene ; 19(10): 1277-87, 2000 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-10713669

RESUMO

The transforming growth factor beta (TGF-beta) plays an important role in constraining cellular proliferation, but it is also a potent inducer of programmed cell death or apoptosis. Here, we demonstrate that TGF-beta can have an opposite effect, acting as a survival factor to prevent c-Myc-induced cell death in Rat-1 fibroblasts. However, in marked contrast to TGF-beta, Smad2, which is a critical intracellular mediator of the TGF-beta signaling pathway, functions as an antagonist to induce increased cell death. The protective activity of TGF-beta was associated with the activation of c-Jun N-terminal Kinase (JNK) and was not linked to the ability of TGF-beta to promote cell cycle progression. Expression of dominant-interfering forms of various components of the JNK signaling pathway, including Rac1, Cdc42, mitogen-activated protein kinase kinase 4 (MKK4), and c-Jun, abolished TGF-beta-mediated cell survival. Furthermore, overexpression of the constitutively activated mutant RacL61F37A, which selectively stimulates JNK cascade but not G1 cell cycle progression or actin polymerization, was sufficient to prevent apoptosis induced by c-Myc. These findings describe a differential effect of two separated signaling pathways of TGF-beta and indicate for the first time that Smad2 can act as antagonist to suppress TGF-beta-dependent cell survival. Oncogene (2000) 19, 1277 - 1287.


Assuntos
Morte Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Ciclo Celular/fisiologia , Ativação Enzimática , Proteínas Quinases JNK Ativadas por Mitógeno , Ratos , Transdução de Sinais , Proteína Smad2 , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
20.
Oncogene ; 21(44): 6740-50, 2002 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-12360401

RESUMO

Bile acids are implicated in colorectal carcinogenesis as evidenced by epidemiological and experimental studies. We examined whether bile acids stimulate cellular invasion of human colorectal and dog kidney epithelial cells at different stages of tumor progression. Colon PC/AA/C1, PCmsrc, and HCT-8/E11 cells and kidney MDCKT23 cells were seeded on top of collagen type I gels and invasive cells were counted after 24 h incubation. Activation of the Rac1 and RhoA small GTPases was investigated by pull-down assays. Haptotaxis was analysed with modified Boyden chambers. Lithocholic acid, chenodeoxycholic acid, cholic acid and deoxycholic acid stimulated cellular invasion of SRC- and RhoA-transformed PCmsrc and MDCKT23-RhoAV14 cells, and of HCT-8/E11 cells originating from a sporadic tumor, but were ineffective in premalignant PC/AA/C1 and MDCKT23 cells. Bile acid-stimulated invasion occurred through stimulation of haptotaxis and was dependent on the RhoA/Rho-kinase pathway and signaling cascades using protein kinase C, mitogen-activated protein kinase, and cyclooxygenase-2. Accordingly, BA-induced invasion was associated with activation of the Rac1 and RhoA GTPases and expression of the farnesoid X receptor. We conclude that bile acids stimulate invasion and haptotaxis in colorectal cancer cells via several cancer invasion signaling pathways.


Assuntos
Ácidos e Sais Biliares/farmacologia , Neoplasias Colorretais/patologia , Genes src/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Ciclo-Oxigenase 2 , Relação Dose-Resposta a Droga , Guanosina Trifosfato/metabolismo , Humanos , Integrina beta1/fisiologia , Isoenzimas/fisiologia , Proteínas de Membrana , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/fisiologia , Lesões Pré-Cancerosas/patologia , Prostaglandina-Endoperóxido Sintases/fisiologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA