Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 10(9): e1004341, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25188454

RESUMO

An overwhelming neutrophil-driven response causes both acute symptoms and the lasting sequelae that result from infection with Neisseria gonorrhoeae. Neutrophils undergo an aggressive opsonin-independent response to N. gonorrhoeae, driven by the innate decoy receptor CEACAM3. CEACAM3 is exclusively expressed by human neutrophils, and drives a potent binding, phagocytic engulfment and oxidative killing of Opa-expressing bacteria. In this study, we sought to explore the contribution of neutrophils to the pathogenic inflammatory process that typifies gonorrhea. Genome-wide microarray and biochemical profiling of gonococcal-infected neutrophils revealed that CEACAM3 engagement triggers a Syk-, PKCδ- and Tak1-dependent signaling cascade that results in the activation of an NF-κB-dependent transcriptional response, with consequent production of pro-inflammatory cytokines. Using an in vivo model of N. gonorrhoeae infection, we show that human CEACAM-expressing neutrophils have heightened migration toward the site of the infection where they may be further activated upon Opa-dependent binding. Together, this study establishes that the role of CEACAM3 is not restricted to the direct opsonin-independent killing by neutrophils, since it also drives the vigorous inflammatory response that typifies gonorrhea. By carrying the potential to mobilize increasing numbers of neutrophils, CEACAM3 thereby represents the tipping point between protective and pathogenic outcomes of N. gonorrhoeae infection.


Assuntos
Biomarcadores/metabolismo , Gonorreia/imunologia , Mediadores da Inflamação/metabolismo , Inflamação/etiologia , Neisseria gonorrhoeae/patogenicidade , Neutrófilos/imunologia , Animais , Aderência Bacteriana , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/metabolismo , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica , Gonorreia/metabolismo , Gonorreia/microbiologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Neisseria gonorrhoeae/imunologia , Neutrófilos/microbiologia , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo , Fagocitose/fisiologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Quinase Syk
2.
Inflamm Bowel Dis ; 18(2): 294-304, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21688348

RESUMO

BACKGROUND: Abnormal expression of CEACAM6 observed on the ileal epithelium in Crohn's disease (CD) patients allows adherent-invasive Escherichia coli (AIEC) to colonize gut mucosa. Since intestinal permeability is significantly increased in CD patients, we aimed at investigating whether and how AIEC alter barrier function. METHODS: Tissue microarray was performed on ileal biopsies from CD patients in quiescent and active phases. CEABAC10 or wildtype mice were orally challenged with 10(9) bacteria. Intestinal permeability was assessed by measuring 4 kDa dextran-FITC flux in serum, barrier integrity was analyzed using biotin tracer experiment, and claudin-2 protein immunostaining. Bacterial translocation was analyzed in Ussing chambers. RESULTS: Pore-forming tight junction protein claudin-2 is strongly expressed in the ileum of 51% patients in quiescent phase and in 49% of the patients with active CD. Infection of CEABAC10 transgenic mice expressing human CEACAMs with AIEC, but not with nonpathogenic E. coli, led to a significant 3.0-fold increase in intestinal permeability and to disruption of mucosal integrity in a type 1 pili-dependent mechanism. This is consistent with the claudin-2 abnormal expression at the plasma membrane of intestinal epithelial cells observed in AIEC-infected CEABAC10 mice. AIEC bacteria were able to translocate through CEABAC10 intestinal mucosa. CONCLUSIONS: These findings strongly support the hypothesis that AIEC type 1 pili-mediated interaction with CEACAM6 abnormally expressed in the quiescent phase of CD may disrupt intestinal barrier integrity before the onset of inflammation. Thus, therapeutic targeting claudin-2 induced by AIEC infection could be a new clinical strategy for preserving intestinal barrier function in CD patients.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Claudinas/biossíntese , Doença de Crohn/metabolismo , Infecções por Escherichia coli/metabolismo , Mucosa Intestinal/metabolismo , Adulto , Animais , Antígenos CD/genética , Aderência Bacteriana , Moléculas de Adesão Celular/genética , Doença de Crohn/microbiologia , Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Humanos , Íleo/metabolismo , Íleo/microbiologia , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Permeabilidade , Análise Serial de Tecidos/métodos
3.
J Exp Med ; 206(10): 2179-89, 2009 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-19737864

RESUMO

Abnormal expression of CEACAM6 is observed at the apical surface of the ileal epithelium in Crohn's disease (CD) patients, and CD ileal lesions are colonized by pathogenic adherent-invasive Escherichia coli (AIEC). We investigated the ability of AIEC reference strain LF82 to colonize the intestinal mucosa and to induce inflammation in CEABAC10 transgenic mice expressing human CEACAMs. AIEC LF82 virulent bacteria, but not nonpathogenic E. coli K-12, were able to persist in the gut of CEABAC10 transgenic mice and to induce severe colitis with reduced survival rate, marked weight loss, increased rectal bleeding, presence of erosive lesions, mucosal inflammation, and increased proinflammatory cytokine expression. The colitis depended on type 1 pili expression by AIEC bacteria and on intestinal CEACAM expression because no sign of colitis was observed in transgenic mice infected with type 1 pili-negative LF82-Delta fimH isogenic mutant or in wild-type mice infected with AIEC LF82 bacteria. These findings strongly support the hypothesis that in CD patients having an abnormal intestinal expression of CEACAM6, AIEC bacteria via type 1 pili expression can colonize the intestinal mucosa and induce gut inflammation. Thus, targeting AIEC adhesion to gut mucosa represents a new strategy for clinicians to prevent and/or to treat ileal CD.


Assuntos
Antígenos CD/fisiologia , Moléculas de Adesão Celular/fisiologia , Colite/etiologia , Doença de Crohn/microbiologia , Escherichia coli/patogenicidade , Trato Gastrointestinal/microbiologia , Adesinas de Escherichia coli/fisiologia , Animais , Aderência Bacteriana , Proteínas de Fímbrias/fisiologia , Proteínas Ligadas por GPI , Humanos , Camundongos , Camundongos Transgênicos
4.
PLoS One ; 2(12): e1353, 2007 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18159236

RESUMO

CEA and CEACAM6 are immunoglobulin family intercellular adhesion molecules that are up-regulated without structural mutations in approximately 70% of human cancers. Results in in vitro systems showing tumorigenic effects for these molecules suggest that this correlation could indicate an instrumental role in tumorigenesis. To test whether this applies in vivo, transgenic mice harboring 187 kb of the human genome containing four CEA family member genes including the CEA and CEACAM6 genes were created and their copy numbers increased by mating until colonocyte expression levels reached levels seen in human colorectal carcinomas. The colonocyte surface level of integrin alpha5 and the activation of AKT increased progressively with the expression levels of CEA/CEACAM6. Colonic crypts showed a progressive increase in colonocyte proliferation, an increase in crypt fission, and a strong inhibition of both differentiation and anoikis/apoptosis. All transgenic mice showed massively enlarged colons comprising a continuous mosaic of severe hyperplasia, dysplasia and serrated adenomatous morphology. These results suggest that up-regulated non-mutated adhesion molecules could have a significant instrumental role in human cancer.


Assuntos
Antígeno Carcinoembrionário/imunologia , Colo/patologia , Reto/patologia , Animais , Antígeno Carcinoembrionário/genética , Colo/imunologia , Humanos , Integrinas/metabolismo , Camundongos , Camundongos Transgênicos , Reto/imunologia , Frações Subcelulares/imunologia
5.
J Cell Biol ; 177(2): 211-8, 2007 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-17438079

RESUMO

Exchanging the glycophosphatidylinositol (GPI) anchor signal sequence of neural cell adhesion molecule (NCAM) for the signal sequence of carcinoembryonic antigen (CEA) generates a mature protein with NCAM external domains but CEA-like tumorigenic activity. We hypothesized that this resulted from the presence of a functional specificity signal within this sequence and generated CEA/NCAM chimeras to identify this signal. Replacing the residues (GLSAG) 6-10 amino acids downstream of the CEA anchor addition site with the corresponding NCAM residues resulted in GPI-anchored proteins lacking the CEA-like biological functions of integrin modulation and differentiation blockage. Transferring this region from CEA into NCAM in conjunction with the upstream proline (PGLSAG) was sufficient to specify the addition of the CEA anchor. Therefore, this study identifies a novel specificity signal consisting of six amino acids located within the GPI anchor attachment signal, which is necessary and sufficient to specify the addition of a particular functional GPI anchor and, thereby, the ultimate function of the mature protein.


Assuntos
Antígeno Carcinoembrionário/química , Glicosilfosfatidilinositóis/química , Moléculas de Adesão de Célula Nervosa/química , Sequência de Aminoácidos , Animais , Células CHO , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/metabolismo , Adesão Celular , Diferenciação Celular , Linhagem Celular , Cricetinae , Cricetulus , Fibronectinas/metabolismo , Humanos , Dados de Sequência Molecular , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
6.
J Cell Physiol ; 211(3): 791-802, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17286276

RESUMO

CEA functions as an intercellular adhesion molecule and is up-regulated in a wide variety of human cancers, including colon, breast and lung. Its over-expression inhibits cellular differentiation, blocks cell polarization, distorts tissue architecture, and inhibits anoikis of many different cell types. Here we report results concerning the molecular mechanism involved in these biological effects, where relatively rapid molecular changes not requiring alterations in gene expression were emphasized. Confocal microscopy experiments showed that antibody-mediated clustering of a deletion mutant of CEA (DeltaNCEA), normally incapable of self binding and clustering, led to the co-localization of integrin alpha5beta1 with patches of DeltaNCEA on the cell surface. Activation of alpha5, as defined by an anti-alpha5 mAb-sensitive increase in cell adhesion to immobilized fibronectin, and an increased binding of soluble fibronectin to cells, was also observed. This was accompanied by the recruitment of integrin-linked kinase (ILK), protein kinase B (PKB/Akt), and the mitogen-activated protein kinase (MAPK) to membrane microdomains and the phosphorylation of Akt and MAPK. Inhibition of PI3-K and ILK, but not MAPK, prevented the alpha5beta1 integrin activation. Conversely, anti-alpha5 antibody inhibited the PI3-K-mediated activation of Akt, implying the involvement of outside-in and inside-out signaling in integrin activation. Therefore we propose that CEA-mediated signaling involves clustering of CEA and co-clustering and activation of the alpha5beta1 and associated specific signaling elements on the internal surfaces of membrane microdomains. These changes may represent a molecular mechanism for the biological effects of CEA.


Assuntos
Antígenos CD/metabolismo , Antígeno Carcinoembrionário/metabolismo , Moléculas de Adesão Celular/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Integrina alfa5beta1/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Agregação de Receptores/fisiologia , Animais , Anoikis/fisiologia , Anticorpos/farmacologia , Antígenos CD/imunologia , Células CHO , Células CACO-2 , Antígeno Carcinoembrionário/imunologia , Moléculas de Adesão Celular/imunologia , Diferenciação Celular/fisiologia , Cricetinae , Cricetulus , Proteínas Ligadas por GPI , Humanos , Integrina alfa2/imunologia , Integrina alfa2/metabolismo , Integrina alfa5beta1/imunologia , Microdomínios da Membrana/metabolismo , Mioblastos/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transfecção
7.
Mol Biol Cell ; 18(4): 1366-74, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17287394

RESUMO

GPI membrane anchors of cell surface glycoproteins have been shown to confer functional properties that are different from their transmembrane (TM)-anchored counterparts. For the human carcinoembryonic antigen (CEA) family, a subfamily of the immunoglobulin superfamily, conversion of the mode of membrane linkage from TM to GPI confers radical changes in function: from tumor suppression or neutrality toward inhibition of differentiation and anoikis and distortion of tissue architecture, thereby contributing to tumorigenesis. We show here that GPI anchorage in the CEA family evolved twice independently in primates, very likely from more primitive TM anchors, by different packages of mutations. Both mutational packages, one package found in many primates, including humans, and a second, novel package found only in the Cebidae radiation of New World monkeys, give rise to efficiently processed GPI-linked proteins. Both types of GPI anchors mediate inhibition of cell differentiation. The estimated rate of nonsynonymous mutations (Ka) in the anchor-determining domain for conversion from TM to GPI anchorage in the CEA family that were fixed during evolution in these primates is 7 times higher than the average Ka in primates, indicating positive selection. These results suggest therefore that the functional changes mediated by CEA GPI anchors, including the inhibition of differentiation and anoikis, could be adaptive and advantageous.


Assuntos
Antígeno Carcinoembrionário/fisiologia , Evolução Molecular , Glicosilfosfatidilinositóis/metabolismo , Primatas , Animais , Sequência de Bases , Antígeno Carcinoembrionário/química , Antígeno Carcinoembrionário/metabolismo , Testes de Carcinogenicidade , Diferenciação Celular , Humanos , Dados de Sequência Molecular , Mutação , Radiação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
8.
J Cell Physiol ; 210(3): 757-65, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17167768

RESUMO

Carcinoembryonic antigen (CEA) and CEA family member CEACAM6 are glycophosphatidyl inositol (GPI)-anchored, intercellular adhesion molecules that are up-regulated in a wide variety of human cancers, including colon, breast, and lung. When over-expressed in a number of cellular systems, these molecules are capable of inhibiting cellular differentiation and anoikis, as well as disrupting cell polarization and tissue architecture, thus increasing tumorigenicity. The present study shows that perturbation of the major fibronectin receptor, integrin alpha5beta1, underlies some of these biological effects. Using confocal microscopy and specific antibodies, CEA and CEACAM6 were demonstrated to co-cluster with integrin alpha5beta1 on the cell surface. The presence of CEA and CEACAM6 was shown to lead to an increase in the binding of the integrin alpha5beta1 receptor to its ligand fibronectin, without changing its cell surface levels, resulting in increased adhesion of CEA/CEACAM6-expressing cells to fibronectin. More tenacious binding of free fibronectin to cells led to enhanced fibronectin matrix assembly and the formation of a polymerized fibronectin "cocoon" around the cells. Disruption of this process with specific monoclonal antibodies against either fibronectin or integrin alpha5beta1 led to the restoration of cellular differentiation and anoikis in CEA/CEACAM6 producing cells.


Assuntos
Antígenos CD/fisiologia , Antígeno Carcinoembrionário/fisiologia , Moléculas de Adesão Celular/fisiologia , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Animais , Anoikis/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Cães , Matriz Extracelular/metabolismo , Proteínas Ligadas por GPI , Regulação da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Microscopia Confocal , Mioblastos/citologia , Ligação Proteica/fisiologia , Ratos
9.
J Cell Biol ; 175(4): 647-59, 2006 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-17101695

RESUMO

The functional specificity conferred by glycophosphatidylinositol (GPI) anchors on certain membrane proteins may arise from their occupancy of specific membrane microdomains. We show that membrane proteins with noninteractive external domains attached to the same carcinoembryonic antigen (CEA) GPI anchor, but not to unrelated neural cell adhesion molecule GPI anchors, colocalize on the cell surface, confirming that the GPI anchor mediates association with specific membrane domains and providing a mechanism for specific signaling. This directed targeting was exploited by coexpressing an external domain-defective protein with a functional protein, both with the CEA GPI anchor. The result was a complete loss of signaling capabilities (through integrin-ECM interaction) and cellular effect (differentiation blockage) of the active protein, which involved an alteration of the size of the microdomains occupied by the active protein. This work clarifies how the GPI anchor can determine protein function, while offering a novel method for its modulation.


Assuntos
Glicosilfosfatidilinositóis/antagonistas & inibidores , Glicosilfosfatidilinositóis/metabolismo , Animais , Anticorpos/imunologia , Células CHO , Antígeno Carcinoembrionário/química , Antígeno Carcinoembrionário/metabolismo , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Reagentes de Ligações Cruzadas/farmacologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Glicosilfosfatidilinositóis/química , Humanos , Integrinas/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Proteínas Mutantes/metabolismo , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Moléculas de Adesão de Célula Nervosa/química , Moléculas de Adesão de Célula Nervosa/metabolismo , Ligação Proteica/efeitos dos fármacos , Ratos
10.
Carcinogenesis ; 27(9): 1909-16, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16632476

RESUMO

Human carcinoembryonic antigen (CEA), a widely used clinical tumor marker, and its close relative, CEACAM6, are often overexpressed in many cancers. This correlation suggests a possible instrumental role in tumorigenesis, which is supported by extensive results obtained with several in vitro systems. The implication that these results could also apply in vivo warrants investigation. Since mice do not possess homologs of the glycophosphatidyl inositol (GPI)-anchored CEACAM family genes CEA, CEACAM6 and CEACAM7, we have constructed transgenic mice harboring a 187 kb portion of the human CEACAM family gene locus contained in a bacterial artificial chromosome (CEABAC) that includes genes coding for CEA, CEACAM6 and CEACAM7. In this study, we treated the CEABAC mice and their wild-type littermates with azoxymethane (AOM) in order to induce colon tumor formation. At 20 weeks post-treatment, the CEABAC transgenics showed more than a 2-fold increase in mean tumor load relative to their wild-type littermates. Cell surface expression of CEA and CEACAM6 increased by 2- and 20-fold, respectively, in colonocytes from the tumors relative to colonocytes from non-AOM treated transgenics and a de-regulated spatial pattern of CEA/CEACAM6 expression was found in 'normal' crypts adjacent to the tumors, thus mimicking closely the situation in human colon tumorigenesis. A modestly increased incidence of beta-catenin mutations also observed in the AOM-induced CEABAC tumors. These results show that expression of the human GPI-anchored CEACAM family genes predisposes mice to acquire and/or retain essential mutations necessary for sporadic colon tumor development.


Assuntos
Azoximetano , Carcinógenos , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Animais , Antígenos CD/genética , Antígeno Carcinoembrionário , Moléculas de Adesão Celular/genética , Proteínas Ligadas por GPI , Predisposição Genética para Doença , Glicosilfosfatidilinositóis/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Mutação , beta Catenina/metabolismo
11.
Mol Ther ; 9(6): 775-85, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15194045

RESUMO

Many novel cancer therapies, including immunotherapy and gene therapy, are specifically targeted to tumor-associated molecules, among which carcinoembryonic antigen (CEA) represents a popular example. Discrepancies between preclinical experimental data in animal models and clinical outcome in terms of therapeutic response and toxicity, however, often arise. Preclinical testing can be compromised by the lack of CEA and other closely related human CEA family members in rodents, which lack analogous genes for most human CEA family members. Here, we report the construction of a transgenic mouse with a 187-kb human bacterial artificial chromosome (CEABAC) that contains part of the human CEA family gene cluster including complete human CEA (CEACAM5), CEACAM3, CEACAM6, and CEACAM7 genes. The spatiotemporal expression pattern of these genes in the CEABAC mice was found to be remarkably similar to that of humans. This novel mouse will ensure better assessment than previously utilized models for the preclinical testing of CEA-targeted therapies and perhaps allow the testing of CEACAM6, which is overexpressed in many solid tumors and leukemias, as a therapeutic target. Moreover, expression of CEA family genes in gastrointestinal, breast, hematopoietic, urogenital, and respiratory systems could facilitate other clinical applications, such as the development of therapeutic agents against Neisseria gonorrhoeae infections, which use CEA family members as major receptors.


Assuntos
Antígeno Carcinoembrionário/genética , Cromossomos Artificiais Bacterianos/genética , Ensaios de Seleção de Medicamentos Antitumorais , Camundongos Transgênicos/genética , Modelos Animais , Animais , Antígenos CD , Antígenos de Neoplasias/análise , Antígenos de Neoplasias/genética , Antígeno Carcinoembrionário/análise , Moléculas de Adesão Celular/análise , Moléculas de Adesão Celular/genética , Proteínas Ligadas por GPI , Expressão Gênica , Humanos , Camundongos , Família Multigênica/genética , Distribuição Tecidual
12.
J Cell Sci ; 117(Pt 5): 761-9, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14734654

RESUMO

GPI anchorage in the CEA family results in the acquisition of radically different functions relative to TM anchorage, including inhibition of differentiation and anoikis, disruption of tissue architecture and promotion of tumorigenicity. CEA GPI anchors, as determined by the carboxy-terminal exon of CEA, demonstrate biological specificity in their ability to confer these functional changes. CEA family GPI anchorage appears to have evolved twice independently during the primate radiation, in a manner suggestive of evolution from more primitive TM-anchored CEACAM1. We show here that very few mutations in the TM exon of present-day human CEACAM1 are required to give efficient GPI anchorage and the biological specificity of CEA GPI anchors, i.e., to give the differentiation-blocking function of GPI-anchored CEA. Such a change in anchorage could therefore represent a relatively facile means for producing radical change in molecular function of Ig superfamily members during evolution.


Assuntos
Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Imunoglobulinas/química , Mutação/genética , Animais , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciação/química , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Sequência de Bases , Antígeno Carcinoembrionário/química , Adesão Celular , Moléculas de Adesão Celular , Diferenciação Celular , Linhagem Celular , Cricetinae , Cricetulus , Citometria de Fluxo , Glicosilfosfatidilinositóis/química , Glicosilfosfatidilinositóis/genética , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida/genética , Fosfatidilinositol Diacilglicerol-Liase/metabolismo , Processamento de Proteína Pós-Traducional , Ratos , Análise de Sequência
13.
J Biol Chem ; 278(17): 14632-9, 2003 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-12571231

RESUMO

The external domains of Ig superfamily members are involved in multiple binding interactions, both homophilic and heterophilic, that initiate molecular events leading to the execution of diverse cell functions. Human carcinoembryonic antigen (CEA), an Ig superfamily cell surface glycoprotein used widely as a clinical tumor marker, undergoes homophilic interactions that mediate intercellular adhesion. Recent evidence supports the view that deregulated overexpression of CEA has an instrumental role in tumorigenesis through the inhibition of cell differentiation and the disruption of tissue architecture. The CEA-mediated block of the myogenic differentiation of rat L6 myoblasts depends on homophilic binding of its external domains. We show here that L6 transfectant cells expressing CEA can "trans-block" the myogenesis of juxtaposed differentiation-competent L6 transfectant cells expressing a deletion mutant of CEA (DeltaNCEA). This result implies the efficacy of antiparallel CEA-CEA interactions between cells in the differentiation block. In addition, DeltaNCEA can acquire differentiation blocking activity by cross-linking with specific anti-CEA antibodies, thus implying the efficacy of parallel CEA-CEA interactions on the same cell surface. The myogenic differentiation blocking activity of CEA was demonstrated by site-directed mutations to involve three subdomains of the amino-terminal domain, shown previously to be critical for its intercellular adhesion function. Monovalent Fab fragments of monoclonal antibodies binding to the region bridging subdomains 1 and 2 could both inhibit intercellular adhesion and release the myogenic differentiation block. Amino acid substitutions Q80A, Q80R, and D82N in subdomain 3, QNDTG, however, were found to completely ablate the differentiation blocking activity of CEA but had no effect on intercellular adhesion activity. A cyclized peptide representing this subdomain was the most effective at releasing the differentiation block.


Assuntos
Antígenos de Neoplasias/fisiologia , Antígeno Carcinoembrionário/fisiologia , Moléculas de Adesão Celular/fisiologia , Substituição de Aminoácidos , Animais , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígeno Carcinoembrionário/química , Antígeno Carcinoembrionário/genética , Adesão Celular , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Transformada , Humanos , Fragmentos Fab das Imunoglobulinas/farmacologia , Imunoglobulinas , Peptídeos Cíclicos/farmacologia , Estrutura Terciária de Proteína , Ratos , Transfecção
14.
Neoplasia ; 4(2): 151-63, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11896570

RESUMO

Human carcinoembryonic antigen (CEA) and the CEA family member CEACAM6 (formerly nonspecific cross-reacting antigen [NCA]) function in vitro, at least, as homotypic intercellular adhesion molecules and, in model systems, can block the terminal differentiation and anoikis of several different cell types. We have recently demonstrated that the increased cell surface levels of CEA and CEACAM6 in purified human colonocytes from freshly excised, well to poorly differentiated colon carcinomas are inversely correlated with the degree of cellular differentiation. Thus, deregulated expression of CEA/CEACAM6 could directly contribute to colon tumorigenesis by the inhibition of terminal differentiation and anoikis. Evidence against this view includes the common observation of increased CEA/CEACAM6 expression as normal colonocytes differentiate in their migration up colonic crypt walls. We report here the direct effects of deregulated overexpression of CEA/CEACAM6, at levels observed in colorectal carcinomas, on the differentiation of two human colonic cell lines, SW-1222 and Caco-2. Stable transfectants of both of these cell lines that constitutively express 10- to 30-fold higher cell surface levels of CEA/CEACAM6 than endogenous levels failed to polarize and differentiate into glandular structures in monolayer or 3D culture or to form colonic crypts in a tissue architecture assay in nude mice. In addition, these transfectants were found to exhibit increased tumorigenicity in nude mice. These results thus support the contention that deregulated overexpression of CEA and CEACAM6 could provide a tumorigenic contribution to colon carcinogenesis.


Assuntos
Antígenos de Neoplasias/metabolismo , Antígeno Carcinoembrionário/metabolismo , Moléculas de Adesão Celular , Colo/metabolismo , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Glicoproteínas de Membrana/metabolismo , Animais , Antígenos CD , Antígenos de Neoplasias/genética , Antígeno Carcinoembrionário/genética , Diferenciação Celular , Células Cultivadas/metabolismo , Colágeno/metabolismo , Colo/citologia , Progressão da Doença , Citometria de Fluxo , Proteínas Ligadas por GPI , Humanos , Técnicas Imunoenzimáticas , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...