Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Biol ; 115(1): 179-90, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1918135

RESUMO

Using histochemical detection, we have visualized in situ the complete metabolic pathway for the degradation of purine nucleotides. From the tongue to the ileum, diverse epithelial cell types lining the lumen of the mouse gastrointestinal (GI) tract strongly coexpress each of the five key purine catabolic enzymes. Dramatic increases in the expression of each enzyme occurred during postnatal maturation of the GI tract. Using in situ hybridization, an intense accumulation of adenosine deaminase (ADA) mRNA was detected only within GI epithelial cells undergoing postmitotic differentiation. In a similar manner, at the developing maternal-fetal interface, high level expression of the purine catabolic pathway also occurred in a unique subset of maternal decidual cells previously known to express high levels of alkaline phosphatase and ADA. This induction occurred almost immediately after implantation in the periembryonic maternal decidual cells, shortly thereafter in antimesometrial decidual cells, and later in cells of the placental decidua basalis: all of which contain cell types thought to be undergoing programmed cell death. The expression of the pathway at the site of embryo implantation appears to be critical because its pharmacologic inhibition during pregnancy has been found to be embryolethal or teratogenic. Purine destruction at these nutritional interfaces (placenta and gastrointestinal tract) seem to override any potential economy of purine salvage, and may represent biochemical adaptation to nucleic acid breakdown occurring in the context of dietary digestion or extensive programmed cell death.


Assuntos
Adenosina Desaminase/metabolismo , Decídua/fisiologia , Fenômenos Fisiológicos do Sistema Digestório , Purinas/metabolismo , 5'-Nucleotidase/metabolismo , Adenosina Desaminase/genética , Animais , Decídua/anatomia & histologia , Sistema Digestório/anatomia & histologia , Desenvolvimento Embrionário , Feminino , Expressão Gênica , Guanina Desaminase/metabolismo , Humanos , Camundongos , Microvilosidades/enzimologia , Hibridização de Ácido Nucleico , Gravidez , Purina-Núcleosídeo Fosforilase/metabolismo , RNA Mensageiro/genética , Xantina Oxidase/metabolismo
2.
J Thromb Haemost ; 16(11): 2258-2269, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30152921

RESUMO

Essentials Protease activated receptor-1 (PAR-1) has been proposed to drive cancer progression. Surprisingly, PAR-1 deletion accelerated tumor progression in two distinct experimental settings. PAR-1 deletion was shown to limit the apoptosis of transformed epithelial cells. Thrombin- and activated protein C-mediated PAR-1 activation have unique effects on tumor cell biology. SUMMARY: Background Multiple studies have implicated protease-activated receptor-1 (PAR-1), a G-protein-coupled receptor activated by proteolytic cleavage of its N-terminus, as one target coupling thrombin-mediated proteolysis to tumor progression. Objective To analyze the role of PAR-1 in the setting of two distinct spontaneously developing tumor models in mice. Methods We interbred PAR-1-deficient mice with Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mice, which spontaneously develop prostate tumors, and adenomatous polyposis coli Min (APCMin/+ ) mice, which spontaneously develop intestinal adenomas. Results Analyses of TRAMP mice with advanced disease (30 weeks) revealed that PAR-1 deficiency resulted in significantly larger and more aggressive prostate tumors. Prostates collected at an earlier time point (12 weeks of age) revealed that PAR-1 promotes apoptosis in transformed epithelia. In vitro analyses of TRAMP-derived cells revealed that activated protein C-mediated PAR-1 cleavage can induce tumor cell apoptosis, suggesting that tumor cell-intrinsic PAR-1 functions can limit tumor progression. Paralleling results in TRAMP mice, PAR-1-deficient APCMin/+ mice developed three-fold more adenomas than PAR-1-expressing mice, and the adenomas that formed were significantly larger. Moreover, loss of PAR-1 expression was shown to limit apoptosis in transformed intestinal epithelial cells. Conclusions Together, these results demonstrate a previously unrecognized role for PAR-1 in impeding tumor progression in vivo. These results also offer a cautionary note suggesting that long-term PAR-1 inhibition could increase malignancy risk in some contexts.


Assuntos
Progressão da Doença , Neoplasias Intestinais/metabolismo , Neoplasias da Próstata/metabolismo , Receptor PAR-1/metabolismo , Animais , Apoptose , Transformação Celular Neoplásica , Cruzamentos Genéticos , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tumores Neuroendócrinos/metabolismo , Neoplasias da Próstata/genética , Proteína C/metabolismo , Trombina/metabolismo
3.
J Clin Invest ; 92(3): 1451-8, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8376597

RESUMO

Complement activation is associated with a variety of immunologically-mediated renal diseases. Proximal tubular epithelial cells in situ constitutively express messenger RNA for C4 of the complement system. These same epithelial cells in culture have been reported to contain message for C3 and to secrete this protein when stimulated by IL-2. The present study compared the in situ localization of C3 and C4 message in parallel in a variety of renal biopsy and nephrectomy specimens. All adequate tissue samples (n = 23) had C4 mRNA throughout in the cortical tubular epithelium. Although C3 message was also expressed in tubular epithelial cells, there was much greater variation in its distribution. mRNA for C3 was not detected in histologically normal specimens (n = 4) either by in situ or Northern hybridization. Focal C3 message correlated with focal histologic abnormalities (e.g., focal glomerulosclerosis), while more generalized C3 signal occurred in specimens with more diffuse inflammatory processes (e.g., SLE). Infiltrating inflammatory cells and cells of the glomeruli were uniformly negative for C3 (and C4) message. Tubular C3 and C4 mRNA appeared to be translated, since selected specimens showed cytoplasmic staining by monoclonal antibodies to C3c and C4c. These observations are consistent with the hypothesis that local production of inflammatory mediators could induce C3 synthesis in the renal interstitium, with the possibility that subsequent complement activation could enhance the pathogenic process.


Assuntos
Complemento C3/metabolismo , Complemento C4/metabolismo , Rim/metabolismo , Complemento C3/genética , Complemento C4/genética , Expressão Gênica , Humanos , Hibridização In Situ , Rim/anatomia & histologia , RNA Mensageiro/genética
4.
J Clin Invest ; 101(5): 1184-94, 1998 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-9486990

RESUMO

A critical role of the coagulation system in the development of atherosclerosis has been frequently postulated based on a variety of indirect observations, including the expression of procoagulants and fibrinolytic factors within atherosclerotic vessels, the presence of substantial amounts of fibrin(ogen) and fibrin degradation products within intimal lesions, the cellular infiltration and assimilation of mural thrombi into developing plaques, and the identification of high plasma fibrinogen (Fib) levels as an independent risk factor for the development of ischemic heart disease. To directly examine the role of fibrin(ogen) in atherogenesis, Fib-deficient mice were crossed to atherosclerosis-prone apolipoprotein E (apo E)-deficient mice. Both apo E-/- and apo E-/-/Fib-/- mice developed lesions throughout the entire aortic tree, ranging in appearance from simple fatty streaks to complex fibrous plaques. Furthermore, remarkably little difference in lesion size and complexity was observed within the aortae of age- and gender-matched apo E-/- and apo E-/-/Fib-/- mice. These results indicate that the contribution of fibrin(ogen) to intimal mass and local cell adhesion, migration, and proliferation is not strictly required for the development of advanced atherosclerotic disease in mice with a severe defect in lipid metabolism.


Assuntos
Afibrinogenemia/complicações , Arteriosclerose/genética , Arteriosclerose/metabolismo , Fibrinogênio/genética , Fibrinogênio/metabolismo , Alelos , Animais , Aorta/patologia , Apolipoproteínas E/genética , Arteriosclerose/patologia , Fibrina/metabolismo , Fibrina/fisiologia , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Músculo Liso/citologia , Reação em Cadeia da Polimerase
5.
J Clin Invest ; 103(9): 1277-85, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10225971

RESUMO

The Ron/STK receptor tyrosine kinase is a member of the c-Met family of receptors and is activated by hepatocyte growth factor-like protein (HGFL). Ron activation results in a variety of cellular responses in vitro, such as activation of macrophages, proliferation, migration, and invasion, suggesting a broad biologic role in vivo. Nevertheless, HGFL-deficient mice grow to adulthood with few appreciable phenotypic abnormalities. We report here that in striking contrast to the loss of its only known ligand, complete loss of Ron leads to early embryonic death. Embryos that are devoid of Ron (Ron-/-) are viable through the blastocyst stage of development but fail to survive past the peri-implantation period. In situ hybridization analysis demonstrates that Ron is expressed in the trophectoderm at embryonic day (E) 3.5 and is maintained in extraembryonic tissue through E7.5, compatible with an essential function at this stage of development. Hemizygous mice (Ron+/-) grow to adulthood; however, these mice are highly susceptible to endotoxic shock and appear to be compromised in their ability to downregulate nitric oxide production. These results demonstrate a novel role for Ron in early mouse development and suggest that Ron plays a limiting role in the inflammatory response.


Assuntos
Implantação do Embrião , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Sequência de Bases , Primers do DNA , Desenvolvimento Embrionário e Fetal , Feminino , Morte Fetal/genética , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Hibridização In Situ , Camundongos , Óxido Nítrico/fisiologia , Receptores Proteína Tirosina Quinases/genética , Receptores de Superfície Celular/genética , Choque Séptico/genética , Células-Tronco/metabolismo
6.
J Clin Invest ; 106(9): 1105-13, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11067863

RESUMO

Apolipoprotein J/clusterin (apoJ/clusterin), an intriguing protein with unknown function, is induced in myocarditis and numerous other inflammatory injuries. To test its ability to modify myosin-induced autoimmune myocarditis, we generated apoJ-deficient mice. ApoJ-deficient and wild-type mice exhibited similar initial onset of myocarditis, as evidenced by the induction of two early markers of the T cell-mediated immune response, MHC-II and TNF receptor p55. Furthermore, autoantibodies against the primary antigen cardiac myosin were induced to the same extent. Although the same proportion of challenged animals exhibited some degree of inflammatory infiltrate, inflammation was more severe in apoJ-deficient animals. Inflammatory lesions were more diffuse and extensive in apoJ-deficient mice, particularly in females. In marked contrast to wild-type animals, the development of a strong generalized secondary response against cardiac antigens in apoJ-deficient mice was predictive of severe myocarditis. Wild-type mice with a strong Ab response to secondary antigens appeared to be protected from severe inflammation. After resolution of inflammation, apoJ-deficient, but not wild-type, mice exhibited cardiac function impairment and severe myocardial scarring. These results suggest that apoJ limits progression of autoimmune myocarditis and protects the heart from postinflammatory tissue destruction.


Assuntos
Doenças Autoimunes/etiologia , Glicoproteínas/fisiologia , Chaperonas Moleculares , Miocardite/etiologia , Animais , Antígenos CD/biossíntese , Autoanticorpos/biossíntese , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Sequência de Bases , Clusterina , Primers do DNA/genética , Feminino , Glicoproteínas/deficiência , Glicoproteínas/genética , Antígenos de Histocompatibilidade Classe II/biossíntese , Masculino , Camundongos , Camundongos Knockout , Miocardite/imunologia , Miocardite/patologia , Miosinas/imunologia , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores Tipo I de Fatores de Necrose Tumoral , Linfócitos T/imunologia
7.
Mol Cell Biol ; 15(10): 5707-15, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7565722

RESUMO

Locus control regions (LCRs) are powerful assemblies of cis elements that organize the actions of cell-type-specific trans-acting factors. A 2.3-kb LCR in the human adenosine deaminase (ADA) gene first intron, which controls expression in thymocytes, is composed of a 200-bp enhancer domain and extended flanking sequences that facilitate activation from within chromatin. Prior analyses have demonstrated that the enhancer contains a 28-bp core region and local adjacent augmentative cis elements. We now show that the core contains a single critical c-Myb binding site. In both transiently cotransfected human cells and stable chromatin-integrated yeast cells, c-Myb strongly transactivated reporter constructs that contained polymerized core sequences. c-Myb protein was strongly evident in T lymphoblasts in which the enhancer was active and was localized within discrete nuclear structures. Fetal murine thymus exhibited a striking concordance of endogenous c-myb expression with that of mouse ADA and human ADA LCR-directed transgene expression. Point mutation of the c-Myb site within the intact 2.3-kb LCR severely attenuated enhancer activity in transfections and LCR activity in transgenic thymocytes. Within the context of a complex enhancer and LCR, c-Myb can act as an organizer of thymocyte-specific gene expression via a single binding site.


Assuntos
Elementos Facilitadores Genéticos/genética , Proteínas Proto-Oncogênicas/metabolismo , Timo/fisiologia , Transativadores/metabolismo , Adenosina Desaminase/genética , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Íntrons/genética , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação Puntual , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas c-myb , Saccharomyces cerevisiae/genética , Linfócitos T , Timo/citologia , Timo/enzimologia , Transativadores/biossíntese
8.
Mol Cell Biol ; 15(2): 1123-35, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7823928

RESUMO

Using transgenic mice, we have defined novel gene regulatory elements, termed "facilitators." These elements bilaterally flank, by up to 1 kb, a 200-bp T-cell-specific enhancer domain in the human adenosine deaminase (ADA) gene. Facilitators were essential for gene copy-proportional and integration site-independent reporter expression in transgenic thymocytes, but they had no effect on the enhancer in transfected T cells. Both segments were required. Individual segments had no activity. A lack of facilitator function caused positional susceptibility and prevented DNase I-hypersensitive site formation at the enhancer. The segments were required to be at opposed ends of the enhancer, and they could not be grouped together. Reversing the orientation of a facilitator segment caused a partial loss of function, suggesting involvement of a stereospecific chromatin structure. trans-acting factor access to enhancer elements was modeled by exposing nuclei to a restriction endonuclease. The enhancer domain was accessible to the 4-cutter DpnII in a tissue- and cell-type-specific fashion. However, unlike DNase I hypersensitivity and gene expression, accessibility to the endonuclease could occur without the facilitator segments, suggesting that an accessible chromatin domain is an intermediate state in the activational pathway. These results suggest that facilitators (i) are distinct from yet positionally constrained to the enhancer, (ii) participate in a chromatin structure transition that is necessary for the DNase I hypersensitivity and the transcriptional activating function of the enhancer, and (iii) act after cell-type-specific accessibility to the enhancer sequences is established by factors that do not require the facilitators to be present.


Assuntos
Adenosina Desaminase/genética , Elementos Facilitadores Genéticos , Expressão Gênica , Adenosina Desaminase/biossíntese , Animais , Núcleo Celular/metabolismo , Cloranfenicol O-Acetiltransferase/biossíntese , Cloranfenicol O-Acetiltransferase/metabolismo , Cromatina/fisiologia , Cruzamentos Genéticos , Desoxirribonuclease I , Desoxirribonucleases de Sítio Específico do Tipo II , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Genéticos , Especificidade de Órgãos , RNA Mensageiro/análise , Sequências Reguladoras de Ácido Nucleico , Mapeamento por Restrição , Timo/enzimologia
9.
Oncogene ; 18(4): 1103-11, 1999 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-10023687

RESUMO

The c-myb gene encodes a sequence specific transactivator that is required for fetal hematopoiesis, but its potential role in other tissues is less clear because of the early fetal demise of mice with targeted deletions of the c-myb gene and incomplete of knowledge about c-myb's expression pattern. In the hematopoietic system, c-Myb protein acts on target genes whose expression is restricted to individual lineages, despite Myb's presence and role in multiple immature lineages. This suggests that c-Myb actions within different cell type-specific contexts are strongly affected by combinatorial interactions. To consider the possibility of similar c-Myb actions could extend into non-hematopoietic systems in other cell and tissue compartments, we characterized c-myb expression in developing and adult mice using in situ hybridization and correlated this with stage-specific differentiation and mitotic activity. Diverse tissues exhibited strong c-myb expression during development, notably tooth buds, the thyroid primordium, developing trachea and proximal branching airway epithelium, hair follicles, hematopoietic cells, and gastrointestinal crypt epithelial cells. The latter three of these all maintained high expression into adulthood, but with characteristic restriction to immature cell lineages prior to their terminal differentiation. In all sites, during fetal and adult stages, loss of c-Myb expression correlated strikingly with the initiation of terminal differentiation, but not the loss of mitotic activity. Based on these data, we hypothesize that c-Myb's function during cellular differentiation is both an activator of immature gene expression and a suppressor of terminal differentiation in diverse lineages.


Assuntos
Diferenciação Celular/fisiologia , Desenvolvimento Embrionário e Fetal/genética , Oncogenes , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Fatores Etários , Animais , Sistema Digestório/embriologia , Folículo Piloso/embriologia , Hibridização In Situ , Fígado/embriologia , Camundongos , Mitose , Odontogênese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myb , Timo/embriologia , Glândula Tireoide/embriologia , Traqueia/embriologia , Transativadores/genética , Transativadores/metabolismo
10.
Oncogene ; 14(18): 2175-88, 1997 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-9174053

RESUMO

Anaplastic Lymphoma Kinase (ALK) was originally identified as a member of the insulin receptor subfamily of receptor tyrosine kinases that acquires transforming capability when truncated and fused to nucleophosmin (NPM) in the t(2;5) chromosomal rearrangement associated with non-Hodgkin's lymphoma, but further insights into its normal structure and function are lacking. Here, we characterize a full-length normal human ALK cDNA and its product, and determine the pattern of expression of its murine homologue in embryonic and adult tissues as a first step toward the functional assessment of the receptor. Analysis of the 6226 bp ALK cDNA identified an open reading frame encoding a 1620-amino acid (aa) protein of predicted mass approximately 177 kDa that is most closely related to leukocyte tyrosine kinase (LTK), the two exhibiting 57% aa identity and 71% similarity over their region of overlap. Biochemical analysis demonstrated that the approximately 177 kDa ALK polypeptide core undergoes co-translational N-linked glycosylation, emerging in its mature form as a 200 kDa single chain receptor. Surface labeling studies indicated that the 200 kDa glycoprotein is exposed at the cell membrane, consistent with the prediction that ALK serves as the receptor for an unidentified ligand(s). In situ hybridization studies revealed Alk expression beginning on embryonic day 11 and persisting into the neonatal and adult periods of development. Alk transcripts were confined to the nervous system and included several thalamic and hypothalamic nuclei; the trigeminal, facial, and acoustic cranial ganglia; the anterior horns of the spinal cord in the region of the developing motor neurons; the sympathetic chain; and the ganglion cells of the gut. Thus, ALK is a novel orphan receptor tyrosine kinase that appears to play an important role in the normal development and function of the nervous system.


Assuntos
Cromossomos Humanos Par 2 , Linfoma não Hodgkin/genética , Fenômenos Fisiológicos do Sistema Nervoso , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Sequência de Aminoácidos , Quinase do Linfoma Anaplásico , Animais , Clonagem Molecular , DNA Complementar , Regulação da Expressão Gênica no Desenvolvimento , Glicosilação , Humanos , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , Músculo Esquelético/patologia , Sistema Nervoso/embriologia , Receptores Proteína Tirosina Quinases/genética , Rabdomiossarcoma/genética , Rabdomiossarcoma/patologia , Análise de Sequência , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Transcrição Gênica , Células Tumorais Cultivadas
11.
Oncogene ; 16(24): 3097-104, 1998 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-9671388

RESUMO

To investigate the role of plasmin(ogen) in mammary tumor development and progression, plasminogen-deficient mice were crossed with transgenic mice expressing Polyoma middle T antigen under the control of the mouse mammary tumor virus long terminal repeat. Virgin females carrying the Polyoma middle T antigen uniformly developed multiple, bilateral mammary tumors, regardless of the presence or absence of circulating plasminogen. Both the age at which these tumors became palpable and subsequent tumor growth were indistinguishable between plasminogen-deficient mice and plasminogen-expressing littermates. However, plasminogen was found to greatly modify the metastatic potential in this model system; lung metastasis in plasminogen-deficient mice was significantly reduced as compared to littermate controls with respect to frequency of occurrence, total number of metastases, and total metastatic tumor burden. Plasminogen activators, as well as other key factors that govern the conversion of plasminogen to plasmin, were expressed within the mammary tumors, suggesting that the plasminogen/plasmin system may promote metastasis by contributing to tumor-associated extracellular proteolysis. The data provide direct evidence that plasmin(ogen) is a tumor progression factor in PymT-induced mammary cancer, and support the hypothesis that hemostatic factors play an important role in tumor biology.


Assuntos
Antígenos Transformantes de Poliomavirus/fisiologia , Neoplasias Pulmonares/patologia , Neoplasias Mamárias Experimentais/patologia , Plasminogênio/genética , Animais , Sequência de Bases , Northern Blotting , Primers do DNA , Feminino , Imuno-Histoquímica , Hibridização In Situ , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/etiologia , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Transgênicos , Inibidor 1 de Ativador de Plasminogênio/genética , Ativador de Plasminogênio Tipo Uroquinase/genética
12.
Biochim Biophys Acta ; 1136(3): 272-82, 1992 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-1387801

RESUMO

The CHRF-288-11 cell line has been previously shown to exhibit properties consistent with a megakaryocytic origin. The response of these cells to thrombin has now been investigated. Thrombin treatment of CHRF-288-11 cells results in both an increase in intracellular free calcium levels and secretion of mitogenic activity and beta-thromboglobulin. Cell viability is not affected. The mitogenic activity released from the cells is due primarily to the presence of basic fibroblast growth factor. Immunohistochemical data indicate a packaging of basic fibroblast growth factor into granular structures. Trypsin and phorbol 12-myristate 13-acetate also initiate release of mitogenic activity from this cell line, whereas under non-stirred conditions collagen and ADP do not. Through measurements of intracellular calcium levels it was determined that thrombin pretreatment of cells ablates a further response to thrombin, but does not block an increase in intracellular calcium levels due to trypsin. This suggests that these two agonists may act through different mechanisms. The thrombin-induced release reaction is inhibited almost completely by the reagents hirudin and dipyridamole, and only partially by indomethacin. These data indicate that the CHRF-288-11 cell line should provide an excellent model system in which to study the packaging of factors into granules which undergo regulated release.


Assuntos
Cálcio/metabolismo , Megacariócitos/efeitos dos fármacos , Mitógenos/metabolismo , Trombina/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Fator 2 de Crescimento de Fibroblastos/análise , Humanos , Megacariócitos/metabolismo , Modelos Biológicos , Fator Plaquetário 4/análise , Acetato de Tetradecanoilforbol/farmacologia , Trombina/antagonistas & inibidores , Fator de Crescimento Transformador beta/análise , Tripsina/farmacologia , beta-Tromboglobulina/análise
13.
Mech Dev ; 50(2-3): 177-86, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7619729

RESUMO

A novel murine dispersed homeobox gene, designated Gsh-2, is described. Analysis of cDNA sequence, including the full open reading frame, reveals an encoded homeodomain that is surprisingly similar to those of the Antennapedia-type clustered Hox genes. In addition, the encoded protein includes polyhistidine and polyalanine tracts, as observed for several other genes of developmental significance. In situ hybridizations showed Gsh-2 expression in the developing central nervous system, including the ganglionic eminences of the forebrain, the diencephalon, which gives rise to the thalamus and hypothalamus, and in the hindbrain. Furthermore, a random oligonucleotide selection and PCR amplification procedure was used to define a target DNA binding sequence, CNAATTAG, as a first step towards the identification of downstream target genes.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes Homeobox , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Camundongos , Dados de Sequência Molecular
14.
Mech Dev ; 88(2): 237-41, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10534623

RESUMO

Here we describe the cloning and expression pattern of a new bHLH-PAS domain gene, Npas3. Npas3 shares 50.2% amino acid sequence identity with Npas1 and a lesser similarity with other members of the bHLH-PAS domain family of transcription factors. Northern blot analysis detected Npas3 mRNA between 11.5 and 17.5 d.p.c. in embryonic development and exclusively in the adult brain. Whole-mount and section in situ hybridization assays revealed expression of Npas3 between 9.5 and 11.5 d.p.c. in the developing neural tube. In addition, Npas3 mRNA was expressed throughout the neuroepithelium of the developing central nervous system between 10. 5 and 12.5 d.p.c. Interestingly, at 14.5 d.p.c., the expression of Npas3 mRNA became restricted to the neopallial layer of the cortex. At 12.5 d.p.c., Npas3 mRNA was evident in nonneural tissues such as the developing dermis and mesenchyme surrounding the otic and nasal placodes. Expression was also detected in the developing cardiac valves, limb and developing kidney.


Assuntos
Sequências Hélice-Alça-Hélice/genética , Proteínas do Tecido Nervoso/genética , Sistema Nervoso/embriologia , Sistema Nervoso/crescimento & desenvolvimento , Fatores de Transcrição/genética , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Northern Blotting , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Epitélio/embriologia , Etiquetas de Sequências Expressas , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Mesoderma/metabolismo , Camundongos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
15.
Exp Hematol ; 29(12): 1392-402, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11750097

RESUMO

OBJECTIVE: The aim of this study was to identify granulocyte-macrophage colony-stimulating factor (GM-CSF) responsive genes. MATERIALS AND METHODS: Potential GM-CSF responsive genes were identified by comparing the mRNA expression pattern of the murine myeloid cell line PGMD1 grown in either interleukin-3 (IL-3) or GM-CSF by differential display. Human and murine cDNA clones of one of the bands having increased expression in GM-CSF were isolated. mRNA expression of the gene was examined by Northern blot. Immunohistochemistry and studies with a green fluorescent fusion protein were used to determine its intracellular location. Growth factor-stimulated proliferation of PGMD1 cells transfected with constitutively expressed sense and anti-sense cDNA constructs of the gene was measured by 3H-thymidine incorporation. RESULTS: A gene, named Magmas (mitochondria-associated granulocyte macrophage CSF signaling molecule), was shown to be rapidly induced when cells were switched from IL-3 to GM-CSF. Analysis of the amino acid sequence of Magmas showed it contained a mitochondrial signal peptide, but not any other known functional domains. The human and murine clones encode nearly identical 13-kDa proteins that localized to the mitochondria. Magmas mRNA expression was observed in all tissues examined. PGMD1 cells that overexpressed Magmas proliferated similarly to untransfected cells when cultured in IL-3 or GM-CSF. In contrast, cells with reduced protein levels grew normally in IL-3, but had impaired proliferation in GM-CSF. CONCLUSION: Magmas is a mitochondrial protein involved in GM-CSF signal transduction.


Assuntos
Fatores Estimuladores de Colônias/fisiologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Biblioteca Gênica , Humanos , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/genética , Alinhamento de Sequência
16.
Transplant Proc ; 47(7): 2239-42, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26361688

RESUMO

BACKGROUND: Long-term outcomes of kidney transplantation with organs from donors with disseminated intravascular coagulation (DIC) are comparable with those from other deceased donors. The use of tranexamic acid to impair fibrinolysis in the treatment of DIC is becoming increasingly frequent, particularly in the trauma setting. However, the effects of tranexamic acid on a transplanted kidney allograft are unknown. RESULTS: We report 2 cases of kidney transplantation following administration of tranexamic acid to the donor prior to organ donation. Microthrombi were present in the renal allografts. Both recipients experienced clinically significant hemolytic anemia, which typically occurs at a very low frequency. CONCLUSIONS: These cases illustrate a potential concern for the use of tranexamic acid in deceased kidney donors with DIC.


Assuntos
Anemia Hemolítica/diagnóstico , Coagulação Intravascular Disseminada/diagnóstico , Glomerulonefrite/cirurgia , Falência Renal Crônica/cirurgia , Trombocitopenia/diagnóstico , Ácido Tranexâmico/uso terapêutico , Idoso , Anemia Hemolítica/complicações , Antifibrinolíticos/efeitos adversos , Antifibrinolíticos/uso terapêutico , Diabetes Mellitus Tipo 2/complicações , Coagulação Intravascular Disseminada/etiologia , Feminino , Fibrinólise , Glomerulonefrite/complicações , Humanos , Hipertensão/complicações , Rim/efeitos dos fármacos , Falência Renal Crônica/complicações , Transplante de Rim , Masculino , Pessoa de Meia-Idade , Trombocitopenia/complicações , Doadores de Tecidos , Ácido Tranexâmico/efeitos adversos , Transplante Homólogo , Adulto Jovem
17.
Oncogene ; 34(7): 868-77, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-24608431

RESUMO

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common malignancy worldwide, and patient outcomes using current treatments remain poor. Tumor development is etiologically associated with tobacco or alcohol use and/or human papillomavirus (HPV) infection. HPV-positive HNSCCs, which frequently harbor wild-type p53, carry a more favorable prognosis and are a biologically distinct subgroup when compared with their HPV-negative counterparts. HPV E7 induces expression of the human DEK gene, both in vitro and in vivo. In keratinocytes, DEK overexpression is sufficient for causing oncogenic phenotypes in the absence of E7. Conversely, DEK loss results in cell death in HPV-positive cervical cancer cells at least in part through p53 activation, and Dek knockout mice are relatively resistant to the development of chemically induced skin papillomas. Despite the established oncogenic role of DEK in HPV-associated cervical cancer cell lines and keratinocytes, a functional role of DEK has not yet been explored in HNSCC. Using an established transgenic mouse model of HPV16 E7-induced HNSCC, we demonstrate that Dek is required for optimal proliferation of E7-transgenic epidermal cells and for the growth of HNSCC tumors. Importantly, these studies also demonstrate that DEK protein is universally upregulated in both HPV-positive and -negative human HNSCC tumors relative to adjacent normal tissue. Furthermore, DEK knockdown inhibited the proliferation of HPV-positive and -negative HNSCC cells, establishing a functional role for DEK in human disease. Mechanistic studies reveal that attenuated HNSCC cell growth in response to DEK loss was associated with reduced expression of the oncogenic p53 family member, ΔNp63. Exogenous ΔNp63 expression rescued the proliferative defect in the absence of DEK, thereby establishing a functional DEK-ΔNp63 oncogenic pathway that promotes HNSCC. Taken together, our data demonstrate that DEK stimulates HNSCC cellular growth and identify ΔNp63 as a novel DEK effector.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Proliferação de Células , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Papillomavirus Humano 16/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecções por Papillomavirus/metabolismo , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Neoplasias de Cabeça e Pescoço , Papillomavirus Humano 16/genética , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Camundongos , Camundongos Knockout , Proteínas Oncogênicas/genética , Proteínas E7 de Papillomavirus/genética , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/patologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
18.
Endocrinology ; 142(8): 3598-606, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11459808

RESUMO

The response of arterial smooth muscle cells to injury is governed by a complex series of events. Significant among them is the paracrine production of peptide growth factors. To determine the impact of local IGF-I gene expression on vascular injury, the left carotid arteries of SMP8-IGF-I mice (in which IGF-I is selectively overexpressed in smooth muscle cells by means of a smooth muscle alpha-actin promoter) and wild-type controls were injured mechanically with an epon resin probe. After 7 and 14 d, a progressive increase in medial area was seen in both SMP8-IGF-I and wild-type mice, but they were not significantly different from each other. However, by 14 d there was a more than 4-fold increase in neointimal area in transgenic vs. wild-type. The intima/media ratios were also strikingly increased at 14 d in the IGF-I-overexpressing animals. The mitotic index, determined in animals injected daily with bromodeoxyuridine for 3 d before death, was markedly elevated in both the media and neointima 7 d after injury in SMP8-IGF-I mice, but the effect had subsided by 14 d. Despite a higher rate of cell division, the relative increase in medial area was less in the SMP8-IGF-I mice than in wild-type mice at both 7 and 14 d, consistent with a stimulation of cell migration to the neointima. The experiments reported here provide compelling evidence that paracrine expression of IGF-I is a powerful stimulus for smooth muscle cell proliferation and migration in vivo.


Assuntos
Lesões das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/fisiopatologia , Fator de Crescimento Insulin-Like I/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Túnica Íntima/crescimento & desenvolvimento , Actinas/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/genética , Camundongos , Camundongos Transgênicos/genética , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , RNA Mensageiro/metabolismo , Túnica Íntima/efeitos dos fármacos
19.
Endocrinology ; 139(5): 2605-14, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9564877

RESUMO

Insulin-like growth factor I (IGF-I) has been postulated to function as a smooth muscle cell (SMC) mitogen and to play a role in the pathogenesis of bladder hypertrophy, estrogen-induced uterine growth, and restenosis after arterial angioplasty. IGF-binding protein-4 (IGFBP-4) inhibits IGF-I action in vitro and is the most abundant IGFBP in the rodent arterial wall. To explore the function of this binding protein in vivo, transgenic mouse lines were developed harboring fusion genes consisting of a rat IGFBP-4 complementary DNA cloned downstream of either a -724 bp fragment of the mouse smooth muscle alpha-actin 5'-flanking region (SMP2-BP-4) or -1074 bp, 63 bp of 5'-untranslated region, and 2.5 kb of intron 1 of smooth muscle alpha-actin (SMP8-BP-4). SMP2-BP-4 mice expressed low levels of the exogenous IGFBP-4 messenger RNA (mRNA), which was not specifically targeted to SMC-rich tissue environments, and were therefore not analyzed further. Six SMP8-BP-4 transgenic lines derived from separate founders were characterized. Mating of hemizygous SMP8-BP-4 mice with controls produced about 50% transgenic offspring, with equal sex distribution. Expression of IGFBP-4 mRNA in nontransgenic littermates was maximal in liver and kidney. By contrast, transgenic IGFBP-4 mRNA expression, distinguished because of a smaller transcript size, was confined to SMC-containing tissues, with the following hierarchy: bladder > aorta > stomach = uterus. There was no transgene expression in skeletal muscle, brain, or cardiac myocytes. The abundance of IGFBP-4 measured by Western ligand blotting or by immunoblotting, was 8- to 10-fold higher in aorta and bladder of SMP8-BP-4 mice than in their nontransgenic littermates, with no change in plasma IGFBP-4 levels. Transgenic mice exhibited a significant reduction in wet weight of SMC-rich tissues, including bladder, intestine, aorta, uterus, and stomach, with no change in total body or carcass weight. In situ hybridization showed that transgene expression was targeted exclusively to the muscular layers of the arteries, veins, bladder, ureter, stomach, intestine, and uterus. Overexpression of IGFBP-4 was associated with SMC hypoplasia, a reciprocal phenotype to that of transgenic mice overexpressing IGF-I under control of the same promoter (SMP8-IGF-I). Double transgenic mice derived from mating SMP8-BP-4 with SMP8-IGF-I animals showed a modest decrease in wet weight at selected SMC tissues. Although we cannot exclude that the effects of IGFBP-4 may be IGF independent, these data suggest that IGFBP-4 is a functional antagonist of IGF-I action on SMC in vivo.


Assuntos
Actinas/genética , Expressão Gênica , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Músculo Liso/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Feminino , Mucosa Gástrica/metabolismo , Hibridização In Situ , Masculino , Camundongos , Camundongos Transgênicos , Músculo Liso/patologia , Tamanho do Órgão , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão , Estômago/patologia , Bexiga Urinária/metabolismo , Bexiga Urinária/patologia , Útero/metabolismo , Útero/patologia
20.
Gene ; 218(1-2): 37-47, 1998 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-9751800

RESUMO

Prosaposin is a multifunctional protein that encodes four glycoproteins, named saposins A, B, C and D. They participate in the catabolism of glycosphingolipids in lysosomes. When secreted, intact prosaposin may function as a neuritogenic factor. Human and mouse prosaposin displayed similar temporal and spatial regulation of expression. To gain insight into the transcriptional regulation of this locus, the 5' region was characterized from the human prosaposin gene. The putative human promoter was shown to be TATA-less, i.e. it belonged to the TATA-less housekeeping gene family. The transcription initiation sites were localized to -23, -27, -31 and -83bp 5' to ATG, compared to -87 and -94bp in the mouse. In SK-N-SH neuroblastoma cells, positive regulatory elements were detected -343 to -813bp upstream of ATG. A negative regulatory region existed between -813 and -2500bp using SK-N-SH, H441 and NS20Y cells. EMSA and DNA-footprint analysis showed that Sp1 and Sp3 are involved in human prosaposin gene regulation. Compared to the mouse promoter, the human promoter is missing a Sp1 cluster within a 310-bp upstream segment, and has AP-1, Oct-1 and two RORalpha sites that are protected from DNaseI by selected nuclear extracts.


Assuntos
Glicoproteínas/genética , Regiões Promotoras Genéticas , Precursores de Proteínas/genética , Animais , Sequência de Bases , Linhagem Celular , DNA , Pegada de DNA , Regulação da Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Mapeamento por Restrição , Saposinas , Fatores de Transcrição/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA