Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
Nat Genet ; 21(3): 326-9, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10080190

RESUMO

c-Jun is a major component of the heterodimeric transcription factor AP-1 and is essential for embryonic development, as fetuses lacking Jun die at mid-gestation with impaired hepatogenesis and primary Jun-/- fibroblasts have a severe proliferation defect and undergo premature senescence in vitro. c-Jun and AP-1 activities are regulated by c-Jun N-terminal phosphorylation (JNP) at serines 63 and 73 through Jun N-terminal kinases(JNKs). JNP is thought to be required for the anti-apoptotic function of c-Jun during hepatogenesis, as mice lacking the JNK kinase SEK1 exhibit liver defects similar to those seen in Jun-/- fetuses. To investigate the physiological relevance of JNP, we replaced endogenous Jun by a mutant Jun allele with serines 63 and 73 mutated to alanines (Jun(tm1wag); hereafter referred to as JunAA). Here we show that primary JunAA fibroblasts have proliferation- and stress-induced apoptotic defects, accompanied by reduced AP-1 activity. JunAA mice are viable and fertile, smaller than controls and resistant to epileptic seizures and neuronal apoptosis induced by the excitatory amino acid kainate. Primary mutant neurons are also protected from apoptosis and exhibit unaltered JNK activity. Our results provide evidence that JNP is dispensable for mouse development, and identify c-Jun as the essential substrate of JNK signalling during kainate-induced neuronal apoptosis.


Assuntos
Apoptose/genética , Proteínas Quinases Ativadas por Mitógeno , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Estresse Fisiológico/genética , Animais , Animais Recém-Nascidos , Sequência de Bases , Divisão Celular/fisiologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Antagonistas GABAérgicos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Hipocampo/efeitos dos fármacos , Homozigoto , Ácido Caínico/farmacologia , Metilnitronitrosoguanidina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Mutantes , Proteína Quinase 12 Ativada por Mitógeno , Dados de Sequência Molecular , Mutagênicos/farmacologia , Mutação , Neurônios/efeitos dos fármacos , Neurônios/patologia , Pentilenotetrazol/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/metabolismo , Convulsões/induzido quimicamente , Raios Ultravioleta
2.
Nat Genet ; 17(4): 479-82, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9398855

RESUMO

Poly(ADP-ribose) polymerase (PARP) and DNA-dependent protein kinase (DNA-PK) are DNA break-activated molecules, Although mice that lack PARP display no gross phenotype and normal DNA excision repair, they exhibit high levels of sister chromatid exchange, indicative of elevated recombination rates. Mutation of the gene for DNA-PK catalytic subunit (Prkdc) cases defective antigen receptor V(D)J recombination and arrests B- and T-lymphocyte development in severe combined immune-deficiency (SCID) mice. SCID V(D)J recombination can be partly rescued in T-lymphocytes by either DNA-damaging agents (gamma-irradiation and bieomycin) or a null mutation of the p53 gene, possibly because of transiently elevated DNA repair activity in response to DNA damage or to delayed apoptosis in the absence of p53. To determine whether the increased chromosomal recombination observed in PARP-deficient cells affects SCID V(D)J recombination, we generated mice lacking both PARP and DNA-PK. Here, we show that thymocytes of SCID mice express both CD4 and CD8 co-receptors, bypassing the SCID block. Double-mutant T-cells in the periphery express TCR beta, which is attributable to productive TCR beta joints. Double-mutant mice develop a high frequency of T-cell lymphoma. These results demonstrate that increased recombination activity after the loss of PARP anti-recombinogenic function can rescue V(D)J recombination in SCID mice and indicate that PARP and DNA-PK cooperate to minimize genomic damage caused by DNA strand breaks.


Assuntos
Proteínas de Ligação a DNA , Rearranjo Gênico , Genes de Imunoglobulinas , Região Variável de Imunoglobulina/genética , Linfoma de Células T/genética , Poli(ADP-Ribose) Polimerases/genética , Proteínas Serina-Treonina Quinases/genética , Recombinação Genética , Animais , Diferenciação Celular/genética , Proteína Quinase Ativada por DNA , Linfoma de Células T/enzimologia , Linfoma de Células T/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Proteínas Nucleares , Baço/metabolismo , Baço/patologia , Timo/metabolismo , Timo/patologia
3.
Nat Genet ; 24(2): 184-7, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10655067

RESUMO

Osteoclasts are bone-resorbing cells derived from haematopoietic precursors of the monocyte-macrophage lineage. Mice lacking Fos (encoding c-Fos) develop osteopetrosis due to an early differentiation block in the osteoclast lineage. c-Fos is a component of the dimeric transcription factor activator protein-1 (Ap-1), which is composed mainly of Fos (c-Fos, FosB, Fra-1 and Fra-2) and Jun proteins (c-Jun, JunB and JunD). Unlike Fra-1 (encoded by Fosl1), c-Fos contains transactivation domains required for oncogenesis and cellular transformation. The mechanism by which c-Fos exerts its specific function in osteoclast differentiation is not understood. Here we show by retroviral-gene transfer that all four Fos proteins, but not the Jun proteins, rescue the differentiation block in vitro. Structure-function analysis demonstrated that the major carboxy-terminal transactivation domains of c-Fos and FosB are dispensable and that Fra-1 (which lacks transactivation domains) has the highest rescue activity. Moreover, a transgene expressing Fra-1 rescues the osteopetrosis of c-Fos-mutant mice in vivo. The osteoclast differentiation factor Rankl (also known as TRANCE, ODF and OPGL; refs 8-11) induces transcription of Fosl1 in a c-Fos-dependent manner, thereby establishing a link between Rank signalling and the expression of Ap-1 proteins in osteoclast differentiation.


Assuntos
Osteoclastos/citologia , Osteoclastos/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Transcrição Gênica , Animais , Proteínas de Transporte/metabolismo , Diferenciação Celular , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Dimerização , Antígeno 2 Relacionado a Fos , Genes fos , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-fos/deficiência , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Fatores de Transcrição/metabolismo
4.
Nat Med ; 3(3): 346-9, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9055866

RESUMO

Apoptotic cell death in the retina was recently demonstrated in animal models of the hereditary human retinal dystrophy known as retinitis pigmentosa. Although recent evidence indicates that the proto-oncogene c-fos is a mediator of apoptosis, its precise role is unclear. In fact, under some conditions, c-fos may even protect against apoptotic cell death. In the retina, c-fos is physiologically expressed in a diurnal manner and is inducible by light. We previously observed a light-elicited, dose-dependent apoptotic response in rat photoreceptors. To determine whether c-fos is involved in the light-induced apoptotic pathway we have used control mice and mice lacking c-fos. We found that following dark adaptation and two hours of light exposure both groups of animals exhibited only a few apoptotic cells. However, at 12 and 24 additional hours after light exposure, apoptosis increased dramatically in controls but was virtually absent in those mice lacking c-fos. Therefore, c-fos is essential for light-induced apoptosis of photoreceptors. Notably, c-fos is continuously upregulated concomitant with apoptotic photoreceptor death in our system and in animal models of retinitis pigmentosa (Agarwal, N. et al., Invest. Ophthalmol. Vis.Sci. Suppl. 36, S638 and Rich, K.A. et al., Invest. Ophthalmol. Vis. Sci. Suppl. 35, 1833). Inhibition of c-fos expression might therefore represent a novel therapeutic strategy to retard the time course of retinal dystrophies and light-induced retinal degeneration.


Assuntos
Apoptose/genética , Regulação da Expressão Gênica , Genes fos , Degeneração Retiniana/patologia , Animais , Humanos , Luz , Camundongos , Camundongos Knockout , Proto-Oncogene Mas , Ratos , Degeneração Retiniana/genética
5.
Nat Med ; 6(9): 980-4, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10973316

RESUMO

Bone formation by osteoblasts is essential for skeletal growth and remodeling. Fra-1 is a c-Fos-related protein belonging to the AP-1 family of transcription factors. Here we show that transgenic mice overexpressing Fra-1 in various organs develop a progressive increase in bone mass leading to osteosclerosis of the entire skeleton, which is due to a cell-autonomous increase in the number of mature osteoblasts. Moreover, osteoblast differentiation, but not proliferation, was enhanced and osteoclastogenesis was also elevated in vitro. These data indicate that, unlike c-Fos, which causes osteosarcomas, Fra-1 specifically enhances bone formation, which may be exploited to stimulate bone formation in pathological conditions.


Assuntos
Calcinose/genética , Osteoblastos/citologia , Osteosclerose/genética , Proteínas Proto-Oncogênicas c-fos/genética , Animais , Diferenciação Celular , Camundongos , Camundongos Transgênicos , Osteoblastos/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-fos/biossíntese
6.
Nat Med ; 5(3): 314-9, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10086388

RESUMO

Human type 1 diabetes results from the selective destruction of insulin-producing pancreatic beta cells during islet inflammation. Cytokines and reactive radicals released during this process contribute to beta-cell death. Here we show that mice with a disrupted gene coding for poly (ADP-ribose) polymerase (PARP-/- mice) are completely resistant to the development of diabetes induced by the beta-cell toxin streptozocin. The mice remained normoglycemic and maintained normal levels of total pancreatic insulin content and normal islet ultrastructure. Cultivated PARP-/- islet cells resisted streptozocin-induced lysis and maintained intracellular NAD+ levels. Our results identify NAD+ depletion caused by PARP activation as the dominant metabolic event in islet-cell destruction, and provide information for the development of strategies to prevent the progression or manifestation of the disease in individuals at risk of developing type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/enzimologia , Poli(ADP-Ribose) Polimerases/fisiologia , Animais , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Modelos Animais de Doenças , Resistência a Medicamentos , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , NAD/metabolismo , Poli(ADP-Ribose) Polimerases/genética , Estreptozocina
7.
J Exp Med ; 193(3): 317-28, 2001 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-11157052

RESUMO

Apoptotic and mitogenic stimuli activate c-Jun NH2-terminal kinases (JNKs) in T cells. Although T cells express both JNK1 and JNK2 isozymes, the absence of JNK2 alone can result in resistance to anti-CD3-induced thymocyte apoptosis and defective mature T cell proliferation. Similar defects in thymocyte apoptosis and mature T cell proliferation, the latter due to reduced interleukin 2 production, are also caused by JNK1 deficiency. Importantly, T cell function was compromised in Jnk1(+/-)Jnk2(+/-) double heterozygous mice, indicating that JNK1 and JNK2 play similar roles in regulating T cell function. The reduced JNK dose results in defective c-Jun NH2-terminal phosphorylation in thymocytes but not in peripheral T cells, in which nuclear factors of activated T cells (NK-ATs)-DNA binding activity is affected. Thus, JNK1 and JNK2 control similar functions during T cell maturation through differential targeting of distinct substrates.


Assuntos
Apoptose , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteínas Nucleares , Linfócitos T/fisiologia , Animais , Linfócitos B/imunologia , Complexo CD3/imunologia , Diferenciação Celular , Divisão Celular , Sobrevivência Celular , Proteínas de Ligação a DNA/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 8 Ativada por Mitógeno , Proteína Quinase 9 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/genética , Fatores de Transcrição NFATC , Fosforilação , Linfócitos T/citologia , Fatores de Transcrição/metabolismo
8.
J Exp Med ; 193(3): 353-64, 2001 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-11157055

RESUMO

Invasive Salmonella induces macrophage apoptosis via the activation of caspase-1 by the bacterial protein SipB. Here we show that infection of macrophages with Salmonella causes the activation and degradation of Raf-1, an important intermediate in macrophage proliferation and activation. Raf-1 degradation is SipB- and caspase-1-dependent, and is prevented by proteasome inhibitors. To study the functional significance of Raf-1 in this process, the c-raf-1 gene was inactivated by Cre-loxP-mediated recombination in vivo. Macrophages lacking c-raf-1 are hypersensitive towards pathogen-induced apoptosis. Surprisingly, activation of the antiapoptotic mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) and nuclear factor kappaB pathways is normal in Raf-1-deficient macrophages, and mitochondrial fragility is not increased. Instead, pathogen-mediated activation of caspase-1 is enhanced selectively, implying that Raf-1 antagonizes stimulus-induced caspase-1 activation and apoptosis.


Assuntos
Apoptose , Macrófagos/citologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Salmonella typhimurium/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/enzimologia , Caspase 1/metabolismo , Células Cultivadas , Ativação Enzimática , Leupeptinas/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Inibidores de Proteases/farmacologia , Proteínas Proto-Oncogênicas c-raf/metabolismo
9.
Ann Rheum Dis ; 69 Suppl 1: i86-88, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19995753

RESUMO

The Fos and Jun proteins are members of the AP-1 transcription factor complex, which is a central regulator for many cellular functions. This paper summarises the important functions of Fos proteins in bone development, with special emphasis on the Fos-related proteins Fra-1 and Fra-2. These factors determine the functions of osteoblasts and osteoclasts and regulate cytokine signalling during bone development. Likewise, the Jun proteins control the expression of cytokines and chemokines and are probably causally involved in inflammatory skin diseases such as psoriasis. Investigations into the molecular mechanisms responsible for skin inflammation have revealed that Jun proteins control cytokine expression, such as granulocyte colony-stimulating factor, IL-6 and tumour necrosis factor alpha by transcriptional and posttranscriptional pathways. Finally, the paper discusses the relevance of the Jun-dependent mouse model for psoriasis for preclinical studies in the field of anti-angiogenic therapies.


Assuntos
Desenvolvimento Ósseo/fisiologia , Inflamação/fisiopatologia , Fator de Transcrição AP-1/fisiologia , Inibidores da Angiogênese/uso terapêutico , Animais , Dermatite/fisiopatologia , Modelos Animais de Doenças , Humanos , Inflamação/tratamento farmacológico , Camundongos , Proteínas Proto-Oncogênicas c-fos/fisiologia , Proteínas Proto-Oncogênicas c-jun/fisiologia , Psoríase/metabolismo
10.
Cell Death Differ ; 15(1): 89-93, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17762881

RESUMO

During the development and organogenesis of all multicellular organisms, cell fate decisions determine whether cells undergo proliferation, differentiation, or aging. Two independent stress kinase signaling pathways, p38-MAPK, and JNKs, have evolved that relay developmental and environmental cues to determine cell responses. Although multiple stimuli can activate these two stress kinase pathways, the functional interactions and molecular cross-talks between these common second signaling cascades are poorly elucidated. Here we report that JNK and p38-MAPK pathways antagonistically control cellular senescence, oncogenic transformation, and proliferation in primary mouse embryonic fibroblasts (MEFs). Similarly, genetic inactivation of the JNK pathway results in impaired proliferation of fetal hepatoblasts in vitro and defective adult liver regeneration in vivo, which is rescued by inhibition of the p38-MAPK pathway. Thus, the balance between the two stress-signaling pathways, MKK7-JNK and MKK3/6-p38-MAPK, determines cell fate and links environmental and developmental stress to cell cycle arrest, senescence, oncogenic transformation, and adult tissue regeneration.


Assuntos
Proliferação de Células , Transformação Celular Neoplásica , Senescência Celular , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Regeneração Hepática , Sistema de Sinalização das MAP Quinases , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Proteína Quinase CDC2/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Hepatócitos/metabolismo , Camundongos , Camundongos Mutantes
11.
J Cell Biol ; 122(3): 685-701, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8335693

RESUMO

We have generated transgenic mice expressing the proto-oncogene c-fos from an H-2Kb class I MHC promoter as a tool to identify and isolate cell populations which are sensitive to altered levels of Fos protein. All homozygous H2-c-fosLTR mice develop osteosarcomas with a short latency period. This phenotype is specific for c-fos as transgenic mice expressing the fos- and jun-related genes, fosB and c-jun, from the same regulatory elements do not develop any pathology despite high expression in bone tissues. The c-fos transgene is not expressed during embryogenesis but is expressed after birth in bone tissues before the onset of tumor formation, specifically in putative preosteoblasts, bone-forming osteoblasts, osteocytes, as well as in osteoblastic cells present within the tumors. Primary and clonal cell lines established from c-fos-induced tumors expressed high levels of exogenous c-fos as well as the bone cell marker genes, type I collagen, alkaline phosphatase, and osteopontin/2ar. In contrast, osteocalcin/BGP expression was either low or absent. All cell lines were tumorigenic in vivo, some of which gave rise to osteosarcomas, expressing exogenous c-fos mRNA, and Fos protein in osteoblastic cells. Detailed analysis of one osteogenic cell line, P1, and several P1-derived clonal cell lines indicated that bone-forming osteoblastic cells were transformed by Fos. The regulation of osteocalcin/BGP and alkaline phosphatase gene expression by 1,25-dihydroxyvitamin D3 was abrogated in P1-derived clonal cells, whereas glucocorticoid responsiveness was unaltered. These results suggest that high levels of Fos perturb the normal growth control of osteoblastic cells and exert specific effects on the expression of the osteoblast phenotype.


Assuntos
Neoplasias Ósseas/genética , Transformação Celular Neoplásica , Genes fos , Osteoblastos/metabolismo , Osteossarcoma/genética , Animais , Sequência de Bases , Calcitriol/farmacologia , Dexametasona/farmacologia , Expressão Gênica , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Osteoblastos/citologia , Osteogênese , Fenótipo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Células Tumorais Cultivadas
12.
J Cell Biol ; 145(5): 1049-61, 1999 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-10352021

RESUMO

Mice lacking the AP-1 transcription factor c-Jun die around embryonic day E13.0 but little is known about the cell types affected as well as the cause of embryonic lethality. Here we show that a fraction of mutant E13.0 fetal livers exhibits extensive apoptosis of both hematopoietic cells and hepatoblasts, whereas the expression of 15 mRNAs, including those of albumin, keratin 18, hepatocyte nuclear factor 1, beta-globin, and erythropoietin, some of which are putative AP-1 target genes, is not affected. Apoptosis of hematopoietic cells in mutant livers is most likely not due to a cell-autonomous defect, since c-jun-/- fetal liver cells are able to reconstitute all hematopoietic compartments of lethally irradiated recipient mice. A developmental analysis of chimeras showed contribution of c-jun-/- ES cell derivatives to fetal, but not to adult livers, suggesting a role of c-Jun in hepatocyte turnover. This is in agreement with the reduced mitotic and increased apoptotic rates found in primary liver cell cultures derived from c-jun-/- fetuses. Furthermore, a novel function for c-Jun was found in heart development. The heart outflow tract of c-jun-/- fetuses show malformations that resemble the human disease of a truncus arteriosus persistens. Therefore, the lethality of c-jun mutant fetuses is most likely due to pleiotropic defects reflecting the diversity of functions of c-Jun in development, such as a role in neural crest cell function, in the maintenance of hepatic hematopoiesis and in the regulation of apoptosis.


Assuntos
Coração/embriologia , Coração/fisiologia , Fígado/embriologia , Fígado/fisiologia , Proteínas Proto-Oncogênicas c-jun/fisiologia , Fator de Transcrição AP-1/fisiologia , Animais , Apoptose , Desenvolvimento Embrionário e Fetal , Deleção de Genes , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/fisiologia , Fígado/patologia , Camundongos , Camundongos Knockout
13.
J Cell Biol ; 127(3): 835-45, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7525597

RESUMO

We generated mice, null mutant in the adhesion molecule on glia (AMOG), the beta 2 subunit of the murine Na,K-ATPase gene. These mice exhibit motor incoordination at 15 d of age, subsequently tremor and paralysis of extremities, and die at 17-18 d after birth. At these ages, the mutants have enlarged ventricles, degenerating photoreceptor cells, and swelling and degeneration of astrocytic endfeet, leading to vacuoles adjoining capillaries of brain stem, thalamus, striatum, and spinal cord. In tissue homogenates from entire brains of 16-17-d-old mutants, Na,K-ATPase activity and expression of the beta 1 subunit of the Na,K-ATPase and of the neural adhesion molecules L1, N-CAM, and MAG appear normal. We suggest that the mutant phenotype can be related primarily to reduced pump activity, with neural degeneration as a possible consequence of osmotic imbalance.


Assuntos
Encéfalo/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Proteínas da Matriz Extracelular/genética , Degeneração Neural , Neuroglia/metabolismo , Neurônios/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , Medula Espinal/metabolismo , Adenosina Trifosfatases , Sequência de Aminoácidos , Animais , Sequência de Bases , Southern Blotting , Encéfalo/citologia , Capilares/patologia , Capilares/ultraestrutura , Proteínas de Transporte de Cátions , Córtex Cerebelar/citologia , Córtex Cerebelar/fisiologia , Córtex Cerebelar/ultraestrutura , Circulação Cerebrovascular , Clonagem Molecular , Embrião de Mamíferos , Hibridização In Situ , Substâncias Macromoleculares , Camundongos , Camundongos Mutantes Neurológicos , Microscopia Eletrônica , Dados de Sequência Molecular , Atividade Motora , Neuroglia/citologia , Neuroglia/ultraestrutura , Neurônios/citologia , Neurônios/ultraestrutura , Oligodesoxirribonucleotídeos , Reação em Cadeia da Polimerase , Mapeamento por Restrição , Medula Espinal/citologia , Células-Tronco/metabolismo , Vacúolos/ultraestrutura
14.
J Cell Biol ; 137(4): 953-63, 1997 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-9151696

RESUMO

The middle T antigen of murine Polyomavirus (PymT) rapidly transforms endothelial cells, leading to the formation of vascular tumors in newborn mice. Transformed endothelial (End.) cell lines established from such tumors exhibit altered proteolytic activity as a result of increased expression of urokinase-type plasminogen activator (uPA) and are capable of inducing vascular tumors efficiently when injected into adult mice. In this study we have used mice lacking components of the PA/plasmin system to analyze the role of this system in the transformation process and in tumor growth. We found that the proteolytic status of the host is not a critical determinant for PymT-induced vascular tumor formation. In addition, the lack of either uPA or tissue-type PA (tPA) activity is not limiting for the establishment and proliferation of End. cells in vitro, although the combined loss of both PA activities leads to a marked reduction in proliferation rates. Furthermore, the in vitro morphogenetic properties of mutant End. cells in fibrin gels could only be correlated with an altered proteolytic status in cells lacking both uPA and tPA. However, in contrast with tumors induced by PymT itself, the tumorigenic potential of mutant and wild-type End. cell lines was found to be highly dependent on the proteolytic status of both the tumor cells and the host. Thus, genetic alterations in the PA/plasmin system affect vascular tumor development, indicating that this system is a causal component in PymTmediated oncogenesis.


Assuntos
Antígenos Transformantes de Poliomavirus , Transformação Celular Viral , Endotélio Vascular/citologia , Fibrinolisina/fisiologia , Ativadores de Plasminogênio/fisiologia , Neoplasias Vasculares/etiologia , Animais , Animais Recém-Nascidos , Linhagem Celular , Endotélio Vascular/enzimologia , Fibrina , Géis , Expressão Gênica , Camundongos , Camundongos Knockout , Morfogênese , Inibidor 1 de Ativador de Plasminogênio/deficiência , RNA Mensageiro/genética , Ativador de Plasminogênio Tecidual/deficiência , Ativador de Plasminogênio Tipo Uroquinase/deficiência
15.
J Cell Biol ; 151(7): 1537-48, 2000 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-11134081

RESUMO

Ras is a universal eukaryotic intracellular protein integrating extracellular signals from multiple receptor types. To investigate its role in the adult central nervous system, constitutively activated V12-Ha-Ras was expressed selectively in neurons of transgenic mice via a synapsin promoter. Ras-transgene protein expression increased postnatally, reaching a four- to fivefold elevation at day 40 and persisting at this level, thereafter. Neuronal Ras was constitutively active and a corresponding activating phosphorylation of mitogen-activated kinase was observed, but there were no changes in the activity of phosphoinositide 3-kinase, the phosphorylation of its target kinase Akt/PKB, or expression of the anti-apoptotic proteins Bcl-2 or Bcl-X(L). Neuronal Ras activation did not alter the total number of neurons, but induced cell soma hypertrophy, which resulted in a 14.5% increase of total brain volume. Choline acetyltransferase and tyrosine hydroxylase activities were increased, as well as neuropeptide Y expression. Degeneration of motorneurons was completely prevented after facial nerve lesion in Ras-transgenic mice. Furthermore, neurotoxin-induced degeneration of dopaminergic substantia nigra neurons and their striatal projections was greatly attenuated. Thus, the Ras signaling pathway mimics neurotrophic effects and triggers neuroprotective mechanisms in adult mice. Neuronal Ras activation might become a tool to stabilize donor neurons for neural transplantation and to protect neuronal populations in neurodegenerative diseases.


Assuntos
Hipertrofia/patologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Doenças Neurodegenerativas/patologia , Fármacos Neuroprotetores/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Axotomia , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Contagem de Células , Tamanho Celular , Células Cultivadas , Colina O-Acetiltransferase/metabolismo , Ativação Enzimática , Hipertrofia/enzimologia , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/enzimologia , Mutação/genética , Doenças Neurodegenerativas/enzimologia , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Oxidopamina/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Transdução de Sinais/efeitos dos fármacos , Substância Negra/efeitos dos fármacos , Substância Negra/patologia , Tirosina 3-Mono-Oxigenase/metabolismo
16.
Science ; 269(5221): 234-8, 1995 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-7618085

RESUMO

Mice and cells lacking the epidermal growth factor receptor (EGFR) were generated to examine its physiological role in vivo. Mutant fetuses are retarded in growth and die at mid-gestation in a 129/Sv genetic background, whereas in a 129/Sv x C57BL/6 cross some survive until birth and even to postnatal day 20 in a 129/Sv x C57BL/6 x MF1 background. Death in utero probably results from a defect in the spongiotrophoblast layer of the placenta. Newborn mutant mice have open eyes, rudimentary whiskers, immature lungs, and defects in the epidermis, correlating with the expression pattern of the EGFR as monitored by beta-galactosidase activity. These defects are probably cell-autonomous because chimeric mice generated with EGFR-/- embryonic stem cells contribute small amounts of mutant cells to some organs. These results indicate that the EGFR regulates epithelial proliferation and differentiation and that the genetic background influences the resulting phenotype.


Assuntos
Desenvolvimento Embrionário e Fetal , Células Epiteliais , Receptores ErbB/fisiologia , Animais , Diferenciação Celular , Divisão Celular , Receptores ErbB/genética , Feminino , Marcação de Genes , Hematopoese , Pulmão/citologia , Pulmão/embriologia , Masculino , Camundongos , Camundongos Endogâmicos , Mutação , Fenótipo , Placenta/fisiologia , Pele/citologia , Pele/embriologia , Especificidade da Espécie , Células-Tronco/citologia , Trofoblastos/citologia
17.
Science ; 217(4564): 1046-8, 1982 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-6287575

RESUMO

Foreign gene sequences were retained in two adult mice (out of 62 analyzed) from fertilized eggs injected with a recombinant plasmid containing the human beta-globin genomic region and the herpes simplex viral thymidine kinase gene. The intact human and viral genes were found in DNA of one of the animals and, in the other, at least part of the human globin gene was present. The latter individual transmitted these sequences to its progeny in a Mendelian ration. Thus, human DNA may be incorporated into the germ line of mice for in vivo studies of regulation of gene expression in development, genetic diseases, and malignancy.


Assuntos
Globinas/genética , Recombinação Genética , Animais , Sequência de Bases , DNA/genética , Enzimas de Restrição do DNA , DNA Recombinante , Feminino , Genes , Genes Virais , Células Germinativas , Humanos , Camundongos , Microinjeções , Hibridização de Ácido Nucleico , Simplexvirus/enzimologia , Timidina Quinase/genética , Zigoto
18.
Science ; 266(5184): 443-8, 1994 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-7939685

RESUMO

Mice lacking the proto-oncogene c-fos develop the bone disease osteopetrosis. Fos mutant mice were found to have a block in the differentiation of bone-resorbing osteoclasts that was intrinsic to hematopoietic cells. Bone marrow transplantation rescued the osteopetrosis, and ectopic c-fos expression overcame this differentiation block. The lack of Fos also caused a lineage shift between osteoclasts and macrophages that resulted in increased numbers of bone marrow macrophages. These results identify Fos as a key regulator of osteoclast-macrophage lineage determination in vivo and provide insights into the molecular mechanisms underlying metabolic bone diseases.


Assuntos
Remodelação Óssea/fisiologia , Células-Tronco Hematopoéticas/citologia , Macrófagos/citologia , Osteoclastos/citologia , Proteínas Proto-Oncogênicas c-fos/fisiologia , Animais , Transplante de Medula Óssea , Diferenciação Celular , Células Cultivadas , Genes fos , Transplante de Células-Tronco Hematopoéticas , Camundongos , Camundongos Mutantes , Osteogênese , Osteopetrose/metabolismo , Osteopetrose/patologia , Proteínas Proto-Oncogênicas c-fos/genética
19.
Oncogene ; 26(33): 4863-71, 2007 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-17297445

RESUMO

Loss of JunB has been observed in human leukemia and lymphoma, but it remains unknown, whether this loss is relevant to disease progression. Here, we investigated the consequences of JunB deficiency using Abelson-induced B-lymphoid leukemia as a model system. Mice deficient in JunB expression succumbed to Abelson-induced leukemia with increased incidence and significantly reduced latency. Similarly, bcr/abl p185-transformed JunB-deficient (junB(Delta/Delta)) cells induced leukemia in RAG2(-/-) mice displaying a more malignant phenotype. These observations indicated that cell intrinsic effects within the junB(Delta/Delta) tumor cells accounted for the accelerated leukemia development. Indeed, explantated bcr/abl p185 transformed junB(Delta/Delta) cells proliferated faster than the control cells. The proliferative advantage emerged slowly after the initial transformation process and was associated with increased expression levels of the cell cycle kinase cdk6 and with decreased levels of the cell cycle inhibitor p16(INK4a). These alterations were due to irreversible reprogramming of the cell, because - once established - accelerated disease induced by junB(Delta/Delta) cells was not reverted by re-introducing JunB. Consistent with this observation, we found that the p16 promoter was methylated. Thus, JunB functions as a gatekeeper during tumor evolution. In its absence, transformed leukemic cells acquire an enhanced proliferative capacity, which presages a more malignant disease.


Assuntos
Leucemia Linfoide/patologia , Proteínas Proto-Oncogênicas c-jun/fisiologia , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Citometria de Fluxo , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Proteínas de Fusão bcr-abl/fisiologia , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Leucemia Experimental/patologia , Leucemia Linfoide/genética , Leucemia Linfoide/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção
20.
Curr Opin Genet Dev ; 11(5): 527-32, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11532394

RESUMO

The skeleton is a single organ composed of >200 different elements spread throughout the body. These skeletal elements comprise two tissues: cartilage and bone. Both tissues contain specific cell type(s): chondrocytes in cartilage and osteoblasts and osteoclasts in bone. We are beginning to understand the genetic control of the differentiation and function of these cells through recent developments in mouse and human genetics, and also through the use of molecular biological and biochemical techniques. The most recent advances in terms of cell differentiation in the skeleton are presented in this review.


Assuntos
Desenvolvimento Ósseo/genética , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Cartilagem/crescimento & desenvolvimento , Cartilagem/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Animais , Cartilagem/citologia , Diferenciação Celular , Condrócitos/citologia , Condrócitos/metabolismo , Humanos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA