Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
3.
PLoS Genet ; 5(6): e1000527, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19543366

RESUMO

Damage initiates a pleiotropic cellular response aimed at cellular survival when appropriate. To identify genes required for damage survival, we used a cell-based RNAi screen against the Drosophila genome and the alkylating agent methyl methanesulphonate (MMS). Similar studies performed in other model organisms report that damage response may involve pleiotropic cellular processes other than the central DNA repair components, yet an intuitive systems level view of the cellular components required for damage survival, their interrelationship, and contextual importance has been lacking. Further, by comparing data from different model organisms, identification of conserved and presumably core survival components should be forthcoming. We identified 307 genes, representing 13 signaling, metabolic, or enzymatic pathways, affecting cellular survival of MMS-induced damage. As expected, the majority of these pathways are involved in DNA repair; however, several pathways with more diverse biological functions were also identified, including the TOR pathway, transcription, translation, proteasome, glutathione synthesis, ATP synthesis, and Notch signaling, and these were equally important in damage survival. Comparison with genomic screen data from Saccharomyces cerevisiae revealed no overlap enrichment of individual genes between the species, but a conservation of the pathways. To demonstrate the functional conservation of pathways, five were tested in Drosophila and mouse cells, with each pathway responding to alkylation damage in both species. Using the protein interactome, a significant level of connectivity was observed between Drosophila MMS survival proteins, suggesting a higher order relationship. This connectivity was dramatically improved by incorporating the components of the 13 identified pathways within the network. Grouping proteins into "pathway nodes" qualitatively improved the interactome organization, revealing a highly organized "MMS survival network." We conclude that identification of pathways can facilitate comparative biology analysis when direct gene/orthologue comparisons fail. A biologically intuitive, highly interconnected MMS survival network was revealed after we incorporated pathway data in our interactome analysis.


Assuntos
Drosophila/fisiologia , Redes Reguladoras de Genes , Interferência de RNA , Transdução de Sinais , Animais , Linhagem Celular , Células Cultivadas , Dano ao DNA/efeitos dos fármacos , Drosophila/efeitos dos fármacos , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Genoma de Inseto/efeitos dos fármacos , Metanossulfonato de Metila/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Mutagênicos/farmacologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Hum Mol Genet ; 18(13): 2472-82, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19383632

RESUMO

Proper limb development requires concerted communication between cells within the developing limb bud. Several molecules have been identified which contribute to the formation of a circuitry loop consisting in large part of secreted proteins. The intracellular actin nucleator, Formin 1 (Fmn1), has previously been implicated in limb development, but questions remain after the identification of a Gremlin transcriptional enhancer within the 3' end of the Fmn 1 locus. To resolve this issue, a knockout mouse devoid of Fmn1 protein was created and characterized. The mice exhibit a reduction of digit number to four, a deformed posterior metatarsal, phalangeal soft tissue fusion as well as the absence of a fibula to 100% penetrance in the FVB genetic background. Importantly, this mutant allele does not genetically disrupt the characterized Gremlin enhancer, and indeed Gremlin RNA expression is upregulated at the 35 somite stage of development. Our data reveal increased Bone Morphogenetic Protein (Bmp) activity in mice which carry a disruption in Fmn1, as evidenced by upregulation of Msx1 and a decrease in Fgf4 within the apical ectodermal ridge. Additionally, these studies show enhanced activity downstream of the Bmp receptor in cells where Fmn1 is perturbed, suggesting a role for Fmn1 in repression of Bmp signaling.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Fetais/genética , Inativação Gênica , Deformidades Congênitas dos Membros/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas Nucleares/genética , Transdução de Sinais , Regulação para Cima , Animais , Receptores de Proteínas Morfogenéticas Ósseas/genética , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Modelos Animais de Doenças , Extremidades/embriologia , Proteínas Fetais/metabolismo , Forminas , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Deformidades Congênitas dos Membros/embriologia , Deformidades Congênitas dos Membros/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo
5.
J Immunol ; 182(1): 347-60, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19109166

RESUMO

The RecQ family helicase BLM is critically involved in the maintenance of genomic stability, and BLM mutation causes the heritable disorder Bloom's syndrome. Affected individuals suffer from a predisposition to a multitude of cancer types and an ill-defined immunodeficiency involving low serum Ab titers. To investigate its role in B cell biology, we inactivated murine Blm specifically in B lymphocytes in vivo. Numbers of developing B lymphoid cells in the bone marrow and mature B cells in the periphery were drastically reduced upon Blm inactivation. Of the major peripheral B cell subsets, B1a cells were most prominently affected. In the sera of Blm-deficient naive mice, concentrations of all Ig isotypes were low, particularly IgG3. Specific IgG Ab responses upon immunization were poor and mutant B cells exhibited a generally reduced Ab class switch capacity in vitro. We did not find evidence for a crucial role of Blm in the mechanism of class switch recombination. However, a modest shift toward microhomology-mediated switch junction formation was observed in Blm-deficient B cells. Finally, a cohort of p53-deficient, conditional Blm knockout mice revealed an increased propensity for B cell lymphoma development. Impaired cell cycle progression and survival as well as high rates of chromosomal structural abnormalities in mutant B cell blasts were identified as the basis for the observed effects. Collectively, our data highlight the importance of BLM-dependent genome surveillance for B cell immunity by ensuring proper development and function of the various B cell subsets while counteracting lymphomagenesis.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/patologia , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Instabilidade Genômica/imunologia , RecQ Helicases/deficiência , RecQ Helicases/genética , Animais , Apoptose/genética , Apoptose/imunologia , Subpopulações de Linfócitos B/enzimologia , Síndrome de Bloom/enzimologia , Síndrome de Bloom/imunologia , Síndrome de Bloom/patologia , Ciclo Celular/genética , Ciclo Celular/imunologia , Diferenciação Celular/genética , Linhagem da Célula/genética , Replicação do DNA/genética , Replicação do DNA/imunologia , Isotipos de Imunoglobulinas/biossíntese , Isotipos de Imunoglobulinas/genética , Isotipos de Imunoglobulinas/metabolismo , Região de Troca de Imunoglobulinas/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neoplasias/enzimologia , Neoplasias/imunologia , Neoplasias/patologia , RecQ Helicases/fisiologia , Recombinação Genética/imunologia
6.
Dev Cell ; 15(4): 547-57, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18854139

RESUMO

The mechanisms responsible for maintaining genomic methylation imprints in mouse embryos are not understood. We generated a knockout mouse in the Zfp57 locus encoding a KRAB zinc finger protein. Loss of just the zygotic function of Zfp57 causes partial neonatal lethality, whereas eliminating both the maternal and zygotic functions of Zfp57 results in a highly penetrant embryonic lethality. In oocytes, absence of Zfp57 results in failure to establish maternal methylation imprints at the Snrpn imprinted region. Intriguingly, methylation imprints are reacquired specifically at the maternally derived Snrpn imprinted region when the zygotic Zfp57 is present in embryos. This suggests that there may be DNA methylation-independent memory for genomic imprints. Zfp57 is also required for the postfertilization maintenance of maternal and paternal methylation imprints at multiple imprinted domains. The effects on genomic imprinting are consistent with the maternal-zygotic lethality of Zfp57 mutants.


Assuntos
Impressão Genômica , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Dedos de Zinco/genética , Zigoto/metabolismo , Sequência de Aminoácidos , Animais , Blastocisto/metabolismo , Metilação de DNA , Embrião de Mamíferos , Feminino , Heterozigoto , Homozigoto , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/metabolismo , Oócitos/metabolismo , Gravidez , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Homologia de Sequência de Aminoácidos
8.
PLoS One ; 3(6): e2497, 2008 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-18560567

RESUMO

BACKGROUND: Regulation of the cytoskeleton is a central feature of cell migration. The formin family of proteins controls the rate of actin nucleation at its barbed end. Thus, formins are predicted to contribute to several important cell processes such as locomotion, membrane ruffling, vesicle endocytosis, and stress fiber formation and disassociation. METHODOLOGY/PRINCIPAL FINDINGS: In this study we investigated the functional role of Formin1-isoform4 (Fmn1-IV) by using genetically null primary cells that displayed augmented protrusive behaviour during wound healing and delayed cell spreading. Cells deficient of Fmn1-IV also showed reduced efficiency of focal adhesion formation. Additionally, we generated an enhanced green fluorescence protein (EGFP)-fused Fmn1-IV knock-in mouse to monitor the endogenous subcellular localization of Fmn1-IV. Its localization was found within the cytoplasm and along microtubules, yet it was largely excluded from adherens junctions. CONCLUSIONS/SIGNIFICANCE: It was determined that Fmn1-IV, as an actin nucleator, contributes to protrusion of the cell's leading edge and focal adhesion formation, thus contributing to cell motility.


Assuntos
Movimento Celular/fisiologia , Proteínas Fetais/fisiologia , Proteínas dos Microfilamentos/fisiologia , Proteínas Nucleares/fisiologia , Isoformas de Proteínas/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Proteínas Fetais/genética , Imunofluorescência , Forminas , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Microscopia Eletrônica , Proteínas Nucleares/genética , Isoformas de Proteínas/genética
9.
BMC Cell Biol ; 8: 37, 2007 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-17725840

RESUMO

BACKGROUND: The mechanism involved in the maintenance and differentiation of embryonic stem (ES) cells is incompletely understood. RESULTS: To address this issue, we have developed a retroviral gene trap vector that can target genes expressed in undifferentiated ES cells. This gene trap vector harbors both GFP and Neo reporter genes. G-418 drug resistance was used to select ES clones in which the vector was integrated into transcriptionally active loci. This was then followed by GFP FACS profiling to identify ES clones with reduced GFP fluorescence and, hence, reduced transcriptional activity when ES cells differentiate. Reduced expression of the GFP reporter in six of three hundred ES clones in our pilot screening was confirmed to be down-regulated by Northern blot analysis during ES cell differentiation. These six ES clones represent four different genes. Among the six integration sites, one was at Zfp-57 whose gene product is known to be enriched in undifferentiated ES cells. Three were located in an intron of a novel isoform of CSL/RBP-J kappa which encodes the key transcription factor of the LIN-12/Notch pathway. Another was inside a gene that may encode noncoding RNA transcripts. The last integration event occurred at a locus that may harbor a novel gene. CONCLUSION: Taken together, we demonstrate the use of a novel retroviral gene trap vector in identifying genes preferentially expressed in undifferentiated ES cells.


Assuntos
Células-Tronco Embrionárias/metabolismo , Expressão Gênica , Vetores Genéticos , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos
10.
Proc Natl Acad Sci U S A ; 104(22): 9381-6, 2007 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-17517653

RESUMO

Regulation of chromatin structure is critical in many fundamental cellular processes. Previous studies have suggested that the Rb tumor suppressor may recruit multiple chromatin regulatory proteins to repress E2F, a key regulator of cell proliferation and differentiation. Taking advantage of the evolutionary conservation of the E2F pathway, we have conducted a genome-wide RNAi screen in cultured Drosophila cells for genes required for repression of E2F activity. Among the genes identified are components of the putative Domino chromatin remodeling complex, as well as the Polycomb Group (PcG) protein-like fly tumor suppressor, L3mbt, and the related CG16975/dSfmbt. These factors are recruited to E2F-responsive promoters through physical association with E2F and are required for repression of endogenous E2F target genes. Surprisingly, their inhibitory activities on E2F appear to be independent of Rb. In Drosophila, domino mutation enhances cell proliferation induced by E2F overexpression and suppresses a loss-of-function cyclin E mutation. These findings suggest that potential chromatin regulation mediated by Domino and PcG-like factors plays an important role in controlling E2F activity and cell growth.


Assuntos
Cromatina/genética , Regulação para Baixo , Drosophila melanogaster/genética , Fatores de Transcrição E2F/antagonistas & inibidores , Genoma de Inseto/genética , Interferência de RNA , Animais , Células Cultivadas , Ciclina H , Ciclinas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Testes Genéticos , Mutação/genética , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Mol Cell Biol ; 27(5): 1947-59, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17210642

RESUMO

Bloom's syndrome is a genetic disorder characterized by increased incidence of cancer and an immunodeficiency of unknown origin. The BLM gene mutated in Bloom's syndrome encodes a DNA helicase involved in the maintenance of genomic integrity. To explore the role of BLM in the immune system, we ablated murine Blm in the T-cell lineage. In the absence of Blm, thymocytes were severely reduced in numbers and displayed a developmental block at the beta-selection checkpoint that was partially p53 dependent. Blm-deficient thymocytes rearranged their T-cell receptor (TCR) beta genes normally yet failed to survive and proliferate in response to pre-TCR signaling. Furthermore, peripheral T cells were reduced in numbers, manifested defective homeostatic and TCR-induced proliferation, and produced extensive chromosomal damage. Finally, CD4(+) and CD8(+) T-cell responses were impaired upon antigen challenge. Thus, by ensuring genomic stability, Blm serves a vital role for development, maintenance, and function of T lymphocytes, suggesting a basis for the immune deficiency in Bloom's syndrome.


Assuntos
Adenosina Trifosfatases/metabolismo , Linhagem da Célula , DNA Helicases/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Linfócitos T/metabolismo , Adenosina Trifosfatases/genética , Alelos , Animais , Células Cultivadas , Aberrações Cromossômicas , DNA Helicases/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , RecQ Helicases , Linfócitos T/imunologia , Timo/citologia
12.
Mol Cell Biol ; 26(17): 6713-26, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16914751

RESUMO

Bloom's syndrome (BS) is a genetic disorder characterized cellularly by increases in sister chromatid exchanges (SCEs) and numbers of micronuclei. BS is caused by mutation in the BLM DNA helicase gene and involves a greatly enhanced risk of developing the range of malignancies seen in the general population. With a mouse model for the disease, we set out to determine the relationship between genomic instability and neoplasia. We used a novel two-step analysis to investigate a panel of eight cell lines developed from mammary tumors that appeared in Blm conditional knockout mice. First, the panel of cell lines was examined for instability. High numbers of SCEs were uniformly seen in members of the panel, and several lines produced chromosomal instability (CIN) manifested by high numbers of chromosomal structural aberrations (CAs) and chromosome missegregation events. Second, to see if Blm mutation was responsible for the CIN, time-dependent analysis was conducted on a tumor line harboring a functional floxed Blm allele. The floxed allele was deleted in vitro, and mutant as well as control subclones were cultured for 100 passages. By passage 100, six of nine mutant subclones had acquired high CIN. Nine mutant subclones produced 50-fold more CAs than did nine control subclones. Finally, chromosome loss preceded the appearance of CIN, suggesting that this loss provides a potential mechanism for the induction of instability in mutant subclones. Such aneuploidy or CIN is a universal feature of neoplasia but has an uncertain function in oncogenesis. Our results show that Blm gene mutation produces this instability, strengthening a role for CIN in the development of human cancer.


Assuntos
Adenosina Trifosfatases/genética , Instabilidade Cromossômica/genética , DNA Helicases/genética , Deleção de Sequência/genética , Alelos , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Células Cultivadas , Cromossomos de Mamíferos/genética , Células Clonais , DNA/genética , Embrião de Mamíferos/citologia , Éxons/genética , Humanos , Integrases/metabolismo , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais , Camundongos , Camundongos Knockout , Células NIH 3T3 , Ploidias , RecQ Helicases , Células-Tronco/metabolismo , Fatores de Tempo
13.
Cancer Cell ; 9(6): 425-34, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16766262

RESUMO

Alterations in cellular metabolism are among the most consistent hallmarks of cancer. Herein we have investigated the relationship between increased aerobic lactate production and mitochondrial physiology in tumor cells. To diminish the ability of malignant cells to metabolize pyruvate to lactate, lactate dehydrogenase A (LDH-A) levels were knocked down by means of LDH-A short hairpin RNAs. Reduction in LDH-A activity resulted in stimulation of mitochondrial respiration and decrease of mitochondrial membrane potential. It also compromised the ability of these tumor cells to proliferate under hypoxia. The tumorigenicity of the LDH-A-deficient cells was severely diminished, and this phenotype was reversed by complementation with the human ortholog LDH-A protein. These results demonstrate that LDH-A plays a key role in tumor maintenance.


Assuntos
Transformação Celular Neoplásica/metabolismo , L-Lactato Desidrogenase/fisiologia , Ácido Láctico/biossíntese , Neoplasias Mamárias Experimentais/metabolismo , Mitocôndrias/fisiologia , Trifosfato de Adenosina/biossíntese , Animais , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/patologia , Feminino , Glucose/metabolismo , Glicólise , Humanos , Indóis/farmacologia , Isoenzimas/biossíntese , Isoenzimas/genética , Isoenzimas/fisiologia , L-Lactato Desidrogenase/biossíntese , L-Lactato Desidrogenase/genética , Lactato Desidrogenase 5 , Ácido Láctico/antagonistas & inibidores , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/patologia , Potenciais da Membrana , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/fisiologia , Transplante de Neoplasias , Fosforilação Oxidativa , Consumo de Oxigênio , Compostos de Piridínio/farmacologia
14.
Exp Cell Res ; 312(7): 1119-26, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16480715

RESUMO

Formin family proteins coordinate actin filaments and microtubules. The mechanisms by which formins bind and regulate the actin cytoskeleton have recently been well defined. However, the molecular mechanism by which formins coordinate actin filaments and microtubules remains poorly understood. We demonstrate here that Isoform-Ib of the Formin-1 protein (Fmn1-Ib) binds to microtubules via a protein domain that is physically separated from the known actin-binding domains. When expressed at low levels in NIH3T3 fibroblasts, Fmn1-Ib protein localizes to cytoplasmic filaments that nocodazole disruption confirmed as interphase microtubules. A series of progressive mutants of Fmn1-Ib demonstrated that deletion of exon-2 caused dissociation from microtubules and a stronger association with actin membrane ruffles. The exon-2-encoded peptide binds purified tubulin in vitro and is also sufficient to localize GFP to microtubules. Exon-2 does not contain any known formin homology domains. Deletion of exon 5, 7, 8, the FH1 domain or FH2 domain did not affect microtubule binding. Thus, our results indicate that exon-2 of Fmn1-Ib encodes a novel microtubule-binding peptide. Since formin proteins associate with actin filaments through the FH1 and FH2 domains, binding to interphase microtubules through this exon-2-encoded domain provides a novel mechanism by which Fmn1-Ib could coordinate actin filaments and microtubules.


Assuntos
Éxons/fisiologia , Proteínas dos Microfilamentos/metabolismo , Microtúbulos/metabolismo , Fragmentos de Peptídeos/metabolismo , Actinas/metabolismo , Animais , Proteínas Fetais , Forminas , Humanos , Camundongos , Proteínas dos Microfilamentos/genética , Células NIH 3T3 , Proteínas Nucleares , Fragmentos de Peptídeos/genética , Estrutura Terciária de Proteína
15.
Cancer Res ; 65(15): 6891-900, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16061673

RESUMO

The HER-2 oncoprotein is commonly overexpressed in a variety of human malignancies and has become an attractive antitumor target. A number of strategies to inhibit the HER-2 receptor tyrosine kinase are currently the focus of intensive preclinical and clinical research. In the present study, we have engineered a bifunctional peptide, BHAP, which consists of two modular domains: a HER-2-targeting/neutralizing domain and a mitochondriotoxic, proapoptotic domain. The chimeric peptide is biologically active and capable of selectively triggering apoptosis of HER-2-overexpressing cancer cells in culture, even those previously described as Herceptin resistant. Furthermore, BHAP slows down growth of HER-2-overexpressing human mammary xenografts established in SCID mice. This approach can be extended to the development of tailored targeted chimeric peptides against a number of overexpressed cellular receptors implicated in the development and progression of cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Mitocôndrias/efeitos dos fármacos , Peptídeos/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos SCID , Mitocôndrias/fisiologia , Dados de Sequência Molecular , Peptídeos/genética , Peptídeos/farmacocinética , Engenharia de Proteínas , Estrutura Terciária de Proteína , Receptor ErbB-2/biossíntese , Receptor ErbB-2/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Hum Mol Genet ; 14(5): 615-25, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15649944

RESUMO

Thalassemia is a disease caused by a variety of mutations affecting both the adult and embryonic alpha- and beta-globin loci. A mouse strain carrying an embryonic zeta-globin gene disrupted by the insertion of a PGK-Neo cassette displays an alpha-thalassemia-like syndrome. Embryonic survival of this zeta-null mouse is variable and strongly influenced by genetic background, the 129/SvEv mouse strain displaying a more severe phenotype than C57BL/6. We have identified two modifying loci on C57BL/6 chromosomes 2 and 5, which affect the penetrance of embryonic lethality in the 129/SvEv mouse. Through this work, we were able to observe an interesting effect on somatic recombination events in thalassemic embryos. We show that these events can occur on multiple chromosomes in very early embryonic cells, prior to their allocation to the germline. Our results demonstrate that somatic recombination events can be transmitted to subsequent generations.


Assuntos
Variação Genética , Globinas/genética , Recombinação Genética , Talassemia/genética , Animais , Mapeamento Cromossômico , Epistasia Genética , Feminino , Marcadores Genéticos , Globinas/metabolismo , Perda de Heterozigosidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linhagem , Fenótipo , Talassemia/metabolismo
17.
Mol Cancer Res ; 2(10): 551-6, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15498929

RESUMO

The majority of patients who succumb to cancer die from metastatic disease progression rather than from the primary tumor. Elucidation of the mechanisms underlying tissue-specific metastasis is essential to the development of effective therapies. The mitogen-activated protein kinase kinase (MEK) pathway is frequently activated in human tumors and has been shown to regulate genes involved in proliferation, migration, and invasion. Studies with MEK-transformed EpH4 mouse mammary epithelial cells showed that these cells are highly tumorigenic but have a limited metastatic ability. Detachment of epithelial cells from the extracellular matrix causes disruption of the actin cytoskeleton and induces apoptosis. Several metastatic breast carcinoma cell lines have been shown to be resistant to cell death following actin disruption. This death-resistant phenotype can be modeled by overexpressing the antiapoptotic Bcl-2 protein in cells. This suggests that mechanisms that regulate survival of extravasated tumor cells may enhance metastatic efficiency. Therefore, we examined whether expression of Bcl-2 in MEK-transformed EpH4 mammary epithelial cells could provide a survival advantage and promote metastasis. Expression of Bcl-2 in parental EpH4 mammary epithelial cells or MEK-transformed cells was insufficient to induce increased migration, invasion, or tumor development. However, Bcl-2 expression markedly enhanced spontaneous lung metastasis from orthotopically implanted primary tumors. These results clearly show that mechanisms that regulate primary tumor development are distinct from those that promote metastasis and that assays designed to isolate genes involved in transformation may fail to identify genes that are critical regulators of metastasis.


Assuntos
Neoplasias da Mama/patologia , Neoplasias Pulmonares/secundário , Glândulas Mamárias Animais/citologia , Metástase Neoplásica/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Animais , Neoplasias da Mama/fisiopatologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Células Epiteliais/citologia , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Neoplasias Pulmonares/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
18.
Immunity ; 21(1): 43-53, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15345219

RESUMO

The developmental origin of type I interferon (IFN)-producing plasmacytoid dendritic cells (PDCs) is controversial. In particular, the rearrangement of immunoglobulin heavy chain (IgH) genes in murine PDCs and the expression of pre-T cell receptor alpha (pTalpha) gene by human PDCs were proposed as evidence for their "lymphoid" origin. Here we demonstrate that PDCs capable of IFN production develop efficiently from both myeloid- and lymphoid-committed progenitors. Rearranged IgH genes as well as RAG transcripts were found in both myeloid- and lymphoid-derived PDCs. The human pTalpha transgenic reporter was activated in both myeloid- and lymphoid-derived PDCs at a level comparable to pre-T cells. PDCs were the only cell population that activated murine RAG1 knockin and human pTalpha transgenic reporters outside the lymphoid lineage. These results highlight a unique developmental program of PDCs that distinguishes them from other cell types including conventional dendritic cells.


Assuntos
Células da Medula Óssea/fisiologia , Células Dendríticas/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Plasmócitos/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Rearranjo Gênico de Cadeia Pesada de Linfócito B , Proteínas de Homeodomínio/metabolismo , Humanos , Interferon Tipo I/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T alfa-beta
19.
Oncogene ; 23(26): 4641-5, 2004 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-15064711

RESUMO

Many mouse models of breast cancer form large primary tumors that rarely metastasize. Models with aggressive metastasis express oncoproteins that simultaneously affect growth and apoptosis pathways. To define the role of apoptotic resistance and to model a challenge faced by tumor cells during metastatic dissemination, we focused on apoptosis induced by cell shape change. Inhibiting actin polymerization with Latrunculin-A causes cell rounding and death within hours in nontumorigenic human 10A-Ras mammary epithelial cells. In contrast, MDA-MB-231 metastatic breast tumor cells resist LA-induced death, and survive for days despite cell rounding. Infecting 10A-Ras cells with a MDA-MB-231 retroviral expression library, and selecting with Latrunculin-A repeatedly identified Bcl-xL as a suppressor of cytoskeleton-dependent death. Although Bcl-xL enhances the spread of metastatic breast tumor cell lines, the distinct effects of apoptotic resistance on tumor growth in the mammary gland and during metastasis have not been compared directly. We find that Bcl-xL overexpression in mouse mammary epithelial cells does not induce primary tumor formation or enhance MEK-induced tumorigenesis within the mammary gland environment. However, it strongly enhances metastatic potential. These results with Bcl-xL provide novel evidence that isolated apoptotic resistance can increase metastatic potential, but remain overlooked by assays based on breast tumor growth.


Assuntos
Neoplasias da Mama/patologia , Citoesqueleto/genética , Metástase Neoplásica/genética , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Animais , Neoplasias da Mama/tratamento farmacológico , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Técnicas Genéticas , Vetores Genéticos , Humanos , MAP Quinase Quinase 1 , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Tiazóis/farmacologia , Tiazolidinas , Proteína bcl-X
20.
Cancer Res ; 64(1): 329-36, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14729642

RESUMO

Mutations that lead to the emergence of resistance to apoptosis are commonly observed among tumor cells. Some of the proteins affected are integral parts of the apoptotic cascade such as pro- and antiapoptotic members of the Bcl-2 family. F16 is a small molecule that accumulates in mitochondria of a variety of tumor cells and interferes with their physiological function. Because this interference ultimately triggers apoptosis in many affected cell lines, we examined the effect of antiapoptotic Bcl-2 overexpression on the response of cells to F16. Our results showed that high levels of Bcl-2 did not block the ability of F16 to induce cell death. However, unlike the apoptotic response that followed F16 treatment of cells with moderate Bcl-2 levels, cells resistant to a variety of apoptotic stimuli by virtue of Bcl-2 overexpression succumbed to F16 by necrosis. Thus, this dual ability of the mitochondriotoxic compound F16 to induce apoptosis and necrosis may represent an added advantage by expanding its spectrum of action toward genetically altered tumor cells incapable of apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma/genética , Carcinoma/patologia , Indóis/toxicidade , Mitocôndrias/efeitos dos fármacos , Compostos de Piridínio/toxicidade , Inibidores de Caspase , Morte Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores de Cisteína Proteinase/farmacologia , Citometria de Fluxo , Homeostase , Humanos , Mitocôndrias/patologia , Necrose , Oxirredução , Proteínas Proto-Oncogênicas c-bcl-2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA