Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
G3 (Bethesda) ; 10(5): 1503-1510, 2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32152007

RESUMO

The UCSF Mouse Inventory Database Application is an open-source Web App that provides information about the mutant alleles, transgenes, and inbred strains maintained by investigators at the university and facilitates sharing of these resources within the university community. The Application is designed to promote collaboration, decrease the costs associated with obtaining genetically-modified mice, and increase access to mouse lines that are difficult to obtain. An inventory of the genetically-modified mice on campus and the investigators who maintain them is compiled from records of purchases from external sources, transfers from researchers within and outside the university, and from data provided by users. These data are verified and augmented with relevant information harvested from public databases, and stored in a succinct, searchable database secured on the university network. Here we describe this resource and provide information about how to implement and maintain such a mouse inventory database application at other institutions.


Assuntos
Aplicativos Móveis , Alelos , Animais , Bases de Dados Factuais , Internet , Camundongos , Transgenes
2.
Dev Biol ; 459(1): 61-64, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31877273

RESUMO

Throughout my professional life I was an academic researcher who had a successful career in developmental biology. When I started the process of closing my lab and retiring from the University, I came to the conclusion that I wanted to do something completely different. I trained for and became a docent at the Asian Art Museum of San Francisco. I apply the skills I acquired as a scientist on a regular basis in my work at the museum, often in unexpected ways. In this article, I discuss my journey from the lab to the museum and what I learned along the way.


Assuntos
Arte , Escolha da Profissão , Mobilidade Ocupacional , Biologia do Desenvolvimento , Museus , Pesquisadores , Aposentadoria , Feminino , Amigos , Humanos , Revisão por Pares
3.
J Clin Invest ; 124(9): 4093-101, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25133424

RESUMO

Endocrine cell proliferation fluctuates dramatically in response to signals that communicate hormone demand. The genetic alterations that override these controls in endocrine tumors often are not associated with oncogenes common to other tumor types, suggesting that unique pathways govern endocrine proliferation. Within the pancreas, for example, activating mutations of the prototypical oncogene KRAS drive proliferation in all pancreatic ductal adenocarcimomas but are never found in pancreatic endocrine tumors. Therefore, we asked how cellular context impacts K-RAS signaling. We found that K-RAS paradoxically suppressed, rather than promoted, growth in pancreatic endocrine cells. Inhibition of proliferation by K-RAS depended on antiproliferative RAS effector RASSF1A and blockade of the RAS-activated proproliferative RAF/MAPK pathway by tumor suppressor menin. Consistent with this model, a glucagon-like peptide 1 (GLP1) agonist, which stimulates ERK1/2 phosphorylation, did not affect endocrine cell proliferation by itself, but synergistically enhanced proliferation when combined with a menin inhibitor. In contrast, inhibition of MAPK signaling created a synthetic lethal interaction in the setting of menin loss. These insights suggest potential strategies both for regenerating pancreatic ß cells for people with diabetes and for targeting menin-sensitive endocrine tumors.


Assuntos
Ilhotas Pancreáticas/citologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas ras/fisiologia , Adulto , Animais , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Fosforilação , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Transdução de Sinais , Proteínas Supressoras de Tumor/fisiologia
4.
Proc Natl Acad Sci U S A ; 109(49): 20023-8, 2012 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-23150596

RESUMO

Expression of Sprouty genes is frequently decreased or absent in human prostate cancer, implicating them as suppressors of tumorigenesis. Here we show they function in prostate tumor suppression in the mouse. Concomitant inactivation of Spry1 and Spry2 in prostate epithelium causes ductal hyperplasia and low-grade prostatic intraepithelial neoplasia (PIN). However, when Spry1 and Spry2 loss-of-function occurs in the context of heterozygosity for a null allele of the tumor suppressor gene Pten, there is a striking increase in PIN and evidence of neoplastic invasion. Conversely, expression of a Spry2 gain-of-function transgene in Pten null prostatic epithelium suppresses the tumorigenic effects of loss of Pten function. We show that Sprouty gene loss-of-function results in hyperactive RAS/ERK1/2 signaling throughout the prostate epithelium and cooperates with heterozygosity for a Pten null allele to promote hyperactive PI3K/AKT signaling. Furthermore, Spry2 gain-of-function can suppress hyperactivation of AKT caused by the absence of PTEN. Together, these results point to a key genetic interaction between Sprouty genes and Pten in prostate tumorigenesis and provide strong evidence that Sprouty genes can function to modulate signaling via the RAS/ERK1/2 and PI3K/AKT pathways. The finding that Sprouty genes suppress tumorigenesis caused by Pten loss-of-function suggests that therapeutic approaches aimed at restoring normal feedback mechanisms triggered by receptor tyrosine kinase signaling, including Sprouty gene expression, may provide an effective strategy to delay or prevent high-grade PIN and invasive prostate cancer.


Assuntos
Genes Supressores de Tumor/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas de Membrana/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Fosfoproteínas/fisiologia , Neoplasia Prostática Intraepitelial/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Primers do DNA/genética , Imunofluorescência , Técnicas Histológicas , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intracelular , Microdissecção e Captura a Laser , Masculino , Proteínas de Membrana/deficiência , Camundongos , PTEN Fosfo-Hidrolase/genética , Fosfoproteínas/deficiência , Reação em Cadeia da Polimerase , Neoplasia Prostática Intraepitelial/fisiopatologia , Proteínas Serina-Treonina Quinases , Proteínas ras/metabolismo
6.
Science ; 333(6040): 342-345, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21764747

RESUMO

During early lung development, airway tubes change shape. Tube length increases more than circumference as a large proportion of lung epithelial cells divide parallel to the airway longitudinal axis. We show that this bias is lost in mutants with increased extracellular signal-regulated kinase 1 (ERK1) and ERK2 activity, revealing a link between the ERK1/2 signaling pathway and the control of mitotic spindle orientation. Using a mathematical model, we demonstrate that change in airway shape can occur as a function of spindle angle distribution determined by ERK1/2 signaling, independent of effects on cell proliferation or cell size and shape. We identify sprouty genes, which encode negative regulators of fibroblast growth factor 10 (FGF10)-mediated RAS-regulated ERK1/2 signaling, as essential for controlling airway shape change during development through an effect on mitotic spindle orientation.


Assuntos
Pulmão/embriologia , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Morfogênese , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Mucosa Respiratória/embriologia , Fuso Acromático/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Polaridade Celular , Proliferação de Células , Forma Celular , Tamanho Celular , Células Epiteliais/citologia , Fator 10 de Crescimento de Fibroblastos/genética , Fator 10 de Crescimento de Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Pulmão/citologia , Pulmão/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mitose , Modelos Biológicos , Mutação , Organogênese , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas p21(ras)/genética , Mucosa Respiratória/citologia , Fuso Acromático/ultraestrutura
7.
Development ; 138(10): 1913-23, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21471156

RESUMO

The essential roles of SHH in anteroposterior (AP) and AER-FGF signalling in proximodistal (PD) limb bud development are well understood. In addition, these morphoregulatory signals are key components of the self-regulatory SHH/GREM1/AER-FGF feedback signalling system that regulates distal progression of limb bud development. This study uncovers an additional signalling module required for coordinated progression of limb bud axis development. Transcriptome analysis using Shh-deficient mouse limb buds revealed that the expression of proximal genes was distally extended from early stages onwards, which pointed to a more prominent involvement of SHH in PD limb axis development. In particular, retinoic acid (RA) target genes were upregulated proximally, while the expression of the RA-inactivating Cyp26b1 enzyme was downregulated distally, pointing to increased RA activity in Shh-deficient mouse limb buds. Further genetic and molecular analysis established that Cyp26b1 expression is regulated by AER-FGF signalling. During initiation of limb bud outgrowth, the activation of Cyp26b1 expression creates a distal 'RA-free' domain, as indicated by complementary downregulation of a transcriptional sensor of RA activity. Subsequently, Cyp26b1 expression increases as a consequence of SHH-dependent upregulation of AER-FGF signalling. To better understand the underlying signalling interactions, computational simulations of the spatiotemporal expression patterns and interactions were generated. These simulations predicted the existence of an antagonistic AER-FGF/CYP26B1/RA signalling module, which was verified experimentally. In summary, SHH promotes distal progression of limb development by enhancing CYP26B1-mediated RA clearance as part of a signalling network linking the SHH/GREM1/AER-FGF feedback loop to the newly identified AER-FGF/CYP26B1/RA module.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Botões de Extremidades/embriologia , Botões de Extremidades/metabolismo , Tretinoína/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/genética , Ectoderma/embriologia , Ectoderma/metabolismo , Ativação Enzimática , Retroalimentação Fisiológica , Feminino , Fatores de Crescimento de Fibroblastos/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Ácido Retinoico 4 Hidroxilase , Transdução de Sinais
8.
Development ; 137(22): 3753-61, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20978073

RESUMO

In many organ systems such as the skin, gastrointestinal tract and hematopoietic system, homeostasis is dependent on the continuous generation of differentiated progeny from stem cells. The rodent incisor, unlike human teeth, grows throughout the life of the animal and provides a prime example of an organ that rapidly deteriorates if newly differentiated cells cease to form from adult stem cells. Hedgehog (Hh) signaling has been proposed to regulate self-renewal, survival, proliferation and/or differentiation of stem cells in several systems, but to date there is little evidence supporting a role for Hh signaling in adult stem cells. We used in vivo genetic lineage tracing to identify Hh-responsive stem cells in the mouse incisor and we show that sonic hedgehog (SHH), which is produced by the differentiating progeny of the stem cells, signals to several regions of the incisor. Using a hedgehog pathway inhibitor (HPI), we demonstrate that Hh signaling is not required for stem cell survival but is essential for the generation of ameloblasts, one of the major differentiated cell types in the tooth, from the stem cells. These results therefore reveal the existence of a positive-feedback loop in which differentiating progeny produce the signal that in turn allows them to be generated from stem cells.


Assuntos
Células-Tronco Adultas/metabolismo , Ameloblastos/citologia , Proteínas Hedgehog/metabolismo , Incisivo/crescimento & desenvolvimento , Camundongos/fisiologia , Transdução de Sinais , Ameloblastos/metabolismo , Animais , Diferenciação Celular , Células Epiteliais/metabolismo , Feminino , Proteínas Hedgehog/antagonistas & inibidores , Incisivo/citologia
9.
Proc Natl Acad Sci U S A ; 107(33): 14662-7, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20675583

RESUMO

Electrical cardiac forces have been previously hypothesized to play a significant role in cardiac morphogenesis and remodeling. In response to electrical forces, cultured cardiomyocytes rearrange their cytoskeletal structure and modify their gene expression profile. To translate such in vitro data to the intact heart, we used a collection of zebrafish cardiac mutants and transgenics to investigate whether cardiac conduction could influence in vivo cardiac morphogenesis independent of contractile forces. We show that the cardiac mutant dco(s226) develops heart failure and interrupted cardiac morphogenesis following uncoordinated ventricular contraction. Using in vivo optical mapping/calcium imaging, we determined that the dco cardiac phenotype was primarily due to aberrant ventricular conduction. Because cardiac contraction and intracardiac hemodynamic forces can also influence cardiac development, we further analyzed the dco phenotype in noncontractile hearts and observed that disorganized ventricular conduction could affect cardiomyocyte morphology and subsequent heart morphogenesis in the absence of contraction or flow. By positional cloning, we found that dco encodes Gja3/Cx46, a gap junction protein not previously implicated in heart formation or function. Detailed analysis of the mouse Cx46 mutant revealed the presence of cardiac conduction defects frequently associated with human heart failure. Overall, these in vivo studies indicate that cardiac electrical forces are required to preserve cardiac chamber morphology and may act as a key epigenetic factor in cardiac remodeling.


Assuntos
Embrião não Mamífero/fisiologia , Sistema de Condução Cardíaco/fisiologia , Coração/fisiologia , Miocárdio/metabolismo , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Conexinas/classificação , Conexinas/genética , Conexinas/metabolismo , Eletrocardiografia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/fisiologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Coração/embriologia , Hibridização In Situ , Camundongos , Camundongos Knockout , Microscopia Confocal , Dados de Sequência Molecular , Mutação , Filogenia , Homologia de Sequência de Aminoácidos , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
10.
PLoS Genet ; 6(1): e1000809, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20084103

RESUMO

GDNF signaling through the Ret receptor tyrosine kinase (RTK) is required for ureteric bud (UB) branching morphogenesis during kidney development in mice and humans. Furthermore, many other mutant genes that cause renal agenesis exert their effects via the GDNF/RET pathway. Therefore, RET signaling is believed to play a central role in renal organogenesis. Here, we re-examine the extent to which the functions of Gdnf and Ret are unique, by seeking conditions in which a kidney can develop in their absence. We find that in the absence of the negative regulator Spry1, Gdnf, and Ret are no longer required for extensive kidney development. Gdnf-/-;Spry1-/- or Ret-/-;Spry1-/- double mutants develop large kidneys with normal ureters, highly branched collecting ducts, extensive nephrogenesis, and normal histoarchitecture. However, despite extensive branching, the UB displays alterations in branch spacing, angle, and frequency. UB branching in the absence of Gdnf and Spry1 requires Fgf10 (which normally plays a minor role), as removal of even one copy of Fgf10 in Gdnf-/-;Spry1-/- mutants causes a complete failure of ureter and kidney development. In contrast to Gdnf or Ret mutations, renal agenesis caused by concomitant lack of the transcription factors ETV4 and ETV5 is not rescued by removing Spry1, consistent with their role downstream of both RET and FGFRs. This shows that, for many aspects of renal development, the balance between positive signaling by RTKs and negative regulation of this signaling by SPRY1 is more critical than the specific role of GDNF. Other signals, including FGF10, can perform many of the functions of GDNF, when SPRY1 is absent. But GDNF/RET signaling has an apparently unique function in determining normal branching pattern. In contrast to GDNF or FGF10, Etv4 and Etv5 represent a critical node in the RTK signaling network that cannot by bypassed by reducing the negative regulation of upstream signals.


Assuntos
Fator 10 de Crescimento de Fibroblastos/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Rim/crescimento & desenvolvimento , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Feminino , Fator 10 de Crescimento de Fibroblastos/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Rim/embriologia , Rim/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organogênese , Fosfoproteínas/genética , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Transdução de Sinais , Ureter/anormalidades , Ureter/metabolismo
11.
Blood ; 113(11): 2478-87, 2009 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-19147787

RESUMO

B-cell lymphoma is the most common immune system malignancy. TCL1 transgenic mice (TCL1-tg), in which TCL1 is ectopically expressed in mature lymphocytes, develop multiple B- and T-cell leukemia and lymphoma subtypes, supporting an oncogenic role for TCL1 that probably involves AKT and MAPK-ERK signaling pathway augmentation. Additional, largely unknown genetic and epigenetic alterations cooperate with TCL1 during lymphoma progression. We examined DNA methylation patterns in TCL1-tg B-cell tumors to discover tumor-associated epigenetic changes, and identified hypermethylation of sprouty2 (Spry2). Sprouty proteins are context-dependent negative or positive regulators of MAPK-ERK pathway signaling, but their role(s) in B-cell physiology or pathology are unknown. Here we show that repression of Spry2 expression in TCL1-tg mouse and human B-cell lymphomas and cell lines is associated with dense DNA hypermethylation and was reversed by inhibition of DNA methylation. Spry2 expression was induced in normal splenic B cells by CD40/B-cell receptor costimulation and regulated a negative feedback loop that repressed MAPK-ERK signaling and decreased B-cell viability. Conversely, loss of Spry2 function hyperactivated MAPK-ERK signaling and caused increased B-cell proliferation. Combined, these results implicate epigenetic silencing of Spry2 expression in B lymphoma progression and suggest it as a companion lesion to ectopic TCL1 expression in enhancing MAPK-ERK pathway signaling.


Assuntos
Linfócitos B/fisiologia , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Linfoma de Células B/genética , Proteínas de Membrana/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linfócitos B/metabolismo , Antígenos CD40/metabolismo , Antígenos CD40/fisiologia , Metilação de DNA/fisiologia , Feminino , Inativação Gênica/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Serina-Treonina Quinases , Células Tumorais Cultivadas
12.
Nature ; 456(7224): 980-4, 2008 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19043405

RESUMO

MicroRNAs comprise a broad class of small non-coding RNAs that control expression of complementary target messenger RNAs. Dysregulation of microRNAs by several mechanisms has been described in various disease states including cardiac disease. Whereas previous studies of cardiac disease have focused on microRNAs that are primarily expressed in cardiomyocytes, the role of microRNAs expressed in other cell types of the heart is unclear. Here we show that microRNA-21 (miR-21, also known as Mirn21) regulates the ERK-MAP kinase signalling pathway in cardiac fibroblasts, which has impacts on global cardiac structure and function. miR-21 levels are increased selectively in fibroblasts of the failing heart, augmenting ERK-MAP kinase activity through inhibition of sprouty homologue 1 (Spry1). This mechanism regulates fibroblast survival and growth factor secretion, apparently controlling the extent of interstitial fibrosis and cardiac hypertrophy. In vivo silencing of miR-21 by a specific antagomir in a mouse pressure-overload-induced disease model reduces cardiac ERK-MAP kinase activity, inhibits interstitial fibrosis and attenuates cardiac dysfunction. These findings reveal that microRNAs can contribute to myocardial disease by an effect in cardiac fibroblasts. Our results validate miR-21 as a disease target in heart failure and establish the therapeutic efficacy of microRNA therapeutic intervention in a cardiovascular disease setting.


Assuntos
Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Fibroblastos/metabolismo , Sistema de Sinalização das MAP Quinases , MicroRNAs/genética , Animais , Cardiomiopatias/patologia , Cardiomiopatias/terapia , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Inativação Gênica , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Ratos
13.
Dev Biol ; 321(1): 77-87, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18585375

RESUMO

FGF signaling is associated with breast cancer and is required for mammary placode formation in the mouse. In this study, we employed a genetic mosaic analysis based on Cre-mediated recombination to investigate FGF receptor 2 (Fgfr2) function in the postnatal mammary gland. Mosaic inactivation of Fgfr2 by the MMTV-Cre transgene enabled us to compare the behavior of Fgfr2 null and Fgfr2 heterozygous cells in the same gland. Fgfr2 null cells were at a competitive disadvantage to their Fgfr2 heterozygous neighbors in the highly proliferative terminal end buds (TEBs) at the invasion front, owing to a negative effect of loss of Fgfr2 function on cell proliferation. However, Fgfr2 null cells were tolerated in mature ducts. In these genetic mosaic mammary glands, the epithelial network is apparently built by TEBs that over time are composed of a progressively larger proportion of Fgfr2-positive cells. However, subsequently, most cells lose Fgfr2 function, presumably due to additional rounds of Cre-mediated recombination. Using an independent strategy to create mosaic mammary glands, which employed an adenovirus-Cre that acts only once, we confirmed that Fgfr2 null cells were out-competed by neighboring Fgfr2 heterozygous cells. Together, our data demonstrate that Fgfr2 functions in the proliferating and invading TEBs, but it is not required in the mature ducts of the pubertal mammary gland.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Glândulas Mamárias Animais/metabolismo , Camundongos , Morfogênese , Maturidade Sexual
14.
J Neurosci ; 28(19): 4938-48, 2008 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-18463247

RESUMO

Mitochondrial dysfunction is commonly associated with neurodegeneration in the aging brain. In addition, the importance of mitochondrial function during brain development is illustrated by the neurological deficits observed in infants with mitochondrial complex deficiencies. However, the extent to which abnormalities in mitochondrial function might impact neurogenesis during brain development is not well understood. Previously, we demonstrated that adult harlequin (Hq) mutant mice, which have an 80% reduction in the mitochondrial protein apoptosis-inducing factor (AIF), exhibited signs of oxidative stress and progressive loss of adult cerebellar and retinal neurons. To assess whether in addition to its role in postmitotic neuron survival Aif is also necessary for cerebellar development, we analyzed embryos in which Aif was deleted in the prospective midbrain and cerebellum at a very early stage of development using an En1 (engrailed 1) promoter-driven cre recombinase gene. These mutant mice, which died at birth, had midbrain defects and dramatic deficits in cerebellar Purkinje and granule cell precursors. Additional analysis revealed that Aif-null Purkinje cell precursors prematurely entered S-phase, but most failed to undergo mitosis and ultimately died via apoptosis. In contrast, proliferation of mutant granule cell precursors was blocked before S-phase. Mice in which Aif was deleted later in embryogenesis using a nestin promoter-driven cre gene survive for several days after birth, and postnatal granule cell precursors in these mice also failed to enter S-phase. Our results indicate that the loss of Aif results in cell cycle abnormalities in a neuron-specific manner during cerebellar development.


Assuntos
Fator de Indução de Apoptose/fisiologia , Cerebelo/embriologia , Animais , Apoptose/fisiologia , Fator de Indução de Apoptose/deficiência , Fator de Indução de Apoptose/genética , Ciclo Celular/fisiologia , Desenvolvimento Embrionário/fisiologia , Fase G1/fisiologia , Deleção de Genes , Mesencéfalo/embriologia , Camundongos , Camundongos Mutantes , Neurônios/fisiologia , Células de Purkinje/citologia , Fase S/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Tempo
15.
Nature ; 453(7196): 745-50, 2008 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-18463632

RESUMO

Mammalian lungs are branched networks containing thousands to millions of airways arrayed in intricate patterns that are crucial for respiration. How such trees are generated during development, and how the developmental patterning information is encoded, have long fascinated biologists and mathematicians. However, models have been limited by a lack of information on the normal sequence and pattern of branching events. Here we present the complete three-dimensional branching pattern and lineage of the mouse bronchial tree, reconstructed from an analysis of hundreds of developmental intermediates. The branching process is remarkably stereotyped and elegant: the tree is generated by three geometrically simple local modes of branching used in three different orders throughout the lung. We propose that each mode of branching is controlled by a genetically encoded subroutine, a series of local patterning and morphogenesis operations, which are themselves controlled by a more global master routine. We show that this hierarchical and modular programme is genetically tractable, and it is ideally suited to encoding and evolving the complex networks of the lung and other branched organs.


Assuntos
Padronização Corporal/fisiologia , Pulmão/anatomia & histologia , Pulmão/embriologia , Organogênese/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Padronização Corporal/genética , Fator 10 de Crescimento de Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Pulmão/citologia , Pulmão/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Modelos Biológicos , Organogênese/genética , Proteínas Serina-Treonina Quinases , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo
16.
Nature ; 453(7193): 401-5, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18449196

RESUMO

Half a century ago, the apical ectodermal ridge (AER) at the distal tip of the tetrapod limb bud was shown to produce signals necessary for development along the proximal-distal (P-D) axis, but how these signals influence limb patterning is still much debated. Fibroblast growth factor (FGF) gene family members are key AER-derived signals, with Fgf4, Fgf8, Fgf9 and Fgf17 expressed specifically in the mouse AER. Here we demonstrate that mouse limbs lacking Fgf4, Fgf9 and Fgf17 have normal skeletal pattern, indicating that Fgf8 is sufficient among AER-FGFs to sustain normal limb formation. Inactivation of Fgf8 alone causes a mild skeletal phenotype; however, when we also removed different combinations of the other AER-FGF genes, we obtained unexpected skeletal phenotypes of increasing severity, reflecting the contribution that each FGF can make to the total AER-FGF signal. Analysis of the compound mutant limb buds revealed that, in addition to sustaining cell survival, AER-FGFs regulate P-D-patterning gene expression during early limb bud development, providing genetic evidence that AER-FGFs function to specify a distal domain and challenging the long-standing hypothesis that AER-FGF signalling is permissive rather than instructive for limb patterning. We discuss how a two-signal model for P-D patterning can be integrated with the concept of early specification to explain the genetic data presented here.


Assuntos
Padronização Corporal/genética , Padronização Corporal/fisiologia , Fator 8 de Crescimento de Fibroblasto/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Botões de Extremidades/embriologia , Animais , Osso e Ossos/embriologia , Osso e Ossos/metabolismo , Sobrevivência Celular , Feminino , Fator 8 de Crescimento de Fibroblasto/deficiência , Fator 8 de Crescimento de Fibroblasto/genética , Fatores de Crescimento de Fibroblastos/deficiência , Fatores de Crescimento de Fibroblastos/genética , Proteínas de Homeodomínio/genética , Botões de Extremidades/citologia , Botões de Extremidades/metabolismo , Masculino , Camundongos , Proteína Meis1 , Proteínas de Neoplasias/genética , Tamanho do Órgão , Transdução de Sinais
17.
Genesis ; 46(2): 69-73, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18257103

RESUMO

Cre-mediated recombination, a method widely used in mice for tissue-specific inactivation of endogenous genes or activation of transgenes, is critically dependent on the availability of mouse lines in which Cre recombinase functions in the tissue of interest or its progenitors. Here we describe a transgenic mouse line, Osr1-cre, in which Cre is active from embryonic day (E)11.5 in a few specific tissues. These include the endoderm of the posterior foregut, midgut, hindgut, and developing urogenital system, the heart left atrium, extra-ocular muscle progenitors, and mesenchyme in particular regions of the limb. Furthermore, starting at E12.5, Cre functions in limb interdigital mesenchyme. Within the urogenital system, recombination appears to be virtually complete in the epithelium of the bladder and urethra just posterior to it by E14.5. In males, some of these urethral cells form the prostate. The spatiotemporal pattern of Cre activity in Osr1-cre makes it a unique resource among the lines available for Cre-mediated recombination experiments.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Técnicas Genéticas , Integrases/genética , Animais , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos
18.
Development ; 135(5): 889-98, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18216176

RESUMO

Prospective midbrain and cerebellum formation are coordinated by FGF ligands produced by the isthmic organizer. Previous studies have suggested that midbrain and cerebellum development require different levels of FGF signaling. However, little is known about the extent to which specific regions within these two parts of the brain differ in their requirement for FGF signaling during embryogenesis. Here, we have explored the effects of inhibiting FGF signaling within the embryonic mouse midbrain (mesencephalon) and cerebellum (rhombomere 1) by misexpressing sprouty2 (Spry2) from an early stage. We show that such Spry2 misexpression moderately reduces FGF signaling, and that this reduction causes cell death in the anterior mesencephalon, the region furthest from the source of FGF ligands. Interestingly, the remaining mesencephalon cells develop into anterior midbrain, indicating that a low level of FGF signaling is sufficient to promote only anterior midbrain development. Spry2 misexpression also affects development of the vermis, the part of the cerebellum that spans the midline. We found that, whereas misexpression of Spry2 alone caused loss of the anterior vermis, reducing FGF signaling further, by decreasing Fgf8 gene dose, resulted in loss of the entire vermis. Our data suggest that cell death is not responsible for vermis loss, but rather that it fails to develop because reducing FGF signaling perturbs the balance between vermis and roof plate development in rhombomere 1. We suggest a molecular explanation for this phenomenon by providing evidence that FGF signaling functions to inhibit the BMP signaling that promotes roof plate development.


Assuntos
Cerebelo/embriologia , Embrião de Mamíferos/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Mesencéfalo/embriologia , Animais , Morte Celular , Cerebelo/anatomia & histologia , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Integrases/metabolismo , Mesencéfalo/anatomia & histologia , Camundongos , Transdução de Sinais
19.
Development ; 135(2): 377-85, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18077585

RESUMO

Rodent incisors grow throughout adult life, but are prevented from becoming excessively long by constant abrasion, which is facilitated by the absence of enamel on one side of the incisor. Here we report that loss-of-function of sprouty genes, which encode antagonists of receptor tyrosine kinase signaling, leads to bilateral enamel deposition, thus impeding incisor abrasion and resulting in unchecked tooth elongation. We demonstrate that sprouty genes function to ensure that enamel-producing ameloblasts are generated on only one side of the tooth by inhibiting the formation of ectopic ameloblasts from self-renewing stem cells, and that they do so by preventing the establishment of an epithelial-mesenchymal FGF signaling loop. Interestingly, although inactivation of Spry4 alone initiates ectopic ameloblast formation in the embryo, the dosage of another sprouty gene must also be reduced to sustain it after birth. These data reveal that the generation of differentiated progeny from a particular stem cell population can be differently regulated in the embryo and adult.


Assuntos
Diferenciação Celular , Fatores de Crescimento de Fibroblastos/metabolismo , Incisivo/citologia , Incisivo/embriologia , Transdução de Sinais , Células-Tronco/citologia , Proteínas Adaptadoras de Transdução de Sinal , Ameloblastos/citologia , Ameloblastos/metabolismo , Animais , Proliferação de Células , Esmalte Dentário/citologia , Esmalte Dentário/embriologia , Esmalte Dentário/metabolismo , Epitélio/embriologia , Epitélio/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Incisivo/anormalidades , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Modelos Biológicos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Especificidade de Órgãos , Fenótipo , Proteínas Serina-Treonina Quinases , Regulação para Cima/genética
20.
Development ; 134(13): 2501-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17537792

RESUMO

Development of the metanephric kidney crucially depends on proper interactions between cells and the surrounding extracellular matrix. For example, we showed previously that in the absence of alpha8beta1 integrin, invasion by the ureteric bud into the metanephric mesenchyme is inhibited, resulting in renal agenesis. Here we present genetic evidence that the extracellular matrix protein nephronectin is an essential ligand that engages alpha8beta1 integrin during early kidney development. We show that embryos lacking a functional nephronectin gene frequently display kidney agenesis or hypoplasia, which can be traced to a delay in the invasion of the metanephric mesenchyme by the ureteric bud at an early stage of kidney development. Significantly, we detected no defects in extracellular matrix organization in the nascent kidneys of the nephronectin mutants. Instead, we found that Gdnf expression was dramatically reduced in both nephronectin- and alpha8 integrin-null mutants specifically in the metanephric mesenchyme at the time of ureteric bud invasion. We show that this reduction is sufficient to explain the agenesis and hypoplasia observed in both mutants. Interestingly, the reduction in Gdnf expression is transient, and its resumption presumably enables the nephronectin-deficient ureteric buds to invade the metanephric mesenchyme and begin branching. Our results thus place nephronectin and alpha8beta1 integrin in a pathway that regulates Gdnf expression and is essential for kidney development.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Integrinas/metabolismo , Rim/embriologia , Rim/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Alelos , Animais , Animais Recém-Nascidos , Membrana Basal/embriologia , Membrana Basal/metabolismo , Proteínas da Matriz Extracelular/deficiência , Proteínas da Matriz Extracelular/genética , Feminino , Redes Reguladoras de Genes , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Integrinas/deficiência , Integrinas/genética , Rim/irrigação sanguínea , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Fosfoproteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA