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










Base de dados
Intervalo de ano de publicação
1.
Dev Dyn ; 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38546215

RESUMO

BACKGROUND: Neuronal lamination is a hallmark of the mammalian central nervous system (CNS) and underlies connectivity and function. Initial formation of this tissue architecture involves the integration of various signaling pathways that regulate the differentiation and migration of neural progenitor cells. RESULTS: Here, we demonstrate that mTORC1 mediates critical roles during neuronal lamination using the mouse retina as a model system. Down-regulation of mTORC1-signaling in retinal progenitor cells by conditional deletion of Rptor led to decreases in proliferation and increased apoptosis during embryogenesis. These developmental deficits preceded aberrant lamination in adult animals which was best exemplified by the fusion of the outer and inner nuclear layer and the absence of an outer plexiform layer. Moreover, ganglion cell axons originating from each Rptor-ablated retina appeared to segregate to an equal degree at the optic chiasm with both contralateral and ipsilateral projections displaying overlapping termination topographies within several retinorecipient nuclei. In combination, these visual pathway defects led to visually mediated behavioral deficits. CONCLUSIONS: This study establishes a critical role for mTORC1-signaling during retinal lamination and demonstrates that this pathway regulates diverse developmental mechanisms involved in driving the stratified arrangement of neurons during CNS development.

2.
Nat Commun ; 14(1): 6446, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37833281

RESUMO

Chromatin organization controls transcription by modulating 3D-interactions between enhancers and promoters in the nucleus. Alterations in epigenetic states and 3D-chromatin organization result in gene expression changes contributing to cancer. Here, we map the promoter-enhancer interactome and regulatory landscape of glioblastoma, the most aggressive primary brain tumour. Our data reveals profound rewiring of promoter-enhancer interactions, chromatin accessibility and redistribution of histone marks in glioblastoma. This leads to loss of long-range regulatory interactions and overall activation of promoters, which orchestrate changes in the expression of genes associated to glutamatergic synapses, axon guidance, axonogenesis and chromatin remodelling. SMAD3 and PITX1 emerge as major transcription factors controlling genes related to synapse organization and axon guidance. Inhibition of SMAD3 and neuronal activity stimulation cooperate to promote proliferation of glioblastoma cells in co-culture with glutamatergic neurons, and in mice bearing patient-derived xenografts. Our findings provide mechanistic insight into the regulatory networks that mediate neurogliomal synaptic communication.


Assuntos
Glioblastoma , Humanos , Animais , Camundongos , Glioblastoma/genética , Elementos Facilitadores Genéticos/genética , Cromatina , Fatores de Transcrição/genética , Expressão Gênica
3.
Dev Dyn ; 251(2): 321-335, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34148274

RESUMO

BACKGROUND: The coordinated wiring of neurons, glia and endothelial cells into neurovascular units is critical for central nervous system development. This is best exemplified in the mammalian retina where interneurons, astrocytes and retinal ganglion cells sculpt their vascular environment to meet the metabolic demands of visual function. Identifying the molecular networks that underlie neurovascular unit formation is an important step towards a deeper understanding of nervous system development and function. RESULTS: Here, we report that cell-to-cell mTORC1-signaling is essential for neurovascular unit formation during mouse retinal development. Using a conditional knockout approach we demonstrate that reduced mTORC1 activity in asymmetrically positioned retinal ganglion cells induces a delay in postnatal vascular network formation in addition to the production of rudimentary and tortuous vessel networks in adult animals. The severity of this vascular phenotype is directly correlated to the degree of mTORC1 down regulation within the neighboring retinal ganglion cell population. CONCLUSIONS: This study establishes a cell nonautonomous role for mTORC1-signaling during retinal development. These findings contribute to our current understanding of neurovascular unit formation and demonstrate how ganglion cells actively sculpt their local environment to ensure that the retina is perfused with an appropriate supply of oxygen and nutrients.


Assuntos
Doenças Retinianas , Células Ganglionares da Retina , Animais , Células Endoteliais/metabolismo , Mamíferos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Retina/metabolismo , Células Ganglionares da Retina/metabolismo
4.
Biol Open ; 8(8)2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31285269

RESUMO

Development of the vertebrate central nervous system involves the co-ordinated differentiation of progenitor cells and the establishment of functional neural networks. This neurogenic process is driven by both intracellular and extracellular cues that converge on the mammalian target of rapamycin complex 1 (mTORC1). Here we demonstrate that mTORC1-signalling mediates multi-faceted roles during central nervous system development using the mouse retina as a model system. Downregulation of mTORC1-signalling in retinal progenitor cells by conditional ablation of Rptor leads to proliferation deficits and an over-production of retinal ganglion cells during embryonic development. In contrast, reduced mTORC1-signalling in postnatal animals leads to temporal deviations in programmed cell death and the consequent production of asymmetric retinal ganglion cell mosaics and associated loss of axonal termination topographies in the dorsal lateral geniculate nucleus of adult mice. In combination these developmental defects induce visually mediated behavioural deficits. These collective observations demonstrate that mTORC1-signalling mediates critical roles during visual pathway development and function.

5.
Dis Model Mech ; 10(3): 245-257, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28250050

RESUMO

Development of the cornea, lens, ciliary body and iris within the anterior segment of the eye involves coordinated interaction between cells originating from the ciliary margin of the optic cup, the overlying periocular mesenchyme and the lens epithelium. Anterior segment dysgenesis (ASD) encompasses a spectrum of developmental syndromes that affect these anterior segment tissues. ASD conditions arise as a result of dominantly inherited genetic mutations and result in both ocular-specific and systemic forms of dysgenesis that are best exemplified by aniridia and Axenfeld-Rieger syndrome, respectively. Extensive clinical overlap in disease presentation amongst ASD syndromes creates challenges for correct diagnosis and classification. The use of animal models has therefore proved to be a robust approach for unravelling this complex genotypic and phenotypic heterogeneity. However, despite these successes, it is clear that additional genes that underlie several ASD syndromes remain unidentified. Here, we report the characterisation of a novel mouse model of ASD. Conditional deletion of Tsc1 during eye development leads to a premature upregulation of mTORC1 activity within the ciliary margin, periocular mesenchyme and lens epithelium. This aberrant mTORC1 signalling within the ciliary margin in particular leads to a reduction in the number of cells that express Pax6, Bmp4 and Msx1 Sustained mTORC1 signalling also induces a decrease in ciliary margin progenitor cell proliferation and a consequent failure of ciliary body and iris development in postnatal animals. Our study therefore identifies Tsc1 as a novel candidate ASD gene. Furthermore, the Tsc1-ablated mouse model also provides a valuable resource for future studies concerning the molecular mechanisms underlying ASD and acts as a platform for evaluating therapeutic approaches for the treatment of visual disorders.


Assuntos
Corpo Ciliar/embriologia , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Deleção de Genes , Iris/embriologia , Proteínas Supressoras de Tumor/genética , Animais , Animais Recém-Nascidos , Segmento Anterior do Olho/embriologia , Segmento Anterior do Olho/patologia , Linhagem da Célula , Corpo Ciliar/patologia , Modelos Animais de Doenças , Integrases/metabolismo , Iris/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos Transgênicos , Especificidade de Órgãos , Fenótipo , Transdução de Sinais , Transcrição Gênica , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
6.
Dis Model Mech ; 8(12): 1517-29, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26449264

RESUMO

Tuberous sclerosis complex (TSC) is an autosomal dominant syndrome that is best characterised by neurodevelopmental deficits and the presence of benign tumours (called hamartomas) in affected organs. This multi-organ disorder results from inactivating point mutations in either the TSC1 or the TSC2 genes and consequent activation of the canonical mammalian target of rapamycin complex 1 signalling (mTORC1) pathway. Because lesions to the eye are central to TSC diagnosis, we report here the generation and characterisation of the first eye-specific TSC mouse model. We demonstrate that conditional ablation of Tsc1 in eye-committed progenitor cells leads to the accelerated differentiation and subsequent ectopic radial migration of retinal ganglion cells. This results in an increase in retinal ganglion cell apoptosis and consequent regionalised axonal loss within the optic nerve and topographical changes to the contra- and ipsilateral input within the dorsal lateral geniculate nucleus. Eyes from adult mice exhibit aberrant retinal architecture and display all the classic neuropathological hallmarks of TSC, including an increase in organ and cell size, ring heterotopias, hamartomas with retinal detachment, and lamination defects. Our results provide the first major insight into the molecular etiology of TSC within the developing eye and demonstrate a pivotal role for Tsc1 in regulating various aspects of visual-pathway development. Our novel mouse model therefore provides a valuable resource for future studies concerning the molecular mechanisms underlying TSC and also as a platform to evaluate new therapeutic approaches for the treatment of this multi-organ disorder.


Assuntos
Olho/patologia , Deleção de Genes , Esclerose Tuberosa/patologia , Proteínas Supressoras de Tumor/metabolismo , Vias Visuais/crescimento & desenvolvimento , Vias Visuais/metabolismo , Animais , Contagem de Células , Diferenciação Celular , Movimento Celular , Núcleo Celular/patologia , Modelos Animais de Doenças , Gliose/patologia , Hamartoma/patologia , Interneurônios/metabolismo , Camundongos Knockout , Mosaicismo , Neuritos/metabolismo , Nervo Óptico/patologia , Especificidade de Órgãos , Células Ganglionares da Retina/patologia , Proteína 1 do Complexo Esclerose Tuberosa , Vias Visuais/patologia
7.
PLoS One ; 8(12): e81158, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24324671

RESUMO

A multitude of signalling pathways are involved in the process of forming an eye. Here we demonstrate that ß-catenin is essential for eye development as inactivation of ß-catenin prior to cellular specification in the optic vesicle caused anophthalmia in mice. By achieving this early and tissue-specific ß-catenin inactivation we find that retinal pigment epithelium (RPE) commitment was blocked and eye development was arrested prior to optic cup formation due to a loss of canonical Wnt signalling in the dorsal optic vesicle. Thus, these results show that Wnt/ß-catenin signalling is required earlier and play a more central role in eye development than previous studies have indicated. In our genetic model system a few RPE cells could escape ß-catenin inactivation leading to the formation of a small optic rudiment. The optic rudiment contained several neural retinal cell classes surrounded by an RPE. Unlike the RPE cells, the neural retinal cells could be ß-catenin-negative revealing that differentiation of the neural retinal cell classes is ß-catenin-independent. Moreover, although dorsoventral patterning is initiated in the mutant optic vesicle, the neural retinal cells in the optic rudiment displayed almost exclusively ventral identity. Thus, ß-catenin is required for optic cup formation, commitment to RPE cells and maintenance of dorsal identity of the retina.


Assuntos
Olho/embriologia , Olho/metabolismo , Via de Sinalização Wnt , Actinas/metabolismo , Animais , Anoftalmia/embriologia , Anoftalmia/metabolismo , Anoftalmia/patologia , Padronização Corporal , Caderinas/metabolismo , Linhagem da Célula , Polaridade Celular , Proliferação de Células , Embrião de Mamíferos/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Camundongos , Mutação/genética , Especificidade de Órgãos , Epitélio Pigmentado da Retina/embriologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo
8.
FASEB J ; 26(8): 3464-72, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22581782

RESUMO

Inactivation of the LIM-homeodomain 2 gene (Lhx2) results in a severe defect in specification of olfactory sensory neurons (OSNs). However, the ramifications of lack of Lhx2-dependent OSN specification for formation of the primary olfactory pathway have not been addressed, since mutant mice die in utero. We have analyzed prenatal and postnatal consequences of conditionally inactivating Lhx2 selectively in OSNs. A cell-autonomous effect is that OSN axons cannot innervate their target, the olfactory bulb. Moreover, the lack of Lhx2 in OSNs causes unpredicted, non-cell-autonomous phenotypes. First, the olfactory bulb shows pronounced hypoplasia in adults, and the data suggest that innervation by correctly specified OSNs is necessary for adult bulb size and organization. Second, absence of an olfactory nerve in the conditional mutant reveals that the vomeronasal nerve is dependent on olfactory nerve formation. Third, the lack of a proper vomeronasal nerve prevents migration of gonadotropin-releasing hormone (GnRH) cells the whole distance to their final positions in the hypothalamus during embryo development. As adults, the conditional mutants do not pass puberty, and these findings support the view of an exclusive nasal origin of GnRH neurons in the mouse. Thus, Lhx2 in OSNs is required for functional development of three separate systems.


Assuntos
Proteínas com Homeodomínio LIM/fisiologia , Neurônios Receptores Olfatórios/fisiologia , Células Receptoras Sensoriais/fisiologia , Fatores de Transcrição/fisiologia , Animais , Movimento Celular/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Masculino , Camundongos , Bulbo Olfatório/embriologia , Condutos Olfatórios/embriologia , Neurônios Receptores Olfatórios/embriologia , Órgão Vomeronasal/embriologia
9.
PLoS One ; 6(8): e23387, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21886788

RESUMO

Progenitor cells committed to eye development become specified in the prospective forebrain and develop subsequently into the optic vesicle and the optic cup. The optic vesicle induces formation of the lens placode in surface ectoderm from which the lens develops. Numerous transcription factors are involved in this process, including the eye-field transcription factors. However, many of these transcription factors also regulate the patterning of the anterior neural plate and their specific role in eye development is difficult to discern since eye-committed progenitor cells are poorly defined. By using a specific part of the Lhx2 promoter to regulate Cre recombinase expression in transgenic mice we have been able to define a distinct progenitor cell population in the forebrain solely committed to eye development. Conditional inactivation of Lhx2 in these progenitor cells causes an arrest in eye development at the stage when the optic vesicle induces lens placode formation in the surface ectoderm. The eye-committed progenitor cell population is present in the Lhx2(-/-) embryonic forebrain suggesting that commitment to eye development is Lhx2-independent. However, re-expression of Lhx2 in Lhx2(-/-) progenitor cells only promotes development of retinal pigment epithelium cells, indicating that Lhx2 promotes the acquisition of the oligopotent fate of these progenitor cells. This approach also allowed us to identify genes that distinguish Lhx2 function in eye development from that in the forebrain. Thus, we have defined a distinct progenitor cell population in the forebrain committed to eye development and identified genes linked to Lhx2's function in the expansion and patterning of these progenitor cells.


Assuntos
Padronização Corporal , Linhagem da Célula , Olho/citologia , Olho/embriologia , Proteínas com Homeodomínio LIM/metabolismo , Células-Tronco/citologia , Fatores de Transcrição/metabolismo , Animais , Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 7/metabolismo , Proliferação de Células , Embrião de Mamíferos/metabolismo , Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Integrases/metabolismo , Proteínas com Homeodomínio LIM/genética , Cristalino/citologia , Cristalino/embriologia , Cristalino/metabolismo , Camundongos , Camundongos Transgênicos , Placa Neural/citologia , Placa Neural/embriologia , Especificidade de Órgãos/genética , Células-Tronco/metabolismo , Fatores de Transcrição/genética
10.
PLoS Genet ; 6(4): e1000904, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20386748

RESUMO

Hair is important for thermoregulation, physical protection, sensory activity, seasonal camouflage, and social interactions. Hair is generated in hair follicles (HFs) and, following morphogenesis, HFs undergo cyclic phases of active growth (anagen), regression (catagen), and inactivity (telogen) throughout life. The transcriptional regulation of this process is not well understood. We show that the transcription factor Lhx2 is expressed in cells of the outer root sheath and a subpopulation of matrix cells during both morphogenesis and anagen. As the HFs enter telogen, expression becomes undetectable and reappears prior to initiation of anagen in the secondary hair germ. In contrast to previously published results, we find that Lhx2 is primarily expressed by precursor cells outside of the bulge region where the HF stem cells are located. This developmental, stage- and cell-specific expression suggests that Lhx2 regulates the generation and regeneration of hair. In support of this hypothesis, we show that Lhx2 is required for anagen progression and HF morphogenesis. Moreover, transgenic expression of Lhx2 in postnatal HFs is sufficient to induce anagen. Thus, our results reveal an alternative interpretation of Lhx2 function in HFs compared to previously published results, since Lhx2 is periodically expressed, primarily in precursor cells distinct from those in the bulge region, and is an essential positive regulator of hair formation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Cabelo/crescimento & desenvolvimento , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Animais , Proliferação de Células , Cabelo/embriologia , Folículo Piloso/crescimento & desenvolvimento , Proteínas de Homeodomínio/metabolismo , Proteínas com Homeodomínio LIM , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfogênese , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
11.
PLoS One ; 3(4): e2025, 2008 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-18431502

RESUMO

The molecular mechanisms regulating the expansion of the hematopoietic system including hematopoietic stem cells (HSCs) in the fetal liver during embryonic development are largely unknown. The LIM-homeobox gene Lhx2 is a candidate regulator of fetal hematopoiesis since it is expressed in the fetal liver and Lhx2(-/-) mice die in utero due to severe anemia. Moreover, expression of Lhx2 in embryonic stem (ES) cell-derived embryoid bodies (EBs) can lead to the generation of HSC-like cell lines. To further define the role of this transcription factor in hematopoietic regulation, we generated ES cell lines that enabled tet-inducible expression of Lhx2. Using this approach we observed that Lhx2 expression synergises with specific signalling pathways, resulting in increased frequency of colony forming cells in developing EB cells. The increase in growth factor-responsive progenitor cells directly correlates to the efficiency in generating HSC-like cell lines, suggesting that Lhx2 expression induce self-renewal of a distinct multipotential hematopoietic progenitor cell in EBs. Signalling via the c-kit tyrosine kinase receptor and the gp130 signal transducer by IL-6 is necessary and sufficient for the Lhx2 induced self-renewal. While inducing self-renewal of multipotential progenitor cells, expression of Lhx2 inhibited proliferation of primitive erythroid precursor cells and interfered with early ES cell commitment, indicating striking lineage specificity of this effect.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Multipotentes/citologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Clonais , Ensaio de Unidades Formadoras de Colônias , Doxiciclina/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Interleucina-6/farmacologia , Proteínas com Homeodomínio LIM , Camundongos , Modelos Biológicos , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Multipotentes/metabolismo , Fator de Células-Tronco/farmacologia
12.
Dev Biol ; 295(2): 615-22, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16712832

RESUMO

Many studies have suggested the hypothesis that the plasminogen activator (PA) system and the matrix metalloproteinase (MMP) system, either separately or in combination, may provide the proteolytic activity that is required for rupture of the follicular wall at the time of ovulation. Our recent studies on ovulation in plasminogen (plg)-deficient mice have, however, shown that plasmin is not required for normal ovulation, leading us to the hypothesis that MMPs may be a more important source of proteolysis for this process. To investigate the role of MMPs and also the possibility of a functional overlap or synergy between the MMP and PA systems during ovulation, we have studied ovulation efficiency in wild-type and plg-deficient mice treated with the broad-spectrum MMP inhibitor galardin. We found that in both wild-type mice and heterozygous plg-deficient (plg+/-) mice that had been treated with galardin prior to ovulation, there was a mild (18-20%) reduction in ovulation efficiency. Surprisingly, galardin treatment of plg-deficient (plg-/-) mice only caused an additional 14% reduction in ovulation efficiency as compared to vehicle-treated plg-/- mice. Our data therefore suggest that although MMPs may play a role in degradation of the follicular wall, they may not be obligatory for ovulation. In contrast to previous studies on tissue remodeling during wound healing and placental development, we have demonstrated that there is no obvious functional overlap or synergy between the PA and MMP systems, which has previously been thought to be essential for the ovulatory process.


Assuntos
Dipeptídeos/farmacologia , Metaloproteinases da Matriz/fisiologia , Ovulação/efeitos dos fármacos , Plasminogênio/deficiência , Animais , Dipeptídeos/uso terapêutico , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Feminino , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Knockout , Folículo Ovariano/enzimologia , Folículo Ovariano/metabolismo , Folículo Ovariano/ultraestrutura , Ativadores de Plasminogênio/fisiologia
13.
Haematologica ; 88(12): 1336-47, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14687986

RESUMO

BACKGROUND AND OBJECTIVES: Chronic myeloproliferative disorders (CMDs) are thought to be due to mutation(s) in a single clone at the level of the hematopoietic stem cell (HSC). Such mutations and additional mutations causing progression of the disease are largely unknown. Chronic myeloid leukemia (CML) is a CMD characterized by a chromosomal translocation between chromosomes 9 and 22 creating the fusion protein BCR-ABL. This translocation has also been suggested to cause mis-expression of the LIM-homeobox gene Lhx2 in hematopoietic cells. We have previously shown that Lhx2 expression in mouse HSC generates cytokine-dependent stem cell-like cell lines that can produce long-term repopulation in stem cell-deficient mice. DESIGN AND METHODS: Since the consequences of Lhx2 expression in hematopoietic cells in vivo were unknown, mice engrafted with the stem cell-like cell lines were analyzed in detail for any pathologic changes. RESULTS: Expression of Lhx2 was maintained in vivo and most engrafted mice developed a myeloproliferative disorder characterized by splenomegaly, extramedullary hematopoiesis and anemia. The disorder was transplantable and the Lhx2-expressing cells could also cause acute leukemia. The anemia was due to both a reduced number of circulating erythrocytes and a reduced mean corpuscular hemoglobin concentration (MCHC). INTERPRETATION AND CONCLUSIONS: These observations suggest that constitutive expression of Lhx2 in hematopoietic cells causes CMD, and also that a novel cell-autonomous mechanism can contribute to anemia.


Assuntos
Globinas/biossíntese , Células-Tronco Hematopoéticas/patologia , Proteínas de Homeodomínio/fisiologia , Transtornos Mieloproliferativos/genética , Fatores de Transcrição/fisiologia , Anemia/genética , Animais , Medula Óssea/patologia , Doença Crônica , Ensaio de Unidades Formadoras de Colônias , Progressão da Doença , Contagem de Eritrócitos , Índices de Eritrócitos , Regulação Leucêmica da Expressão Gênica , Globinas/genética , Glucose-6-Fosfato Isomerase/genética , Hematopoese Extramedular/genética , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Proteínas de Homeodomínio/genética , Proteínas com Homeodomínio LIM , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Leucemia Mieloide/genética , Leucemia Mieloide/patologia , Contagem de Leucócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/patologia , Transtornos Mieloproliferativos/patologia , Proteínas Proto-Oncogênicas c-kit/genética , Provírus/genética , Quimera por Radiação , Baço/patologia , Esplenomegalia/etiologia , Fatores de Transcrição/genética , Transdução Genética
14.
Mol Cell Endocrinol ; 205(1-2): 131-40, 2003 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-12890575

RESUMO

The formation of the corpus luteum (CL) is accompanied with angiogenesis and tissue remodeling and its regression involves tissue degradation. Matrix degrading proteases such as plasminogen activators (PAs) and matrix metalloproteinases (MMPs) are thought to play important roles in such controlled proteolytic processes. In this study, in situ hybridization has been used to examine the regulation and expression pattern of mRNAs coding for proteases and protease inhibitors belonging to the PA- and MMP-systems during the life cycle of the CL in an adult pseudopregnant mouse model. Of the nine proteases and five protease inhibitors that were studied, the majority were found to be temporally expressed during the formation and/or the regression of the CL. However, the mRNAs coding for urokinase type PA (uPA), membrane-type 1 MMP (MT1-MMP), and tissue inhibitor of metalloproteinases type-3 (TIMP-3) were constantly expressed in the mouse CL throughout its whole life span. To study the functional role of uPA in the CL, we analyzed luteal formation and function in uPA deficient mice. Our results revealed no significant difference in ovarian weight, serum progesterone levels, and blood vessel density in the functional CL between uPA deficient and wild type control mice. The temporal and spatial expression pattern of proteases and protease inhibitors during the CL life span suggests that members of the PA- and MMP-systems may play important roles in the angiogenesis and tissue remodeling processes during CL formation, as well as in the tissue degradation during luteal regression. However, the absence of reproductive phenotypes in mice lacking uPA and several other matrix degrading proteases indicates that there are redundancies among different matrix degrading proteases or that tissue remodeling in the ovary may involve other additional unique elements.


Assuntos
Corpo Lúteo/enzimologia , Endopeptidases/metabolismo , Matriz Extracelular/enzimologia , Inibidores de Proteases/metabolismo , Animais , Corpo Lúteo/crescimento & desenvolvimento , Corpo Lúteo/metabolismo , Endopeptidases/fisiologia , Feminino , Perfilação da Expressão Gênica , Lactação , Masculino , Camundongos , Camundongos Knockout/genética , Gravidez , Progesterona/sangue , RNA Mensageiro/metabolismo , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...