Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 150(6)2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36960827

RESUMO

The blood-brain barrier (BBB) is a vascular endothelial cell boundary that partitions the circulation from the central nervous system to promote normal brain health. We have a limited understanding of how the BBB is formed during development and maintained in adulthood. We used quantitative transcriptional profiling to investigate whether specific adhesion molecules are involved in BBB functions, with an emphasis on understanding how astrocytes interact with endothelial cells. Our results reveal a striking enrichment of multiple genes encoding laminin subunits as well as the laminin receptor gene Itga7, which encodes the alpha7 integrin subunit, in astrocytes. Genetic ablation of Itga7 in mice led to aberrant BBB permeability and progressive neurological pathologies. Itga7-/- mice also showed a reduction in laminin protein expression in parenchymal basement membranes. Blood vessels in the Itga7-/- brain showed separation from surrounding astrocytes and had reduced expression of the tight junction proteins claudin 5 and ZO-1. We propose that the alpha7 integrin subunit in astrocytes via adhesion to laminins promotes endothelial cell junction integrity, all of which is required to properly form and maintain a functional BBB.


Assuntos
Astrócitos , Barreira Hematoencefálica , Camundongos , Animais , Barreira Hematoencefálica/metabolismo , Laminina/metabolismo , Células Endoteliais/metabolismo , Integrinas/metabolismo , Junções Íntimas/metabolismo
2.
Development ; 149(6)2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35217866

RESUMO

In the developing mammalian brain, neuroepithelial cells interact with blood vessels to regulate angiogenesis, blood-brain barrier maturation and other key neurovascular functions. Genetic studies in mice have shown that neurovascular development is controlled, in part, by Itgb8, which encodes the neuroepithelial cell-expressed integrin ß8 subunit. However, these studies have involved complete loss-of-function Itgb8 mutations, and have not discerned the relative roles for the ß8 integrin extracellular matrix (ECM) binding region versus the intracellular signaling tail. Here, Cre/lox strategies have been employed to selectively delete the cytoplasmic tail of murine Itgb8 without perturbing its transmembrane and extracellular domains. We report that the ß8 integrin cytoplasmic domain is essential for inside-out modulation of adhesion, including activation of latent-TGFßs in the ECM. Quantitative sequencing of the brain endothelial cell transcriptome identifies TGFß-regulated genes with putative links to blood vessel morphogenesis, including several genes linked to Wnt/ß-catenin signaling. These results reveal that the ß8 integrin cytoplasmic domain is essential for the regulation of TGFß-dependent gene expression in endothelial cells and suggest that cross-talk between TGFßs and Wnt pathways is crucial for neurovascular development.


Assuntos
Células Endoteliais , Cadeias beta de Integrinas , Animais , Encéfalo/metabolismo , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Cadeias beta de Integrinas/genética , Cadeias beta de Integrinas/metabolismo , Integrinas/genética , Integrinas/metabolismo , Mamíferos/metabolismo , Camundongos , Fator de Crescimento Transformador beta/metabolismo
3.
J Neurosci ; 43(47): 8043-8057, 2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-37722850

RESUMO

The malignant brain cancer glioblastoma (GBM) contains groups of highly invasive cells that drive tumor progression as well as recurrence after surgery and chemotherapy. The molecular mechanisms that enable these GBM cells to exit the primary mass and disperse throughout the brain remain largely unknown. Here we report using human tumor specimens and primary spheroids from male and female patients that glial cell adhesion molecule (GlialCAM), which has normal roles in brain astrocytes and is mutated in the developmental brain disorder megalencephalic leukoencephalopathy with subcortical cysts (MLC), is differentially expressed in subpopulations of GBM cells. High levels of GlialCAM promote cell-cell adhesion and a proliferative GBM cell state in the tumor core. In contrast, GBM cells with low levels of GlialCAM display diminished proliferation and enhanced invasion into the surrounding brain parenchyma. RNAi-mediated inhibition of GlialCAM expression leads to activation of proinvasive extracellular matrix adhesion and signaling pathways. Profiling GlialCAM-regulated genes combined with cross-referencing to single-cell transcriptomic datasets validates functional links among GlialCAM, Mlc1, and aquaporin-4 in the invasive cell state. Collectively, these results reveal an important adhesion and signaling axis comprised of GlialCAM and associated proteins including Mlc1 and aquaporin-4 that is critical for control of GBM cell proliferation and invasion status in the brain cancer microenvironment.SIGNIFICANCE STATEMENT Glioblastoma (GBM) contains heterogeneous populations of cells that coordinately drive proliferation and invasion. We have discovered that glial cell adhesion molecule (GlialCAM)/hepatocyte cell adhesion molecule (HepaCAM) is highly expressed in proliferative GBM cells within the tumor core. In contrast, GBM cells with low levels of GlialCAM robustly invade into surrounding brain tissue along blood vessels and white matter. Quantitative RNA sequencing identifies various GlialCAM-regulated genes with functions in cell-cell adhesion and signaling. These data reveal that GlialCAM and associated signaling partners, including Mlc1 and aquaporin-4, are key factors that determine proliferative and invasive cell states in GBM.


Assuntos
Aquaporinas , Glioblastoma , Feminino , Humanos , Masculino , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Proteínas de Ciclo Celular/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patologia , Proteínas de Membrana/metabolismo , Microambiente Tumoral , Proliferação de Células , Invasividade Neoplásica
4.
J Neurosci ; 42(8): 1406-1416, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34965971

RESUMO

In the mammalian brain, perivascular astrocytes (PAs) closely juxtapose blood vessels and are postulated to have important roles in the control of vascular physiology, including regulation of the blood-brain barrier (BBB). Deciphering specific functions for PAs in BBB biology, however, has been limited by the ability to distinguish these cells from other astrocyte populations. In order to characterize selective roles for PAs in vivo, a new mouse model has been generated in which the endogenous megalencephalic leukoencephalopathy with subcortical cysts 1 (Mlc1) gene drives expression of Cre fused to a mutated estrogen ligand-binding domain (Mlc1-T2A-CreERT2). This knock-in mouse model, which we term MLCT, allows for selective identification and tracking of PAs in the postnatal brain. We also demonstrate that MLCT-mediated ablation of PAs causes severe defects in BBB integrity, resulting in premature death. PA loss results in aberrant localization of Claudin 5 and -VE-Cadherin in endothelial cell junctions as well as robust microgliosis. Collectively, these data reveal essential functions for Mlc1-expressing PAs in regulating endothelial barrier integrity in mice and indicate that primary defects in astrocytes that cause BBB breakdown may contribute to human neurologic disorders.SIGNIFICANCE STATEMENT Interlaced among the billions of neurons and glia in the mammalian brain is an elaborate network of blood vessels. Signals from the brain parenchyma control the unique permeability properties of cerebral blood vessels known as the blood-brain barrier (BBB). However, we understand very little about the relative contributions of different neural cell types in the regulation of BBB functions. Here, we show that a specific subpopulation of astrocyte is essential for control of BBB integrity, with ablation of these cells leading to defects in endothelial cell junctions, BBB breakdown, and resulting neurologic deficits.


Assuntos
Astrócitos , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central , Animais , Astrócitos/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Claudina-5/genética , Cistos , Modelos Animais de Doenças , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/genética , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/metabolismo , Mamíferos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos
5.
Development ; 147(18)2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32895288

RESUMO

The central nervous system (CNS) contains a complex network of blood vessels that promote normal tissue development and physiology. Abnormal control of blood vessel morphogenesis and maturation is linked to the pathogenesis of various neurodevelopmental diseases. The CNS-specific genes that regulate blood vessel morphogenesis in development and disease remain largely unknown. Here, we have characterized functions for the gene encoding prion protein 2 (Prnd) in CNS blood vessel development and physiology. Prnd encodes the glycosylphosphatidylinositol (GPI)-linked protein doppel, which is expressed on the surface of angiogenic vascular endothelial cells, but is absent in quiescent endothelial cells of the adult CNS. During CNS vascular development, doppel interacts with receptor tyrosine kinases and activates cytoplasmic signaling pathways involved in endothelial cell survival, metabolism and migration. Analysis of mice genetically null for Prnd revealed impaired CNS blood vessel morphogenesis and associated endothelial cell sprouting defects. Prnd-/- mice also displayed defects in endothelial barrier integrity. Collectively, these data reveal novel mechanisms underlying doppel control of angiogenesis in the developing CNS, and may provide new insights about dysfunctional pathways that cause vascular-related CNS disorders.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica/fisiologia , Proteínas Priônicas/metabolismo , Animais , Sistema Nervoso Central/metabolismo , Citoplasma/metabolismo , Proteínas Ligadas por GPI/metabolismo , Camundongos , Morfogênese/fisiologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia
6.
J Cell Sci ; 133(12)2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32540905

RESUMO

Cells must interpret a complex milieu of extracellular cues to modulate intracellular signaling events linked to proliferation, differentiation, migration and other cellular processes. Integrins are heterodimeric transmembrane proteins that link the extracellular matrix (ECM) to the cytoskeleton and control intracellular signaling events. A great deal is known about the structural and functional properties for most integrins; however, the adhesion and signaling pathways controlled by αvß8 integrin, which was discovered nearly 30 years ago, have only recently been characterized. αvß8 integrin is a receptor for ECM-bound forms of latent transforming growth factor ß (TGFß) proteins and promotes the activation of TGFß signaling pathways. Studies of the brain, lung and immune system reveal that the αvß8 integrin-TGFß axis mediates cell-cell contact and communication within complex multicellular structures. Perturbing components of this axis results in aberrant cell-cell adhesion and signaling leading to the initiation of various pathologies, including neurodegeneration, fibrosis and cancer. As discussed in this Review, understanding the functions for αvß8 integrin, its ECM ligands and intracellular effector proteins is not only an important topic in cell biology, but may lead to new therapeutic strategies to treat human pathologies related to integrin dysfunction.


Assuntos
Integrinas , Transdução de Sinais , Adesão Celular , Humanos , Integrinas/genética , Fator de Crescimento Transformador beta
7.
Glia ; 69(1): 91-108, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32744761

RESUMO

In the developing peripheral nervous system, Schwann cells (SCs) extend their processes to contact, sort, and myelinate axons. The mechanisms that contribute to the interaction between SCs and axons are just beginning to be elucidated. Using a SC-neuron coculture system, we demonstrate that Arg-Gly-Asp (RGD) peptides that inhibit αV -containing integrins delay the extension of SCs elongating on axons. αV integrins in SC localize to sites of contact with axons and are expressed early in development during radial sorting and myelination. Short interfering RNA-mediated knockdown of the αV integrin subunit also delays SC extension along axons in vitro, suggesting that αV -containing integrins participate in axo-glial interactions. However, mice lacking the αV subunit in SCs, alone or in combination with the potentially compensating α5 subunit, or the αV partners ß3 or ß8 , myelinate normally during development and remyelinate normally after nerve crush, indicating that overlapping or compensatory mechanisms may hide the in vivo role of RGD-binding integrins.


Assuntos
Células de Schwann , Animais , Axônios , Integrina alfaV , Integrinas , Camundongos , Oligopeptídeos
8.
Nature ; 527(7576): 100-104, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26479035

RESUMO

The development of life-threatening cancer metastases at distant organs requires disseminated tumour cells' adaptation to, and co-evolution with, the drastically different microenvironments of metastatic sites. Cancer cells of common origin manifest distinct gene expression patterns after metastasizing to different organs. Clearly, the dynamic interaction between metastatic tumour cells and extrinsic signals at individual metastatic organ sites critically effects the subsequent metastatic outgrowth. Yet, it is unclear when and how disseminated tumour cells acquire the essential traits from the microenvironment of metastatic organs that prime their subsequent outgrowth. Here we show that both human and mouse tumour cells with normal expression of PTEN, an important tumour suppressor, lose PTEN expression after dissemination to the brain, but not to other organs. The PTEN level in PTEN-loss brain metastatic tumour cells is restored after leaving the brain microenvironment. This brain microenvironment-dependent, reversible PTEN messenger RNA and protein downregulation is epigenetically regulated by microRNAs from brain astrocytes. Mechanistically, astrocyte-derived exosomes mediate an intercellular transfer of PTEN-targeting microRNAs to metastatic tumour cells, while astrocyte-specific depletion of PTEN-targeting microRNAs or blockade of astrocyte exosome secretion rescues the PTEN loss and suppresses brain metastasis in vivo. Furthermore, this adaptive PTEN loss in brain metastatic tumour cells leads to an increased secretion of the chemokine CCL2, which recruits IBA1-expressing myeloid cells that reciprocally enhance the outgrowth of brain metastatic tumour cells via enhanced proliferation and reduced apoptosis. Our findings demonstrate a remarkable plasticity of PTEN expression in metastatic tumour cells in response to different organ microenvironments, underpinning an essential role of co-evolution between the metastatic cells and their microenvironment during the adaptive metastatic outgrowth. Our findings signify the dynamic and reciprocal cross-talk between tumour cells and the metastatic niche; importantly, they provide new opportunities for effective anti-metastasis therapies, especially of consequence for brain metastasis patients.


Assuntos
Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Exossomos/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , MicroRNAs/genética , PTEN Fosfo-Hidrolase/deficiência , Microambiente Tumoral , Adaptação Fisiológica/genética , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/metabolismo , Proteínas de Ligação ao Cálcio , Proliferação de Células/genética , Quimiocina CCL2/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/genética , Evolução Molecular , Exossomos/metabolismo , Feminino , Genes Supressores de Tumor , Humanos , Masculino , Camundongos , Proteínas dos Microfilamentos , PTEN Fosfo-Hidrolase/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Microambiente Tumoral/genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
9.
Development ; 142(24): 4363-73, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26586223

RESUMO

Angiogenesis in the developing central nervous system (CNS) is regulated by neuroepithelial cells, although the genes and pathways that couple these cells to blood vessels remain largely uncharacterized. Here, we have used biochemical, cell biological and molecular genetic approaches to demonstrate that ß8 integrin (Itgb8) and neuropilin 1 (Nrp1) cooperatively promote CNS angiogenesis by mediating adhesion and signaling events between neuroepithelial cells and vascular endothelial cells. ß8 integrin in the neuroepithelium promotes the activation of extracellular matrix (ECM)-bound latent transforming growth factor ß (TGFß) ligands and stimulates TGFß receptor signaling in endothelial cells. Nrp1 in endothelial cells suppresses TGFß activation and signaling by forming intercellular protein complexes with ß8 integrin. Cell type-specific ablation of ß8 integrin, Nrp1, or canonical TGFß receptors results in pathological angiogenesis caused by defective neuroepithelial cell-endothelial cell adhesion and imbalances in canonical TGFß signaling. Collectively, these data identify a paracrine signaling pathway that links the neuroepithelium to blood vessels and precisely balances TGFß signaling during cerebral angiogenesis.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Cadeias beta de Integrinas/metabolismo , Neovascularização Fisiológica , Neuropilina-1/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Actinas/metabolismo , Animais , Encéfalo/patologia , Adesão Celular , Perda do Embrião/patologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Deleção de Genes , Masculino , Camundongos , Modelos Biológicos , Células Neuroepiteliais/citologia , Células Neuroepiteliais/metabolismo , Peixe-Zebra
10.
Genesis ; 55(10)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28929580

RESUMO

Perivascular astrocyte end feet closely juxtapose cerebral blood vessels to regulate important developmental and physiological processes including endothelial cell proliferation and sprouting as well as the formation of the blood-brain barrier (BBB). The mechanisms underlying these events remain largely unknown due to a lack of experimental models for identifying perivascular astrocytes and distinguishing these cell types from other astroglial populations. Megalencephalic leukoencephalopathy with subcortical cysts 1 (Mlc1) is a transmembrane protein that is expressed in perivascular astrocyte end feet where it controls BBB development and homeostasis. On the basis of this knowledge, we used T2A peptide-skipping strategies to engineer a knock-in mouse model in which the endogenous Mlc1 gene drives expression of enhanced green fluorescent protein (eGFP), without impacting expression of Mlc1 protein. Analysis of fetal, neonatal and adult Mlc1-eGFP knock-in mice revealed a dynamic spatiotemporal expression pattern of eGFP in glial cells, including nestin-expressing neuroepithelial cells during development and glial fibrillary acidic protein (GFAP)-expressing perivascular astrocytes in the postnatal brain. EGFP was not expressed in neurons, microglia, oligodendroglia, or cerebral vascular cells. Analysis of angiogenesis in the neonatal retina also revealed enriched Mlc1-driven eGFP expression in perivascular astrocytes that contact sprouting blood vessels and regulate blood-retinal barrier permeability. A cortical injury model revealed that Mlc1-eGFP expression is progressively induced in reactive astrocytes that form a glial scar. Hence, Mlc1-eGFP knock-in mice are a new and powerful tool to identify perivascular astrocytes in the brain and retina and characterize how these cell types regulate cerebral blood vessel functions in health and disease.


Assuntos
Astrócitos/metabolismo , Técnicas de Introdução de Genes/métodos , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Membrana/metabolismo , Animais , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Biossíntese de Proteínas , Retina/citologia , Retina/embriologia , Retina/metabolismo , Ribossomos/metabolismo
11.
Genesis ; 55(8)2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28653435

RESUMO

Application of CRISPR-Cas9 technology in diverse organisms has resulted in an explosion of genome modification efforts. To expand the toolbox of applications, we have created an E. coli Exonuclease I (sbcB)-Cas9 fusion that has altered enzymatic activity in zebrafish embryos. This Cas9 variant has increased mutation efficiency and favors longer deletions relative to wild-type Cas9. We anticipate that this variant will allow for more efficient screening for F0 phenotypes and mutation of a larger spectrum of genomic targets including deletion of regulatory regions and creating loss of function mutations in transcription units with poor sequence conservation such as lncRNAs where larger deletions may be required for loss of function.


Assuntos
Proteínas de Bactérias/genética , Sistemas CRISPR-Cas , Endonucleases/genética , Marcação de Genes/métodos , Peixe-Zebra/genética , Animais , Proteínas de Bactérias/metabolismo , Proteína 9 Associada à CRISPR , Endonucleases/metabolismo , Deleção de Genes , Marcação de Genes/normas , Mutação com Perda de Função
12.
Development ; 138(23): 5157-66, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22069187

RESUMO

The mouse retina is vascularized after birth when angiogenic blood vessels grow and sprout along a pre-formed latticework of astrocytes. How astrocyte-derived cues control patterns of blood vessel growth and sprouting, however, remains enigmatic. Here, we have used molecular genetic strategies in mice to demonstrate that αvß8 integrin expressed in astrocytes is essential for neovascularization of the developing retina. Selective ablation of αv or ß8 integrin gene expression in astrocytes leads to impaired blood vessel sprouting and intraretinal hemorrhage, particularly during formation of the secondary vascular plexus. These pathologies correlate, in part, with diminished αvß8 integrin-mediated activation of extracellular matrix-bound latent transforming growth factor ßs (TGFßs) and defective TGFß signaling in vascular endothelial cells, but not astrocytes. Collectively, our data demonstrate that αvß8 integrin is a component of a paracrine signaling axis that links astrocytes to blood vessels and is essential for proper regulation of retinal angiogenesis.


Assuntos
Astrócitos/metabolismo , Integrinas/metabolismo , Comunicação Parácrina/fisiologia , Vasos Retinianos/embriologia , Animais , Primers do DNA/genética , Imunofluorescência , Genótipo , Immunoblotting , Integrinas/genética , Camundongos , Comunicação Parácrina/genética
13.
Cancer Res ; 83(8): 1167-1169, 2023 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-37057598

RESUMO

Although brain metastases are 10-fold more prevalent than primary brain cancers, relatively little is understood about the genes and pathways that promote metastatic cell entry, growth, and survival in the brain. Hence, determining how metastatic tumors colonize the brain and thrive within the neural microenvironment is a topic of both fundamental importance and direct clinical relevance. In this issue, a report by Karreman and colleagues explores pathways that are exploited by metastatic tumor cells to arrest in the circulation, cross the endothelial blood-brain barrier (BBB), and thrive in the brain microenvironment. The authors used elegant imaging tools including intravital fluorescence microcopy and serial reconstruction of ultrastructural sections to analyze BBB breach and subsequent colonization of the brain. They show that matrix metalloprotease 9 (MMP9) plays a central role in these events. Pharmacologic or genetic targeting of MMP9 significantly reduced penetration across the BBB and limited micrometastasis formation. Surprisingly, extravasation and brain colonization does not involve significant degradation of canonical MMP9 protein targets such as collagen and laminin in vascular basement membranes, indicating the requirement for other extracellular matrix (ECM) or non-ECM substrates for MMP9. Collectively, these new and important findings reveal cell-cell adhesion and signaling events between cerebral endothelial and metastatic cancer cells as well as identify potential therapeutic targets to prevent metastatic tumor cell dissemination in the brain. See related article by Karreman et al., p. 1299.


Assuntos
Barreira Hematoencefálica , Neoplasias , Humanos , Barreira Hematoencefálica/metabolismo , Endotélio Vascular/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Encéfalo/metabolismo , Linhagem Celular Tumoral , Neoplasias/metabolismo
14.
Biochem Biophys Res Commun ; 426(4): 578-84, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-22982319

RESUMO

Integrins link the extracellular matrix (ECM) to the cytoskeleton to control cell behaviors including adhesion, spreading and migration. Band 4.1 proteins contain 4.1, ezrin, radixin, moesin (FERM) domains that likely mediate signaling events and cytoskeletal reorganization via integrins. However, the mechanisms by which Band 4.1 proteins and integrins are functionally interconnected remain enigmatic. Here we have investigated roles for Band 4.1 proteins in integrin-mediated cell spreading using primary astrocytes as a model system. We demonstrate that Proteins 4.1B and 4.1G show dynamic patterns of sub-cellular localization in astrocytes spreading on fibronectin. During early stages of cell spreading Proteins 4.1B and 4.1G are enriched in ECM adhesion sites but become more diffusely localized at later stages of spreading. Combinatorial inactivation of Protein 4.1B and 4.1G expression leads to impaired astrocyte spreading. Furthermore, in exogenous expression systems we show that the isolated Protein 4.1 FERM domain significantly enhances integrin-mediated cell spreading. Protein 4.1B is dispensable for reactive astrogliosis in experimental models of cortical injury, likely due to functional compensation by related Protein 4.1 family members. Collectively, these findings reveal that Band 4.1 proteins are important intracellular components for integrin-mediated cell spreading.


Assuntos
Astrócitos/fisiologia , Movimento Celular , Proteínas do Citoesqueleto/metabolismo , Cadeias beta de Integrinas/metabolismo , Proteínas de Membrana/metabolismo , Animais , Astrócitos/patologia , Proteínas do Citoesqueleto/genética , Gliose/metabolismo , Gliose/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Mutantes
15.
Am J Pathol ; 178(2): 609-20, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21281793

RESUMO

Integrins are heterodimeric receptors that regulate cell adhesion, migration, and apoptosis. Integrin αvß8 is most abundantly expressed in kidney and brain, and its major ligand is latent transforming growth factor-ß (TGF-ß). Kidney αvß8 localizes to mesangial cells, which appose glomerular endothelial cells and maintain glomerular capillary structure by mechanical and poorly understood paracrine mechanisms. To establish kidney αvß8 function, mice with homozygous Itgb8 deletion (Itgb8(-/-)) were generated on outbred and C57BL/6 congenic backgrounds. Most Itgb8(-/-) mice died in utero, and surviving Itgb8(-/-) mice failed to gain weight, and rarely survived beyond 6 weeks. A renal glomerular phenotype included azotemia and albuminuria, as well as increased platelet endothelial cell adhesion molecule-1 (PECAM-1) expression, which was surprisingly not associated with conventional functions, such as endothelial cell hyperplasia, hypertrophy, or perivascular inflammation. Itgb8(-/-) mesangial cells demonstrated reduced latent TGF-ß binding, resulting in bioactive TGF-ß release, which stimulated glomerular endothelial cell apoptosis. Using PECAM-1 gain and loss of function strategies, we show that PECAM-1 provides endothelial cytoprotection against mesangial cell TGF-ß. These results clarify a singular mechanism of mesangial-to-endothelial cell cross-talk, whereby mesangial cell αvß8 homeostatically arbitrates glomerular microvascular integrity by sequestering TGF-ß in its latent conformation. Under pathological conditions associated with decreased mesangial cell αvß8 expression and TGF-ß secretion, compensatory PECAM-1 modulation facilitates glomerular endothelial cell survival.


Assuntos
Citoproteção , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Integrinas/metabolismo , Células Mesangiais/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Apoptose , Cruzamentos Genéticos , Células Endoteliais/ultraestrutura , Feminino , Regulação da Expressão Gênica , Heterozigoto , Integrinas/deficiência , Masculino , Células Mesangiais/patologia , Células Mesangiais/ultraestrutura , Camundongos , Comunicação Parácrina , Fenótipo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Ligação Proteica
16.
Dev Dyn ; 240(1): 271-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21181944

RESUMO

Morphogenesis of the heart is regulated by various cues, including growth factors and extracellular matrix (ECM) proteins. The mechanisms by which cardiac cells properly integrate these cues to regulate growth, differentiation, and migration remain poorly understood. Here we have used genetic strategies in mice to identify αvß8 integrin and its cytoskeletal adaptor protein, Band 4.1B, as essential regulators of cardiac morphogenesis. We demonstrate that approximately 60% of mouse embryos genetically null for ß8 integrin and Band 4.1B display cardiovascular phenotypes and die by E11.5. This premature death is due, in part, to defective development of the cardiac outflow tract (OFT), with reduced expression of smooth muscle α-actin (SMAα-actin) in OFT cells derived from the cardiac neural crest. These data are the first to identify cell adhesion and signaling pathways regulated by αvß8 integrin and Band 4.1B as essential for normal formation and function of the heart during embryogenesis.


Assuntos
Proteínas do Citoesqueleto/fisiologia , Coração/embriologia , Cadeias beta de Integrinas/fisiologia , Proteínas de Membrana/fisiologia , Animais , Anormalidades Cardiovasculares/embriologia , Anormalidades Cardiovasculares/genética , Anormalidades Cardiovasculares/metabolismo , Sistema Nervoso Central/anormalidades , Sistema Nervoso Central/irrigação sanguínea , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Cadeias beta de Integrinas/genética , Cadeias beta de Integrinas/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
17.
Glia ; 59(11): 1579-87, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21674628

RESUMO

Neurogenesis in the post-natal brain occurs in two primary locations: the subgranular layer of the hippocampal dentate gyrus and the subventricular zone (SVZ) of the lateral ventricles. Following differentiation, neuroblasts within the SVZ migrate several millimeters to the olfactory bulbs (OBs) via a distinct anatomic route, or rostral migratory stream (RMS). The genes that govern neuroblast directional migration, and particularly those encoding cell adhesion and signaling factors, remain largely uncharacterized. Here, we report that the extracellular matrix adhesion receptor, ß8 integrin, is essential for proper neuroblast chain formation and directional navigation in the RMS. Primary neuroblasts isolated from the mouse brain express robust levels of ß8 integrin protein, and selective ablation of ß8 integrin gene expression in neuroblasts leads to aberrant chain migration and size-reduced OBs. These integrin-dependent defects can be recapitulated ex vivo using isolated neurospheres or SVZ explants. Collectively, these data identify essential cell-intrinsic functions for ß8 integrin in regulating neuroblast polarity and directional navigation in the mouse forebrain.


Assuntos
Movimento Celular/fisiologia , Cadeias beta de Integrinas/genética , Cadeias beta de Integrinas/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Neurogênese/fisiologia , Neurônios/fisiologia , Animais , Western Blotting , Movimento Celular/genética , Polaridade Celular/genética , Polaridade Celular/fisiologia , Células Cultivadas , Giro Denteado/citologia , Giro Denteado/fisiologia , Imunofluorescência , Proteínas de Fluorescência Verde , Imuno-Histoquímica , Imunoprecipitação , Ventrículos Laterais/citologia , Ventrículos Laterais/fisiologia , Camundongos , Camundongos Knockout , Bulbo Olfatório/citologia , Bulbo Olfatório/crescimento & desenvolvimento
18.
Lab Invest ; 91(11): 1554-63, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21876535

RESUMO

The various organs of the body harbor blood vessel networks that display unique structural and functional features; however, the mechanisms that control organ-specific vascular development and physiology remain mostly unknown. In the developing mouse brain, αvß8 integrin-mediated TGF-ß activation and signaling is essential for normal blood vessel growth and sprouting. Whether integrins activate TGF-ß signaling pathways in vascular endothelial cells (ECs), neural cells, or both, has yet to be determined. Here, we have generated and characterized mice in which TGF-ß receptors are specifically deleted in neuroepithelial cells via Nestin-Cre, or in ECs via a novel Cre transgenic strain (Alk1(GFPCre)) in which Cre is expressed under control of the endogenous activin receptor-like kinase 1 (Alk1) promoter. We report that deletion of Tgfbr2 in the neuroepithelium does not impact brain vascular development. In contrast, selective deletion of the Tgfbr2 or Alk5 genes in ECs result in embryonic lethality because of brain-specific vascular pathologies, including blood vessel morphogenesis and intracerebral hemorrhage. These data reveal for the first time that αvß8 integrin-activated TGF-ßs regulate angiogenesis in the developing brain via paracrine signaling to ECs.


Assuntos
Cérebro/irrigação sanguínea , Cérebro/embriologia , Células Endoteliais/fisiologia , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Astrócitos/metabolismo , Galactosídeos , Imuno-Histoquímica , Indóis , Integrases , Proteínas de Filamentos Intermediários , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso , Nestina , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Vitronectina/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/genética
19.
J Cutan Pathol ; 38(7): 570-5, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21375561

RESUMO

BACKGROUND: Alpha v integrins are receptors for many extracellular matrix (ECM) protein ligands, including latent transforming growth factor betas (TGFßs). Various studies in mice have shown that ablation of genes encoding αv integrin or TGFß signaling pathway components leads to spontaneous squamous cell carcinomas (SCCs) in the conjunctiva and periocular skin. Here, we have analyzed patterns of αv integrin protein expression and TGFß signaling in human eyelid and periorbital SCC samples. METHODS: An anti-αv integrin antibody was used to immunostain 19 eyelid and periorbital SCC samples. Additionally, tissue lysates from resected normal eyelid and SCC samples were analyzed by immunoblotting for αv integrin protein. Tumor sections were also immunostained with an antibody directed against Smad2, an intracellular signaling protein that is phosphorylated by TGFß receptors. RESULTS: Alpha v integrin protein was highly expressed in the invasive and less-differentiated components of human SCCs. Lower levels of αv integrin protein were detected in more differentiated components of tumors, as well as in SCC in situ. Patterns of phosphorylated Smad2 immunoreactivity correlated with levels αv integrin expression. CONCLUSIONS: Alpha v integrin was expressed at robust levels in tumor cells representing less differentiated, more invasive components of SCC; by contrast, well-differentiated cells as well as SCC in situ expressed low levels of αv integrin protein.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Neoplasias Oculares/metabolismo , Neoplasias Oculares/patologia , Neoplasias Palpebrais/metabolismo , Neoplasias Palpebrais/patologia , Integrina alfaV/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/análise , Western Blotting , Feminino , Humanos , Imuno-Histoquímica , Integrina alfaV/análise , Masculino , Pessoa de Meia-Idade
20.
Curr Opin Hematol ; 16(3): 209-14, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19318941

RESUMO

PURPOSE OF REVIEW: Central nervous system (CNS) neurovascular units are multicellular complexes consisting of neurons and astrocytes, vascular endothelial cells and pericytes, as well as an assortment of growth factors and extracellular matrix (ECM) proteins. Here, I will discuss the current knowledge of signaling networks essential for the development and physiology of CNS neurovascular units, particularly in the brain. RECENT FINDINGS: Molecular genetic studies have identified various signaling proteins that regulate the formation and function of CNS neurovascular units. These include members of the integrin family of ECM adhesion receptors, ECM proteins such as Wnts and latent transforming growth factor betas, and various transcriptional regulators, including beta-catenin and the inhibitors of DNA binding (Ids). SUMMARY: Neurovascular units are the cellular and molecular interfaces between the circulatory system and the CNS. Recent molecular genetic analyses in mice and other model organisms have revealed the first mechanisms underlying bidirectional communication between neural and vascular components. In particular, ECM-mediated adhesion and signaling pathways have been identified as essential for neurovascular development and physiology. Understanding how these various gene products normally control neurovascular unit formation and function will lend new insights into the causes and possible treatments of debilitating neurovascular-related diseases such as birth defects, stroke, and age-related dementia.


Assuntos
Astrócitos/fisiologia , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Células Endoteliais/fisiologia , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Animais , Astrócitos/citologia , Adesão Celular/fisiologia , Células Endoteliais/citologia , Proteínas da Matriz Extracelular/metabolismo , Humanos , Neurônios/citologia , Pericitos/citologia , Pericitos/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA