Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 321(6): C964-C977, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34586897

RESUMO

Pulmonary microvascular endothelial cells (PMVECs) uniquely express an α1G-subtype of voltage-gated T-type Ca2+ channel. We have previously revealed that the α1G channel functions as a background Ca2+ entry pathway that is critical for the cell proliferation, migration, and angiogenic potential of PMVECs, a novel function attributed to the coupling between α1G-mediated Ca2+ entry and constitutive Akt phosphorylation and activation. Despite this significance, mechanism(s) that link the α1G-mediated Ca2+ entry to Akt phosphorylation remain incompletely understood. In this study, we demonstrate that Ca2+/calmodulin-dependent protein kinase (CaMK) 4 serves as a downstream effector of the α1G-mediated Ca2+ entry to promote the angiogenic potential of PMVECs. Notably, CaMK2 and CaMK4 are both expressed in PMVECs. Pharmacological blockade or genetic knockdown of the α1G channel led to a significant reduction in the phosphorylation level of CaMK4 but not the phosphorylation level of CaMK2. Pharmacological inhibition as well as genetic knockdown of CaMK4 significantly decreased cell proliferation, migration, and network formation capacity in PMVECs. However, CaMK4 inhibition or knockdown did not alter Akt phosphorylation status in PMVECs, indicating that α1G/Ca2+/CaMK4 is independent of the α1G/Ca2+/Akt pathway in sustaining the cells' angiogenic potential. Altogether, these findings suggest a novel α1G-CaMK4 signaling complex that regulates the Ca2+-dominated angiogenic potential in PMVECs.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Sinalização do Cálcio , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Células Endoteliais/enzimologia , Pulmão/irrigação sanguínea , Microvasos/enzimologia , Neovascularização Fisiológica , Inibidores da Angiogênese/farmacologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/genética , Movimento Celular , Proliferação de Células , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Masculino , Microvasos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos Sprague-Dawley
2.
Am J Physiol Cell Physiol ; 316(3): C353-C364, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30649917

RESUMO

Pulmonary microvascular endothelial cells (PMVECs) display a rapid angioproliferative phenotype, essential for maintaining homeostasis in steady-state and promoting vascular repair after injury. Although it has long been established that endothelial cytosolic Ca2+ ([Ca2+]i) transients are required for proliferation and angiogenesis, mechanisms underlying such regulation and the transmembrane channels mediating the relevant [Ca2+]i transients remain incompletely understood. In the present study, the functional role of the microvascular endothelial site-specific α1G T-type Ca2+ channel in angiogenesis was examined. PMVECs intrinsically possess an in vitro angiogenic "network formation" capacity. Depleting extracellular Ca2+ abolishes network formation, whereas blockade of vascular endothelial growth factor receptor or nitric oxide synthase has little or no effect, suggesting that the network formation is a [Ca2+]i-dependent process. Blockade of the T-type Ca2+ channel or silencing of α1G, the only voltage-gated Ca2+ channel subtype expressed in PMVECs, disrupts network formation. In contrast, blockade of canonical transient receptor potential (TRP) isoform 4 or TRP vanilloid 4, two other Ca2+ permeable channels expressed in PMVECs, has no effect on network formation. T-type Ca2+ channel blockade also reduces proliferation, cell-matrix adhesion, and migration, three major components of angiogenesis in PMVECs. An in vivo study demonstrated that the mice lacking α1G exhibited a profoundly impaired postinjury cell proliferation in the lungs following lipopolysaccharide challenge. Mechanistically, T-type Ca2+ channel blockade reduces Akt phosphorylation in a dose-dependent manner. Blockade of Akt or its upstream activator, phosphatidylinositol-3-kinase (PI3K), also impairs network formation. Altogether, these findings suggest a novel functional role for the α1G T-type Ca2+ channel to promote the cell's angiogenic potential via a PI3K-Akt signaling pathway.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Células Endoteliais/metabolismo , Pulmão/metabolismo , Neovascularização Patológica/metabolismo , Animais , Cálcio/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Feminino , Lipopolissacarídeos/farmacologia , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Fosfatidilinositol 3-Quinase/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPC/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
PLoS One ; 10(8): e0135533, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26274589

RESUMO

Microparticles (MPs) are released constitutively and from activated cells. MPs play significant roles in vascular homeostasis, injury, and as biomarkers. The unique glycocalyx on the membrane of cells has frequently been exploited to identify specific cell types, however the glycocalyx of the MPs has yet to be defined. Thus, we sought to determine whether MPs, released both constitutively and during injury, from vascular cells have a glycocalyx matching those of the parental cell type to provide information on MP origin. For these studies we used rat pulmonary microvascular and artery endothelium, pulmonary smooth muscle, and aortic endothelial cells. MPs were collected from healthy or cigarette smoke injured cells and analyzed with a panel of lectins for specific glycocalyx linkages. Intriguingly, we determined that the MPs released either constitutively or stimulated by CSE injury did not express the same glycocalyx of the parent cells. Further, the glycocalyx was not unique to any of the specific cell types studied. These data suggest that MPs from both normal and healthy vascular cells do not share the parental cell glycocalyx makeup.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Glicocálix/química , Lectinas/metabolismo , Fumar/efeitos adversos , Animais , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Glicocálix/efeitos dos fármacos , Glicocálix/metabolismo , Microscopia Eletrônica de Transmissão , Ratos
4.
Am J Respir Crit Care Med ; 178(6): 558-64, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18556624

RESUMO

The plexiform lesions of severe pulmonary arterial hypertension (PAH) are similar in histologic appearance, whether the disease is idiopathic or secondary. Both forms of the disease show actively proliferating endothelial cells without evidence of apoptosis. Here, we discuss the pathobiology of the atypical, angioproliferative endothelial cells in severe PAH. The concept of the endothelial cell as a "quasi-malignant" cell provides a new framework for antiproliferative, antiangiogenic therapy in severe PAH.


Assuntos
Hipertensão Pulmonar/fisiopatologia , Proliferação de Células , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/patologia , Imuno-Histoquímica , Neoplasias/fisiopatologia , Artéria Pulmonar/patologia , Terminologia como Assunto , Vasoconstrição/fisiologia , Proteínas Wnt/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 294(3): L431-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17981956

RESUMO

Pulmonary microvascular endothelial cells (PMVECs) are enriched with progenitor cells that underlie their rapid proliferation and vasculogenic capacity. However, the molecular basis for such an enhanced growth potential is unknown. Nucleosome assembly protein-1 (NAP1), and its related family of proteins, have been incriminated in control of cell growth in a range of species. We therefore sought to determine whether NAP1 contributes to the rapid proliferation and vasculogenesis that is observed in PMVECs. NAP1 was expressed at a high level in two fast-growing cell types, including PMVECs and resident microvascular endothelial progenitor cells that were selected from PMVECs, whereas it was expressed at a low level in slow-growing pulmonary artery endothelial cells (PAECs). Heterologous NAP1 expression increased the growth potential of PAECs, whereas inhibiting NAP1 expression reduced the growth potential of PMVECs. Despite its impact on endothelial cell growth, NAP1 did not influence the expression of endothelial cell-selective markers (PECAM-1, VE-cadherin, von Willebrand factor), and it did not influence cell type-specific lectin binding criterion; PMVECs interact with Griffonia simplicifolia lectin, whereas PAECs interact with Helix pomatia lectin. PMVECs possess a higher basal transelectrical resistance than do PAECs, indicative of their more restrictive barrier property. Changing NAP1 expression did not normalize this basal barrier function between PMVECs and PAECs. To examine whether the growth-promoting actions of NAP1 influence blood vessel formation, endothelial cells were mixed into Matrigel and subcutaneously implanted. PMVECs generated eightfold more blood vessels than did PAECs over a 10-day time course. Heterologous NAP1 expression in PAECs increased the number of blood vessels formed by this cell type, where blood vessel growth approached that seen with PMVECs. Thus, our findings indicate that NAP1 functions as an important regulator of the proliferative and vasculogenic endothelial cell phenotype without globally impacting endothelial cell phenotype specification.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Endotélio Vascular/citologia , Microcirculação/fisiologia , Neovascularização Fisiológica/fisiologia , Proteínas Nucleares/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Microcirculação/citologia , Proteína 1 de Modelagem do Nucleossomo , Fenótipo
6.
Ultrastruct Pathol ; 28(2): 109-13, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15205111

RESUMO

A 56-year-old woman presented with a right inguinal mass. Biopsy revealed multiple lymph nodes involved with papillary and gland-like structures extending into the surrounding fibroadipose tissue. There was no history of carcinoma or other malignancy. Ultrastructural findings included long microvilli, desmosomes, and tonofilament bundles, indicating metastatic malignant mesothelioma. Malignant mesothelioma only rarely presents as a lymph node metastasis, and electron microscopy is very useful in establishing this diagnosis.


Assuntos
Canal Inguinal/patologia , Metástase Linfática/patologia , Mesotelioma/patologia , Mesotelioma/secundário , Adenocarcinoma/patologia , Diagnóstico Diferencial , Hemangiossarcoma/patologia , Humanos , Metástase Linfática/ultraestrutura , Masculino , Melanoma/patologia , Mesotelioma/ultraestrutura , Microscopia Eletrônica , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA