Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 20(1): e1010851, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38190417

RESUMO

Blood vessels in different vascular beds vary in size, which is essential for their function and fluid flow along the vascular network. Molecular mechanisms involved in the formation of a vascular lumen of appropriate size, or tubulogenesis, are still only partially understood. Src homology 2 domain containing E (She) protein was previously identified in a screen for proteins that interact with Abelson (Abl)-kinase. However, its biological role has remained unknown. Here we demonstrate that She and Abl signaling regulate vessel size in zebrafish embryos and human endothelial cell culture. Zebrafish she mutants displayed increased endothelial cell number and enlarged lumen size of the dorsal aorta (DA) and defects in blood flow, eventually leading to the DA collapse. Vascular endothelial specific overexpression of she resulted in a reduced diameter of the DA, which correlated with the reduced arterial cell number and lower endothelial cell proliferation. Chemical inhibition of Abl signaling in zebrafish embryos caused a similar reduction in the DA diameter and alleviated the she mutant phenotype, suggesting that She acts as a negative regulator of Abl signaling. Enlargement of the DA size in she mutants correlated with an increased endothelial expression of claudin 5a (cldn5a), which encodes a protein enriched in tight junctions. Inhibition of cldn5a expression partially rescued the enlarged DA in she mutants, suggesting that She regulates DA size, in part, by promoting cldn5a expression. SHE knockdown in human endothelial umbilical vein cells resulted in a similar increase in the diameter of vascular tubes, and also increased phosphorylation of a known ABL downstream effector CRKL. These results argue that SHE functions as an evolutionarily conserved inhibitor of ABL signaling and regulates vessel and lumen size during vascular tubulogenesis.


Assuntos
Peixe-Zebra , Domínios de Homologia de src , Animais , Humanos , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , China , Etnicidade , Transdução de Sinais/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Claudina-5
2.
Am J Pathol ; 194(4): 574-598, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37838010

RESUMO

This study sought to define key molecules and signals controlling major steps in vascular morphogenesis, and how these signals regulate pericyte recruitment and pericyte-induced basement membrane deposition. The morphogenic impact of endothelial cell (EC) expression of activating mutants of Kirsten rat sarcoma virus (kRas), mitogen-activated protein kinase 1 (Mek1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), Akt serine/threonine kinase 1 (Akt1), Ras homolog enriched in brain (Rheb) Janus kinase 2 (Jak2), or signal transducer and activator of transcription 3 (Stat3) expression versus controls was evaluated, along with EC signaling events, pharmacologic inhibitor assays, and siRNA suppression experiments. Primary stimulators of EC lumen formation included kRas, Akt1, and Mek1, whereas PIK3CA and Akt1 stimulated a specialized type of cystic lumen formation. In contrast, the key drivers of EC sprouting behavior were Jak2, Stat3, Mek1, PIK3CA, and mammalian target of rapamycin (mTor). These conclusions are further supported by pharmacologic inhibitor and siRNA suppression experiments. EC expression of active Akt1, kRas, and PIK3CA led to markedly dysregulated lumen formation coupled to strongly inhibited pericyte recruitment and basement membrane deposition. For example, activated Akt1 expression in ECs excessively stimulated lumen formation, decreased EC sprouting behavior, and showed minimal pericyte recruitment with reduced mRNA expression of platelet-derived growth factor-BB, platelet-derived growth factor-DD, and endothelin-1, critical EC-derived factors known to stimulate pericyte invasion. The study identified key signals controlling fundamental steps in capillary morphogenesis and maturation and provided mechanistic details on why EC activating mutations induced a capillary deficiency state with abnormal lumens, impaired pericyte recruitment, and basement deposition: predisposing stimuli for the development of vascular malformations.


Assuntos
Células Endoteliais , Proteínas Proto-Oncogênicas p21(ras) , Proteínas Proto-Oncogênicas p21(ras)/genética , Células Endoteliais/metabolismo , Morfogênese/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Mutação , RNA Interferente Pequeno/metabolismo , Classe I de Fosfatidilinositol 3-Quinases/metabolismo
3.
Am J Pathol ; 193(12): 2203-2217, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37689384

RESUMO

Five growth factors [ie, insulin, fibroblast growth factor-2 (FGF-2), stem cell factor, IL-3, and stromal-derived factor 1α] in combination are necessary for human endothelial cells (ECs) to undergo tube morphogenesis, a process requiring both lumen formation and sprouting behavior. This study investigated why these factors are required by subdividing the factors into 4 separate groups: insulin-only, insulin and FGF-2, no FGF-2 (all factors but without FGF-2), and all factors. The study found that the insulin-only condition failed to support EC morphogenesis or survival, the insulin and FGF-2 condition supported primarily EC lumen formation, and the no FGF-2 condition supported EC sprouting behavior. By comparison, the all-factors condition more strongly stimulated both EC lumen formation and sprouting behavior, and signaling analysis revealed prolonged stimulation of multiple promorphogenic signals coupled with inhibition of proregressive signals. Pharmacologic inhibition of Jak kinases more selectively blocked EC sprouting behavior, whereas inhibition of Raf, phosphatidylinositol 3-kinase, and Akt kinases showed selective blockade of lumen formation. Inhibition of Src family kinases and Notch led to increased sprouting coupled to decreased lumen formation, whereas inhibition of Pak, Mek, and mammalian target of rapamycin kinases blocked both sprouting and lumen formation. These findings reveal novel downstream biological and signaling activities of defined factors that are required for the assembly of human EC-lined capillary tube networks.


Assuntos
Células Endoteliais , Insulinas , Humanos , Células Endoteliais/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células Cultivadas , Morfogênese , Insulinas/metabolismo
4.
Am J Pathol ; 193(9): 1319-1334, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37328101

RESUMO

This study sought to identify potential mechanisms by which k-RasV12-expressing endothelial cell (EC) tubes demonstrate an increased propensity to regress compared with controls. Activated k-Ras mutations play a role in a variety of pathological conditions, including arteriovenous malformations, which are prone to bleed, causing serious hemorrhagic complications. ECs expressing active k-RasV12 demonstrate markedly excessive lumen formation with widened and shortened tubes accompanied by reduced pericyte recruitment and basement membrane deposition, leading to deficient capillary network assembly. The current study showed that active k-Ras-expressing ECs secreted greater amounts of MMP-1 proenzyme compared with control ECs, and readily converted it to increased active MMP-1 levels through the action of plasmin or plasma kallikrein (generated from their added zymogens). Active MMP-1 degraded three-dimensional collagen matrices, leading to more rapid and extensive regression of the active k-Ras-expressing EC tubes, in conjunction with matrix contraction, compared with control ECs. Under conditions where pericytes protect control EC tubes from plasminogen- and MMP-1-dependent tube regression, this failed to occur with k-RasV12 ECs, due to reduced pericyte interactions. In summary, k-RasV12-expressing EC vessels showed an increased propensity to regress in response to serine proteinases through accentuated levels of active MMP-1, a novel pathogenic mechanism that may underlie hemorrhagic events associated with arteriovenous malformation lesions.


Assuntos
Malformações Arteriovenosas , Metaloproteinase 1 da Matriz , Humanos , Metaloproteinase 1 da Matriz/metabolismo , Colágeno/metabolismo , Células Endoteliais/metabolismo , Fibrinolisina/metabolismo , Malformações Arteriovenosas/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 43(9): 1599-1616, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37409533

RESUMO

Because of structural and cellular differences (ie, degrees of matrix abundance and cross-linking, mural cell density, and adventitia), large and medium-sized vessels, in comparison to capillaries, react in a unique manner to stimuli that induce vascular disease. A stereotypical vascular injury response is ECM (extracellular matrix) remodeling that occurs particularly in larger vessels in response to injurious stimuli, such as elevated angiotensin II, hyperlipidemia, hyperglycemia, genetic deficiencies, inflammatory cell infiltration, or exposure to proinflammatory mediators. Even with substantial and prolonged vascular damage, large- and medium-sized arteries, persist, but become modified by (1) changes in vascular wall cellularity; (2) modifications in the differentiation status of endothelial cells, vascular smooth muscle cells, or adventitial stem cells (each can become activated); (3) infiltration of the vascular wall by various leukocyte types; (4) increased exposure to critical growth factors and proinflammatory mediators; and (5) marked changes in the vascular ECM, that remodels from a homeostatic, prodifferentiation ECM environment to matrices that instead promote tissue reparative responses. This latter ECM presents previously hidden matricryptic sites that bind integrins to signal vascular cells and infiltrating leukocytes (in coordination with other mediators) to proliferate, invade, secrete ECM-degrading proteinases, and deposit injury-induced matrices (predisposing to vessel wall fibrosis). In contrast, in response to similar stimuli, capillaries can undergo regression responses (rarefaction). In summary, we have described the molecular events controlling ECM remodeling in major vascular diseases as well as the differential responses of arteries versus capillaries to key mediators inducing vascular injury.


Assuntos
Doenças Vasculares , Lesões do Sistema Vascular , Humanos , Células Endoteliais , Lesões do Sistema Vascular/metabolismo , Matriz Extracelular/metabolismo , Túnica Adventícia , Doenças Vasculares/metabolismo , Remodelação Vascular
6.
Arterioscler Thromb Vasc Biol ; 42(2): 205-222, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34879709

RESUMO

OBJECTIVE: We sought to determine how endothelial cell (EC) expression of the activating k-Ras (kirsten rat sarcoma 2 viral oncogene homolog) mutation, k-RasV12, affects their ability to form lumens and tubes and interact with pericytes during capillary assembly Approach and Results: Using defined bioassays where human ECs undergo observable tubulogenesis, sprouting behavior, pericyte recruitment to EC-lined tubes, and pericyte-induced EC basement membrane deposition, we assessed the impact of EC k-RasV12 expression on these critical processes that are necessary for proper capillary network formation. This mutation, which is frequently seen in human ECs within brain arteriovenous malformations, was found to markedly accentuate EC lumen formation mechanisms, with strongly accelerated intracellular vacuole formation, vacuole fusion, and lumen expansion and with reduced sprouting behavior, leading to excessively widened tube networks compared with control ECs. These abnormal tubes demonstrate strong reductions in pericyte recruitment and pericyte-induced EC basement membranes compared with controls, with deficiencies in fibronectin, collagen type IV, and perlecan deposition. Analyses of signaling during tube formation from these k-RasV12 ECs reveals strong enhancement of Src (Src proto-oncogene, non-receptor tyrosine kinase), Pak2 (P21 [RAC1 (Rac family small GTPase 1)] activated kinase 2), b-Raf (v-raf murine sarcoma viral oncogene homolog B1), Erk (extracellular signal-related kinase), and Akt (AK strain transforming) activation and increased expression of PKCε (protein kinase C epsilon), MT1-MMP (membrane-type 1 matrix metalloproteinase), acetylated tubulin and CDCP1 (CUB domain-containing protein 1; most are known EC lumen regulators). Pharmacological blockade of MT1-MMP, Src, Pak, Raf, Mek (mitogen-activated protein kinase) kinases, Cdc42 (cell division cycle 42)/Rac1, and Notch markedly interferes with lumen and tube formation from these ECs. CONCLUSIONS: Overall, this novel work demonstrates that EC expression of k-RasV12 disrupts capillary assembly due to markedly excessive lumen formation coupled with strongly reduced pericyte recruitment and basement membrane deposition, which are critical pathogenic features predisposing the vasculature to develop arteriovenous malformations.


Assuntos
Membrana Basal/citologia , Capilares/fisiologia , Células Endoteliais/citologia , Neovascularização Fisiológica , Pericitos/citologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Membrana Basal/metabolismo , Linhagem Celular , Células Endoteliais/metabolismo , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Mutação , Pericitos/metabolismo
7.
Proc Natl Acad Sci U S A ; 117(42): 26494-26502, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33020273

RESUMO

During the progression of ocular diseases such as retinopathy of prematurity and diabetic retinopathy, overgrowth of retinal blood vessels results in the formation of pathological neovascular tufts that impair vision. Current therapeutic options for treating these diseases include antiangiogenic strategies that can lead to the undesirable inhibition of normal vascular development. Therefore, strategies that eliminate pathological neovascular tufts while sparing normal blood vessels are needed. In this study we exploited the hyaloid vascular network in murine eyes, which naturally undergoes regression after birth, to gain mechanistic insights that could be therapeutically adapted for driving neovessel regression in ocular diseases. We found that endothelial cells of regressing hyaloid vessels underwent down-regulation of two structurally related E-26 transformation-specific (ETS) transcription factors, ETS-related gene (ERG) and Friend leukemia integration 1 (FLI1), prior to apoptosis. Moreover, the small molecule YK-4-279, which inhibits the transcriptional and biological activity of ETS factors, enhanced hyaloid regression in vivo and drove Human Umbilical Vein Endothelial Cells (HUVEC) tube regression and apoptosis in vitro. Importantly, exposure of HUVECs to sheer stress inhibited YK-4-279-induced apoptosis, indicating that low-flow vessels may be uniquely susceptible to YK-4-279-mediated regression. We tested this hypothesis by administering YK-4-279 to mice in an oxygen-induced retinopathy model that generates disorganized and poorly perfused neovascular tufts that mimic human ocular diseases. YK-4-279 treatment significantly reduced neovascular tufts while sparing healthy retinal vessels, thereby demonstrating the therapeutic potential of this inhibitor.


Assuntos
Olho/irrigação sanguínea , Proteínas Oncogênicas/metabolismo , Proteína Proto-Oncogênica c-fli-1/metabolismo , Regulador Transcricional ERG/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Vasos Sanguíneos/patologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Indóis/farmacologia , Camundongos , Oxigênio/metabolismo , Proteínas Proto-Oncogênicas c-ets/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-ets/metabolismo , Vasos Retinianos/patologia
8.
Am J Pathol ; 191(12): 2245-2264, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34563512

RESUMO

Whether alterations in the microtubule cytoskeleton affect the ability of endothelial cells (ECs) to sprout and form branching networks of tubes was investigated in this study. Bioassays of human EC tubulogenesis, where both sprouting behavior and lumen formation can be rigorously evaluated, were used to demonstrate that addition of the microtubule-stabilizing drugs, paclitaxel, docetaxel, ixabepilone, and epothilone B, completely interferes with EC tip cells and sprouting behavior, while allowing for EC lumen formation. In bioassays mimicking vasculogenesis using single or aggregated ECs, these drugs induce ring-like lumens from single cells or cyst-like spherical lumens from multicellular aggregates with no evidence of EC sprouting behavior. Remarkably, treatment of these cultures with a low dose of the microtubule-destabilizing drug, vinblastine, led to an identical result, with complete blockade of EC sprouting, but allowing for EC lumen formation. Administration of paclitaxel in vivo markedly interfered with angiogenic sprouting behavior in developing mouse retina, providing corroboration. These findings reveal novel biological activities for pharmacologic agents that are widely utilized in multidrug chemotherapeutic regimens for the treatment of human malignant cancers. Overall, this work demonstrates that manipulation of microtubule stability selectively interferes with the ability of ECs to sprout, a necessary step to initiate and form branched capillary tube networks.


Assuntos
Inibidores da Angiogênese/farmacologia , Neovascularização Patológica/tratamento farmacológico , Paclitaxel/farmacologia , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/crescimento & desenvolvimento , Células Cultivadas , Docetaxel/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/crescimento & desenvolvimento , Epotilonas/farmacologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese/efeitos dos fármacos , Neovascularização Patológica/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Paclitaxel/análogos & derivados
9.
Development ; 145(17)2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-30042182

RESUMO

Although major progress in our understanding of the genes and mechanisms that regulate lymphatic vasculature development has been made, we still do not know how lumen formation and maintenance occurs. Here, we identify the Ras-interacting protein Rasip1 as a key player in this process. We show that lymphatic endothelial cell-specific Rasip1-deficient mouse embryos exhibit enlarged and blood-filled lymphatics at embryonic day 14.5. These vessels have patent lumens with disorganized junctions. Later on, as those vessels become fragmented and lumens collapse, cell junctions become irregular. In addition, Rasip1 deletion at later stages impairs lymphatic valve formation. We determined that Rasip1 is essential for lymphatic lumen maintenance during embryonic development by regulating junction integrity, as Rasip1 loss results in reduced levels of junction molecules and defective cytoskeleton organization in vitro and in vivo We determined that Rasip1 regulates Cdc42 activity, as deletion of Cdc42 results in similar phenotypes to those seen following the loss of Rasip1 Furthermore, ectopic Cdc42 expression rescues the phenotypes in Rasip1-deficient lymphatic endothelial cells, supporting the suggestion that Rasip1 regulates Cdc42 activity to regulate cell junctions and cytoskeleton organization, which are both activities required for lymphatic lumen maintenance.


Assuntos
Proteínas de Transporte/metabolismo , Citoesqueleto/metabolismo , Embrião de Mamíferos/embriologia , Células Endoteliais/metabolismo , Vasos Linfáticos/embriologia , Junções Íntimas/metabolismo , Animais , Proteínas de Transporte/genética , Citoesqueleto/genética , Embrião de Mamíferos/citologia , Células Endoteliais/citologia , Peptídeos e Proteínas de Sinalização Intracelular , Vasos Linfáticos/citologia , Camundongos , Camundongos Transgênicos , Junções Íntimas/genética , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo
10.
Cell Immunol ; 364: 104360, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33866285

RESUMO

Ig-GAD2, an antigen-specific immune modulator, requires bone marrow (BM) cell transfer in order to restore beta (ß)-cell formation and induce recovery from established type 1 diabetes (T1D). The BM cells provide endothelial precursor cells (EPCs) that give rise to islet resident endothelial cells (ECs). This study shows that, during development of T1D, the immune attack causes collateral damage to the islet vascular network. The EPC-derived ECs repair and restore islet blood vessel integrity. In addition, ß-cell genetic tracing indicates that the newly formed ß-cells originate from residual ß-cells that escaped the immune attack and, unexpectedly, from ß-cell precursors. This indicates that the rejuvenated islet microenvironment invigorates formation of new ß-cells not only from residual ß-cells but also from precursor cells. This is twofold significant from the perspective of precursor cells as a safe reserve for restoration of ß-cell mass and its promise for therapy of T1D long after diagnosis.


Assuntos
Células da Medula Óssea/fisiologia , Diabetes Mellitus Tipo 1/terapia , Células Progenitoras Endoteliais/fisiologia , Fatores Imunológicos/uso terapêutico , Células Secretoras de Insulina/fisiologia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Diferenciação Celular , Autorrenovação Celular , Células Cultivadas , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Glutamato Descarboxilase/genética , Humanos , Imunoglobulinas/genética , Fatores Imunológicos/genética , Camundongos , Camundongos Endogâmicos NOD , Proteínas Recombinantes de Fusão/genética , Regeneração , Fluxo Sanguíneo Regional
11.
Arterioscler Thromb Vasc Biol ; 40(11): 2632-2648, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32814441

RESUMO

OBJECTIVE: We sought to identify and investigate the functional role of the major endothelial cell (EC)-derived factors that control pericyte recruitment to EC tubes and pericyte-induced tube maturation during capillary network formation. Approach and Results: We identify PDGF (platelet-derived growth factor)-BB, PDGF-DD, ET (endothelin)-1, TGF (transforming growth factor)-ß, and HB-EGF (heparin-binding epidermal growth factor), as the key individual and combined regulators of pericyte assembly around EC tubes. Using novel pericyte only assays, we demonstrate that PDGF-BB, PDGF-DD, and ET-1 are the primary direct drivers of pericyte invasion. Their addition to pericytes induces invasion as if ECs were present. In contrast, TGF-ß and HB-EGF have minimal ability to directly stimulate pericyte invasion. In contrast, TGF-ß1 can act as an upstream pericyte primer to stimulate invasion in response to PDGFs and ET-1. HB-EGF stimulates pericyte proliferation along with PDGFs and ET-1. Using EC-pericyte cocultures, individual, or combined blockade of these EC-derived factors, or their pericyte receptors, using neutralizing antibodies or chemical inhibitors, respectively, interferes with pericyte recruitment and proliferation. As individual factors, PDGF-BB and ET-1 have the strongest impact on these events. However, when the blocking reagents are combined to interfere with each of the above factors or their receptors, more dramatic and profound blockade of pericyte recruitment, proliferation, and pericyte-induced basement membrane deposition occurs. Under these conditions, ECs form tubes that become much wider and less elongated as if pericytes were absent. CONCLUSIONS: Overall, these new studies define and characterize a functional role for key EC-derived factors controlling pericyte recruitment, proliferation, and pericyte-induced basement membrane deposition during capillary network assembly.


Assuntos
Proteínas Angiogênicas/metabolismo , Encéfalo/irrigação sanguínea , Capilares/metabolismo , Movimento Celular , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica , Comunicação Parácrina , Pericitos/metabolismo , Proteínas Angiogênicas/farmacologia , Becaplermina/metabolismo , Capilares/citologia , Capilares/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Endotelina-1/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Linfocinas/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Comunicação Parácrina/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 40(12): 2891-2909, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33086871

RESUMO

OBJECTIVE: In this work, we have sought to define growth factor requirements and the signaling basis for different stages of human vascular morphogenesis and maturation. Approach and Results: Using a serum-free model of endothelial cell (EC) tube morphogenesis in 3-dimensional collagen matrices that depends on a 5 growth factor combination, SCF (stem cell factor), IL (interleukin)-3, SDF (stromal-derived factor)-1α, FGF (fibroblast growth factor)-2, and insulin (factors), we demonstrate that VEGF (vascular endothelial growth factor) pretreatment of ECs for 8 hours (ie, VEGF priming) leads to marked increases in the EC response to the factors which includes; EC tip cells, EC tubulogenesis, pericyte recruitment and proliferation, and basement membrane deposition. VEGF priming requires VEGFR2, and the effect of VEGFR2 is selective to the priming response and does not affect factor-dependent tubulogenesis in the absence of priming. Key molecule and signaling requirements for VEGF priming include RhoA, Rock1 (Rho-kinase), PKCα (protein kinase C α), and PKD2 (protein kinase D2). siRNA suppression or pharmacological blockade of these molecules and signaling pathways interfere with the ability of VEGF to act as an upstream primer of downstream factor-dependent EC tube formation as well as pericyte recruitment. VEGF priming was also associated with the formation of actin stress fibers, activation of focal adhesion components, upregulation of the EC factor receptors, c-Kit, IL-3Rα, and CXCR4 (C-X-C chemokine receptor type 4), and upregulation of EC-derived PDGF (platelet-derived growth factor)-BB, PDGF-DD, and HB-EGF (heparin-binding epidermal growth factor) which collectively affect pericyte recruitment and proliferation. CONCLUSIONS: Overall, this study defines a signaling signature for a separable upstream VEGF priming step, which can activate ECs to respond to downstream factors that are necessary to form branching tube networks with associated mural cells.


Assuntos
Indutores da Angiogênese/farmacologia , Comunicação Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Pericitos/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Comunicação Celular/genética , Células Cultivadas , Técnicas de Cocultura , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Neovascularização Fisiológica/genética , Fosforilação , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/agonistas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 40(2): 365-377, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31852224

RESUMO

OBJECTIVE: In this work, we examine the molecular basis for capillary tube regression and identify key proregressive factors, signaling pathways, and pharmacological antagonists of this process. Approach and Results: We demonstrate that the proinflammatory mediators, IL (interleukin)-1ß, TNF (tumor necrosis factor) α, and thrombin, singly and in combination, are potent regulators of capillary tube regression in vitro. These proregressive factors, when added to endothelial cell-pericyte cocultures, led to selective loss of endothelial cell-lined tube networks, with retention and proliferation of pericytes despite the marked destruction of adjacent capillary tubes. Moreover, treatment of macrophages with the TLR (toll-like receptor) agonists Pam3CSK4 and lipopolysaccharide generates conditioned media with marked proregressive activity, that is completely blocked by a combination of neutralizing antibodies directed to IL-1ß and TNFα but not to other factors. The same combination of blocking antibodies, as well as the anti-inflammatory cytokine IL-10, interfere with macrophage-dependent hyaloid vasculature regression in mice suggesting that proinflammatory cytokine signaling regulates capillary regression in vivo. In addition, we identified a capillary regression signaling signature in endothelial cells downstream of these proregressive agents that is characterized by increased levels of ICAM-1 (intercellular adhesion molecule-1), phospho-p38, and phospho-MLC2 (myosin light chain-2) and decreased levels of phospho-Pak2, acetylated tubulin, phospho-cofilin, and pro-caspase3. Finally, we identified combinations of pharmacological agents (ie, FIST and FISTSB) that markedly rescue the proregressive activities of IL-1ß, TNFα, and thrombin, individually and in combination. CONCLUSIONS: Overall, these new studies demonstrate that the major proinflammatory mediators, IL-1ß, TNFα, and thrombin, are key regulators of capillary tube regression-a critical pathological process regulating human disease.


Assuntos
Capilares/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Inflamação/metabolismo , Trombina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Capilares/patologia , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Endotélio Vascular/patologia , Feminino , Humanos , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
14.
Angiogenesis ; 21(3): 425-532, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29766399

RESUMO

The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.


Assuntos
Bioensaio/métodos , Neoplasias , Neovascularização Patológica , Animais , Bioensaio/instrumentação , Guias como Assunto , Humanos , Camundongos , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
15.
Development ; 142(17): 3058-70, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26253403

RESUMO

The Rho family of small GTPases has been shown to be required in endothelial cells (ECs) during blood vessel formation. However, the underlying cellular events controlled by different GTPases remain unclear. Here, we assess the cellular mechanisms by which Cdc42 regulates mammalian vascular morphogenesis and maintenance. In vivo deletion of Cdc42 in embryonic ECs (Cdc42(Tie2KO)) results in blocked lumen formation and endothelial tearing, leading to lethality of mutant embryos by E9-10 due to failed blood circulation. Similarly, inducible deletion of Cdc42 (Cdc42(Cad5KO)) at mid-gestation blocks angiogenic tubulogenesis. By contrast, deletion of Cdc42 in postnatal retinal vessels leads to aberrant vascular remodeling and sprouting, as well as markedly reduced filopodia formation. We find that Cdc42 is essential for organization of EC adhesion, as its loss results in disorganized cell-cell junctions and reduced focal adhesions. Endothelial polarity is also rapidly lost upon Cdc42 deletion, as seen by failed localization of apical podocalyxin (PODXL) and basal actin. We link observed failures to a defect in F-actin organization, both in vitro and in vivo, which secondarily impairs EC adhesion and polarity. We also identify Cdc42 effectors Pak2/4 and N-WASP, as well as the actomyosin machinery, to be crucial for EC actin organization. This work supports the notion of Cdc42 as a central regulator of the cellular machinery in ECs that drives blood vessel formation.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Citoesqueleto/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Neovascularização Fisiológica , Proteína cdc42 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Animais , Aorta/metabolismo , Apoptose , Vasos Sanguíneos/citologia , Adesão Celular , Polaridade Celular , Proliferação de Células , Sobrevivência Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Matriz Extracelular/metabolismo , Feminino , Deleção de Genes , Integrases/metabolismo , Camundongos Knockout , Modelos Biológicos , Gravidez , Pseudópodes/metabolismo , Receptor TIE-2/metabolismo , Vasos Retinianos/embriologia , Vasos Retinianos/metabolismo , Saco Vitelino/irrigação sanguínea , Saco Vitelino/metabolismo
16.
Circ Res ; 119(7): 810-26, 2016 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-27486147

RESUMO

RATIONALE: Vascular tubulogenesis is essential to cardiovascular development. Within initial vascular cords of endothelial cells, apical membranes are established and become cleared of cell-cell junctions, thereby allowing continuous central lumens to open. Rasip1 (Ras-interacting protein 1) is required for apical junction clearance, as well as for regulation of Rho GTPase (enzyme that hydrolyzes GTP) activity. However, it remains unknown how activities of different Rho GTPases are coordinated by Rasip1 to direct tubulogenesis. OBJECTIVE: The aim of this study is to determine the mechanisms downstream of Rasip1 that drive vascular tubulogenesis. METHODS AND RESULTS: Using conditional mouse mutant models and pharmacological approaches, we dissect GTPase pathways downstream of Rasip1. We show that clearance of endothelial cell apical junctions during vascular tubulogenesis depends on Rasip1, as well as the GTPase Cdc42 (cell division control protein 42 homolog) and the kinase Pak4 (serine/threonine-protein kinase 4). Genetic deletion of Rasip1 or Cdc42, or inhibition of Pak4, all blocks endothelial cell tubulogenesis. By contrast, inactivation of RhoA (Ras homologue gene family member A) signaling leads to vessel overexpansion, implicating actomyosin contractility in control of lumen diameter. Interestingly, blocking activity of NMII (nonmuscle myosin II) either before, or after, lumen morphogenesis results in dramatically different tubulogenesis phenotypes, suggesting time-dependent roles. CONCLUSIONS: Rasip1 controls different pools of GTPases, which in turn regulate different pools of NMII to coordinate junction clearance (remodeling) and actomyosin contractility during vascular tubulogenesis. Rasip1 promotes activity of Cdc42 to activate Pak4, which in turn activates NMII, clearing apical junctions. Once lumens open, Rasip1 suppresses actomyosin contractility via inhibition of RhoA by Arhgap29, allowing controlled expansion of vessel lumens during embryonic growth. These findings elucidate the stepwise processes regulated by Rasip1 through downstream Rho GTPases and NMII.


Assuntos
Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Proteínas de Transporte/fisiologia , Miosina Tipo II/metabolismo , Transdução de Sinais/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Desenvolvimento Embrionário/fisiologia , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Gravidez
17.
Angiogenesis ; 19(2): 173-90, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26897025

RESUMO

Cardiovascular function depends on patent, continuous and stable blood vessel formation by endothelial cells (ECs). Blood vessel development initiates by vasculogenesis, as ECs coalesce into linear aggregates and organize to form central lumens that allow blood flow. Molecular mechanisms underlying in vivo vascular 'tubulogenesis' are only beginning to be unraveled. We previously showed that the GTPase-interacting protein called Rasip1 is required for the formation of continuous vascular lumens in the early embryo. Rasip1(-/-) ECs exhibit loss of proper cell polarity and cell shape, disrupted localization of EC-EC junctions and defects in adhesion of ECs to extracellular matrix. In vitro studies showed that Rasip1 depletion in cultured ECs blocked tubulogenesis. Whether Rasip1 is required in blood vessels after their initial formation remained unclear. Here, we show that Rasip1 is essential for vessel formation and maintenance in the embryo, but not in quiescent adult vessels. Rasip1 is also required for angiogenesis in three models of blood vessel growth: in vitro matrix invasion, retinal blood vessel growth and directed in vivo angiogenesis assays. Rasip1 is thus necessary in growing embryonic blood vessels, postnatal angiogenic sprouting and remodeling, but is dispensable for maintenance of established blood vessels, making it a potential anti-angiogenic therapeutic target.


Assuntos
Proteínas de Transporte/metabolismo , Neovascularização Fisiológica , Vasos Retinianos/embriologia , Vasos Retinianos/metabolismo , Envelhecimento/metabolismo , Animais , Aorta/crescimento & desenvolvimento , Feminino , Deleção de Genes , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Integrases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Gravidez
18.
FASEB J ; 29(12): 4989-5005, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26310270

RESUMO

Talin (tln) binds and activates integrins to couple extracellular matrix-bound integrins to the cytoskeleton; however, its role in heart development is not well characterized. We identified the defective gene and the resulting cardiovascular phenotypes in zebrafish tln1(fl02k) mutants. The ethylnitrosourea-induced fl02k mutant showed heart failure, brain hemorrhage, and diminished cardiac and vessel lumens at 52 h post fertilization. Positional cloning revealed a nonsense mutation of tln1 in this mutant. tln1, but neither tln2 nor -2a, was dominantly expressed in the heart and vessels. Unlike tln1 and -2 in the mouse heart, the unique tln1 expression in the heart enabled us, for the first time, to determine the critical roles of Tln1 in the maintenance of cardiac sarcomeric Z-disks and endothelial/endocardial cell integrity, partly through regulating F-actin networks in zebrafish. The similar expression profiles of tln1 and integrin ß1b (itgb1b) and synergistic function of the 2 genes revealed that itgb1b is a potential partner for tln1 in the stabilization of cardiac Z-disks and vessel lumens. Taken together, the results of this work suggest that Tln1-mediated Itgß1b plays a crucial role in maintaining cardiac sarcomeric Z-disks and endothelial/endocardial cell integrity in zebrafish and may also help to gain molecular insights into congenital heart diseases.


Assuntos
Endotélio Vascular/citologia , Coração/embriologia , Talina/fisiologia , Sequência de Aminoácidos , Animais , Células Endoteliais da Veia Umbilical Humana , Humanos , Integrina beta1/genética , Camundongos , Dados de Sequência Molecular , Mutação , Homologia de Sequência de Aminoácidos , Talina/química , Talina/genética , Peixe-Zebra/embriologia
19.
Blood ; 121(17): 3521-30, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23444400

RESUMO

Vascular tube morphogenesis requires the establishment of endothelial cell (EC) apical-basal polarity in three-dimensional (3D) extracellular matrices. To date, there is little understanding of how EC polarity is controlled during these highly dynamic and rapid morphogenic events. We show that the microtubule tip complex proteins, end binding 1 (EB1), p150(Glued), and Clasp1, control human EC tube formation by (1) inducing microtubule assembly and asymmetric cytoskeletal polarization, whereby acetylated and detyrosinated tubulins distribute in a subapical membrane location and filamentous actin distributes basally; (2) increasing tubulin posttranslational modifications, including required acetylation events; and (3) regulating an EC lumen signaling cascade that involves membrane type 1 matrix metallopatrinase (MT1-MMP)-dependent proteolysis as well as Pak, Raf, and Erk kinases. Another regulator of this process is the microtubule stabilizing protein, tau, which binds p150(Glued) and similarly affects EC lumen formation by controlling the levels of acetylated and detyrosinated tubulins. Increased expression of the tubulin deacetylases, sirtuin 2, and histone deacetylase 6 (HDAC6), blocks EC tube formation and cytoskeletal polarization, while siRNA suppression of these deacetylases stimulates these events. Overall, this work reveals a fundamental role for microtubule tip complex proteins in coordinating microtubule assembly, posttranslational modifications including acetylation, and apical-basal cytoskeletal polarization to control the developing apical membrane surface during blood vessel tubulogenesis in 3D matrix environments.


Assuntos
Polaridade Celular , Células Endoteliais da Veia Umbilical Humana/citologia , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neovascularização Fisiológica , Acetilação , Células Cultivadas , Citoesqueleto/metabolismo , Complexo Dinactina , Matriz Extracelular/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Immunoblotting , Técnicas Imunoenzimáticas , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Morfogênese/fisiologia , Processamento de Proteína Pós-Traducional , RNA Interferente Pequeno/genética , Tubulina (Proteína)/metabolismo , Tirosina/metabolismo
20.
Connect Tissue Res ; 56(5): 392-402, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26305158

RESUMO

This review addresses fundamental mechanisms underlying how capillaries form in three-dimensional extracellular matrices and how endothelial cells (ECs) and pericytes co-assemble to form capillary networks. In addition to playing a critical role in supplying oxygen and nutrients to tissues, recent work suggests that blood vessels supply important signals to facilitate tissue development. Here, we hypothesize that another major function of capillaries is to supply signals to suppress major disease mechanisms including inflammation, infection, thrombosis, hemorrhage, edema, ischemic injury, fibrosis, autoimmune disease and tumor growth/progression. Capillary dysfunction plays a key pathogenic role in many human diseases, and thus, this suppressing function may be attenuated and central toward the initiation and progression of disease. We describe how capillaries form through creation of EC-lined tube networks and vascular guidance tunnels in 3D extracellular matrices. Pericytes recruit to the abluminal EC tube surface within these tunnel spaces, and work together to assemble the vascular basement membrane matrix. These processes occur under serum-free conditions in 3D collagen or fibrin matrices and in response to five key growth factors which are stem cell factor, interleukin-3, stromal-derived factor-1α, fibroblast growth factor-2 and insulin. In addition, we identified a key role for EC-derived platelet-derived growth factor-BB and heparin-binding epidermal growth factor in pericyte recruitment and proliferation to promote EC-pericyte tube co-assembly and vascular basement membrane matrix deposition. A molecular understanding of capillary morphogenesis and maturation should lead to novel therapeutic strategies to repair capillary dysfunction in major human disease contexts including cancer and diabetes.


Assuntos
Células Endoteliais/citologia , Matriz Extracelular/metabolismo , Morfogênese/fisiologia , Neovascularização Fisiológica/fisiologia , Pericitos/citologia , Animais , Membrana Basal/metabolismo , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA