Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
EMBO Mol Med ; 15(4): e16128, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36740996

RESUMO

Vascular endothelial protein tyrosine phosphatase (VE-PTP) influences endothelial barrier function by regulating the activation of tyrosine kinase receptor Tie2. We determined whether this action is linked to the development of atherosclerosis by examining the influence of arterial shear stress on VE-PTP, Tie2 activation, plasma leakage, and atherogenesis. We found that exposure to high average shear stress led to downstream polarization and endocytosis of VE-PTP accompanied by Tie2 activation at cell junctions. In aortic regions with disturbed flow, VE-PTP was not redistributed away from Tie2. Endothelial cells exposed to high shear stress had greater Tie2 activation and less macromolecular permeability than regions with disturbed flow. Deleting endothelial VE-PTP in VE-PTPiECKO mice increased Tie2 activation and reduced plasma leakage in atheroprone regions. ApoE-/- mice bred with VE-PTPiECKO mice had less plasma leakage and fewer atheromas on a high-fat diet. Pharmacologic inhibition of VE-PTP by AKB-9785 had similar anti-atherogenic effects. Together, the findings identify VE-PTP as a novel target for suppression of atherosclerosis.


Assuntos
Aterosclerose , Placa Aterosclerótica , Camundongos , Animais , Células Endoteliais/metabolismo , Placa Aterosclerótica/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Aterosclerose/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo
2.
Am J Pathol ; 190(12): 2355-2375, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33039355

RESUMO

Despite many reports about pulmonary blood vessels in lung fibrosis, the contribution of lymphatics to fibrosis is unknown. We examined the mechanism and consequences of lymphatic remodeling in mice with lung fibrosis after bleomycin injury or telomere dysfunction. Widespread lymphangiogenesis was observed after bleomycin treatment and in fibrotic lungs of prospero homeobox 1-enhanced green fluorescent protein (Prox1-EGFP) transgenic mice with telomere dysfunction. In loss-of-function studies, blocking antibodies revealed that lymphangiogenesis 14 days after bleomycin treatment was dependent on vascular endothelial growth factor (Vegf) receptor 3 signaling, but not on Vegf receptor 2. Vegfc gene and protein expression increased specifically. Extensive extravasated plasma, platelets, and macrophages at sites of lymphatic growth were potential sources of Vegfc. Lymphangiogenesis peaked at 14 to 28 days after bleomycin challenge, was accompanied by doubling of chemokine (C-C motif) ligand 21 in lung lymphatics and tertiary lymphoid organ formation, and then decreased as lung injury resolved by 56 days. In gain-of-function studies, expansion of the lung lymphatic network by transgenic overexpression of Vegfc in club cell secretory protein (CCSP)/VEGF-C mice reduced macrophage accumulation and fibrosis and accelerated recovery after bleomycin treatment. These findings suggest that lymphatics have an overall protective effect in lung injury and fibrosis and fit with a mechanism whereby lung lymphatic network expansion reduces lymph stasis and increases clearance of fluid and cells, including profibrotic macrophages.


Assuntos
Proliferação de Células/fisiologia , Fibrose/patologia , Lesão Pulmonar/patologia , Linfangiogênese/fisiologia , Fator C de Crescimento do Endotélio Vascular/metabolismo , Animais , Fibrose/metabolismo , Vasos Linfáticos/patologia , Macrófagos/metabolismo , Camundongos Transgênicos , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Sci Adv ; 4(8): eaat4758, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30101193

RESUMO

Tumor lymphangiogenesis is accompanied by a higher incidence of sentinel lymph node metastasis and shorter overall survival in several types of cancer. We asked whether tumor lymphangiogenesis might also occur in distant organs with established metastases and whether it might promote further metastatic spread of those metastases to other organs. Using mouse metastasis models, we found that lymphangiogenesis occurred in distant lung metastases and that some metastatic tumor cells were located in lymphatic vessels and draining lymph nodes. In metastasis-bearing lungs of melanoma patients, a higher lymphatic density within and around metastases and lymphatic invasion correlated with poor outcome. Using a transgenic mouse model with inducible expression of vascular endothelial growth factor C (VEGF-C) in the lung, we found greater growth of lung metastases, with more abundant dissemination to other organs. Our findings reveal unexpected contributions of lymphatics in distant organs to the promotion of growth of metastases and their further spread to other organs, with potential clinical implications for adjuvant therapies in patients with metastatic cancer.


Assuntos
Neoplasias da Mama/patologia , Neoplasias Pulmonares/secundário , Linfangiogênese , Vasos Linfáticos/patologia , Melanoma Experimental/patologia , Neovascularização Patológica/patologia , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/metabolismo , Metástase Linfática , Vasos Linfáticos/metabolismo , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo
4.
J Clin Invest ; 126(9): 3511-25, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27548529

RESUMO

Angiopoietin-2 (ANG2) regulates blood vessel remodeling in many pathological conditions through differential effects on Tie2 signaling. While ANG2 competes with ANG1 to inhibit Tie2, it can paradoxically also promote Tie2 phosphorylation (p-Tie2). A related paradox is that both inactivation and overactivation of Tie2 can result in vascular remodeling. Here, we reconciled these opposing actions of ANG2 by manipulating conditions that govern its actions in the vasculature. ANG2 drove vascular remodeling during Mycoplasma pulmonis infection by acting as a Tie2 antagonist, which led to p-Tie2 suppression, forkhead box O1 (FOXO1) activation, increased ANG2 expression, and vessel leakiness. These changes were exaggerated by anti-Tie2 antibody, inhibition of PI3K signaling, or ANG2 overexpression and were reduced by anti-ANG2 antibody or exogenous ANG1. In contrast, under pathogen-free conditions, ANG2 drove vascular remodeling by acting as an agonist, promoting high p-Tie2, low FOXO1 activation, and no leakage. Tie1 activation was strong under pathogen-free conditions, but infection or TNF-α led to Tie1 inactivation by ectodomain cleavage and promoted the Tie2 antagonist action of ANG2. Together, these data indicate that ANG2 activation of Tie2 supports stable enlargement of normal nonleaky vessels, but reduction of Tie1 in inflammation leads to ANG2 antagonism of Tie2 and initiates a positive feedback loop wherein FOXO1-driven ANG2 expression promotes vascular remodeling and leakage.


Assuntos
Angiopoietina-2/metabolismo , Proteína Forkhead Box O1/antagonistas & inibidores , Receptor TIE-2/metabolismo , Animais , Anticorpos Monoclonais/química , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Feminino , Humanos , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mycoplasma pulmonis , Fosfatidilinositol 3-Quinases/metabolismo , Domínios Proteicos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Remodelação Vascular
5.
J Am Soc Nephrol ; 27(1): 69-77, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26038530

RESUMO

Polycystic kidney diseases (PKD) are genetic disorders characterized by progressive epithelial cyst growth leading to destruction of normally functioning renal tissue. Current therapies have focused on the cyst epithelium, and little is known about how the blood and lymphatic microvasculature modulates cystogenesis. Hypomorphic Pkd1(nl/nl) mice were examined, showing that cystogenesis was associated with a disorganized pericystic network of vessels expressing platelet/endothelial cell adhesion molecule 1 and vascular endothelial growth factor receptor 3 (VEGFR3). The major ligand for VEGFR3 is VEGFC, and there were lower levels of Vegfc mRNA within the kidneys during the early stages of cystogenesis in 7-day-old Pkd1(nl/nl) mice. Seven-day-old mice were treated with exogenous VEGFC for 2 weeks on the premise that this would remodel both the VEGFR3(+) pericystic vascular network and larger renal lymphatics that may also affect the severity of PKD. Treatment with VEGFC enhanced VEGFR3 phosphorylation in the kidney, normalized the pattern of the pericystic network of vessels, and widened the large lymphatics in Pkd1(nl/nl) mice. These effects were associated with significant reductions in cystic disease, BUN and serum creatinine levels. Furthermore, VEGFC administration reduced M2 macrophage pericystic infiltrate, which has been implicated in the progression of PKD. VEGFC administration also improved cystic disease in Cys1(cpk/cpk) mice, a model of autosomal recessive PKD, leading to a modest but significant increase in lifespan. Overall, this study highlights VEGFC as a potential new treatment for some aspects of PKD, with the possibility for synergy with current epithelially targeted approaches.


Assuntos
Doenças Renais Policísticas/tratamento farmacológico , Fator C de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Camundongos , Doenças Renais Policísticas/etiologia , Fator C de Crescimento do Endotélio Vascular/fisiologia
6.
Am J Pathol ; 184(6): 1877-89, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24726646

RESUMO

Vascular remodeling is a feature of sustained inflammation in which capillaries enlarge and acquire the phenotype of venules specialized for plasma leakage and leukocyte recruitment. We sought to determine whether neutrophils are required for vascular remodeling in the respiratory tract by using Mycoplasma pulmonis infection as a model of sustained inflammation in mice. The time course of vascular remodeling coincided with the influx of neutrophils during the first few days after infection and peaked at day 5. Depletion of neutrophils with antibody RB6-8C5 or 1A8 reduced neutrophil influx and vascular remodeling after infection by about 90%. Similarly, vascular remodeling after infection was suppressed in Cxcr2(-/-) mice, in which neutrophils adhered to the endothelium of venules but did not extravasate into the tissue. Expression of the venular adhesion molecule P-selectin increased in endothelial cells from day 1 to day 3 after infection, as did expression of the Cxcr2-receptor ligands Cxcl1 and Cxcl2. Tumor necrosis factor α (TNFα) expression increased more than sixfold in the trachea of wild-type and Cxcr2(-/-) mice, but intratracheal administration of TNFα did not induce vascular remodeling similar to that seen in infection. We conclude that neutrophil influx is required for remodeling of capillaries into venules in the airways of mice with Mycoplasma infection and that TNFα signaling is necessary but not sufficient for vascular remodeling.


Assuntos
Endotélio Vascular/metabolismo , Infecções por Mycoplasma/metabolismo , Mycoplasma pulmonis , Neutrófilos/metabolismo , Sistema Respiratório/metabolismo , Remodelação Vascular , Animais , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Endotélio Vascular/patologia , Feminino , Camundongos , Camundongos Knockout , Infecções por Mycoplasma/genética , Infecções por Mycoplasma/patologia , Neutrófilos/patologia , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Sistema Respiratório/patologia
7.
Circ Res ; 114(5): 806-22, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24429550

RESUMO

RATIONALE: Lymphatic vessels in the respiratory tract normally mature into a functional network during the neonatal period, but under some pathological conditions they can grow as enlarged, dilated sacs that result in the potentially lethal condition of pulmonary lymphangiectasia. OBJECTIVE: We sought to determine whether overexpression of the lymphangiogenic growth factor (vascular endothelial growth factor-C [VEGF-C]) can promote lymphatic growth and maturation in the respiratory tract. Unexpectedly, perinatal overexpression of VEGF-C in the respiratory epithelium led to a condition resembling human pulmonary lymphangiectasia, a life-threatening disorder of the newborn characterized by respiratory distress and the presence of widely dilated lymphatics. METHODS AND RESULTS: Administration of doxycycline to Clara cell secretory protein-reverse tetracycline-controlled transactivator/tetracycline operator-VEGF-C double-transgenic mice during a critical period from embryonic day 15.5 to postnatal day 14 was accompanied by respiratory distress, chylothorax, pulmonary lymphangiectasia, and high mortality. Enlarged sac-like lymphatics were abundant near major airways, pulmonary vessels, and visceral pleura. Side-by-side comparison revealed morphological features similar to pulmonary lymphangiectasia in humans. The condition was milder in mice given doxycycline after age postnatal day 14 and did not develop after postnatal day 35. Mechanistic studies revealed that VEGF recptor (VEGFR)-3 alone drove lymphatic growth in adult mice, but both VEGFR-2 and VEGFR-3 were required for the development of lymphangiectasia in neonates. VEGFR-2/VEGFR-3 heterodimers were more abundant in the dilated lymphatics, consistent with the involvement of both receptors. Despite the dependence of lymphangiectasia on VEGFR-2 and VEGFR-3, the condition was not reversed by blocking both receptors together or by withdrawing VEGF-C. CONCLUSIONS: The findings indicate that VEGF-C overexpression can induce pulmonary lymphangiectasia during a critical period in perinatal development.


Assuntos
Pneumopatias/congênito , Linfangiectasia/congênito , Fator C de Crescimento do Endotélio Vascular/genética , Animais , Feminino , Humanos , Lactente , Pneumopatias/genética , Pneumopatias/metabolismo , Pneumopatias/patologia , Linfangiectasia/genética , Linfangiectasia/metabolismo , Linfangiectasia/patologia , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Gravidez , Edema Pulmonar/genética , Edema Pulmonar/metabolismo , Edema Pulmonar/patologia , Transdução de Sinais/fisiologia , Traqueia/metabolismo , Traqueia/patologia , Uteroglobina/genética , Uteroglobina/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Am J Pathol ; 182(4): 1434-47, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23391392

RESUMO

These studies used bi-transgenic Clara cell secretory protein (CCSP)/IL-1ß mice that conditionally overexpress IL-1ß in Clara cells to determine whether IL-1ß can promote angiogenesis and lymphangiogenesis in airways. Doxycycline treatment induced rapid, abundant, and reversible IL-1ß production, influx of neutrophils and macrophages, and conspicuous and persistent lymphangiogenesis, but surprisingly no angiogenesis. Gene profiling showed many up-regulated genes, including chemokines (Cxcl1, Ccl7), cytokines (tumor necrosis factor α, IL-1ß, and lymphotoxin-ß), and leukocyte genes (S100A9, Aif1/Iba1). Newly formed lymphatics persisted after IL-1ß overexpression was stopped. Further studies examined how IL1R1 receptor activation by IL-1ß induced lymphangiogenesis. Inactivation of vascular endothelial growth factor (VEGF)-C and VEGF-D by adeno-associated viral vector-mediated soluble VEGFR-3 (VEGF-C/D Trap) completely blocked lymphangiogenesis, showing its dependence on VEGFR-3 ligands. Consistent with this mechanism, VEGF-C immunoreactivity was present in some Aif1/Iba1-immunoreactive macrophages. Because neutrophils contribute to IL-1ß-induced lung remodeling in newborn mice, we examined their potential role in lymphangiogenesis. Triple-transgenic CCSP/IL-1ß/CXCR2(-/-) mice had the usual IL-1ß-mediated lymphangiogenesis but no neutrophil recruitment, suggesting that neutrophils are not essential. IL1R1 immunoreactivity was found on some epithelial basal cells and neuroendocrine cells, suggesting that these cells are targets of IL-1ß, but was not detected on lymphatics, blood vessels, or leukocytes. We conclude that lymphangiogenesis triggered by IL-1ß overexpression in mouse airways is driven by VEGF-C/D from macrophages, but not neutrophils, recruited by chemokines from epithelial cells that express IL1R1.


Assuntos
Interleucina-1beta/metabolismo , Linfangiogênese , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Traqueia/irrigação sanguínea , Traqueia/patologia , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Epitélio/metabolismo , Epitélio/patologia , Regulação da Expressão Gênica , Humanos , Hipertrofia , Linfangiogênese/genética , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Camundongos , Camundongos Transgênicos , Neovascularização Patológica/genética , Neutrófilos/metabolismo , Neutrófilos/patologia , Transporte Proteico , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-8B/metabolismo
9.
Am J Pathol ; 178(6): 2897-909, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21550017

RESUMO

Blood vessel leakiness is an early, transient event in acute inflammation but can also persist as vessels undergo remodeling in sustained inflammation. Angiopoietin/Tie2 signaling can reduce the leakiness through changes in endothelial cells. The role of pericytes in this action has been unknown. We used the selective PDGF-B-blocking oligonucleotide aptamer AX102 to determine whether disruption of pericyte-endothelial crosstalk alters vascular leakiness or remodeling in the airways of mice under four different conditions: i) baseline, ii) acute inflammation induced by bradykinin, iii) sustained inflammation after 7-day infection by the respiratory pathogen Mycoplasma pulmonis, or iv) leakage after bradykinin challenge in the presence of vascular stabilization by the angiopoietin-1 (Ang1) mimic COMP-Ang1 for 7 days. AX102 reduced pericyte coverage but did not alter the leakage of microspheres from tracheal blood vessels at baseline or after bradykinin; however, AX102 exaggerated leakage at 7 days after M. pulmonis infection and increased vascular remodeling and disease severity at 14 days. AX102 also abolished the antileakage effect of COMP-Ang1 at 7 days. Together, these findings show that pericyte contributions to endothelial stability have greater dependence on PDGF-B during the development of sustained inflammation, when pericyte dynamics accompany vascular remodeling, than under baseline conditions or in acute inflammation. The findings also show that the antileakage action of Ang1 requires PDGF-dependent actions of pericytes in maintaining endothelial stability.


Assuntos
Angiopoietina-1/metabolismo , Inflamação/patologia , Pericitos/patologia , Traqueia/irrigação sanguínea , Traqueia/patologia , Actinas/metabolismo , Animais , Aptâmeros de Nucleotídeos/farmacologia , Bradicinina/farmacologia , Contagem de Células , Forma Celular/efeitos dos fármacos , Desmina/metabolismo , Inflamação/complicações , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Infecções por Mycoplasma/complicações , Infecções por Mycoplasma/patologia , Mycoplasma pulmonis/efeitos dos fármacos , Mycoplasma pulmonis/fisiologia , Pericitos/efeitos dos fármacos , Pericitos/microbiologia , Proteínas Proto-Oncogênicas c-sis/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-sis/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Traqueia/microbiologia
10.
Dev Dyn ; 239(9): 2354-66, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20730909

RESUMO

Recent advances have documented the development of lung vasculature before and after birth, but less is known of the growth and maturation of airway vasculature. We sought to determine whether airway vasculature changes during the perinatal period and when the typical adult pattern develops. On embryonic day 16.5 mouse tracheas had a primitive vascular plexus unlike the adult airway vasculature, but instead resembling the yolk sac vasculature. Soon after birth (P0), the primitive vascular plexus underwent abrupt and extensive remodeling. Blood vessels overlying tracheal cartilage rings regressed from P1 to P3 but regrew from P4 to P7 to form the hierarchical, segmented, ladder-like adult pattern. Hypoxia and HIF-1α were present in tracheal epithelium over vessels that survived but not where they regressed. These findings reveal the plasticity of airway vasculature after birth and show that these vessels can be used to elucidate factors that promote postnatal vascular remodeling and maturation.


Assuntos
Vasos Sanguíneos , Pulmão , Neovascularização Fisiológica/fisiologia , Animais , Animais Recém-Nascidos , Apoptose/fisiologia , Vasos Sanguíneos/anatomia & histologia , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/crescimento & desenvolvimento , Proliferação de Células , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Feminino , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/irrigação sanguínea , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
11.
Blood ; 116(12): 2173-82, 2010 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-20566898

RESUMO

Lymphotoxin-α (LTα), lymphotoxin-ß (LTß), and tumor necrosis factor-α (TNFα) are inflammatory mediators that play crucial roles in lymphoid organ development. We demonstrate here that LTα also contributes to the function of lymphatic vessels and to lymphangiogenesis during inflammation. LTα(-/-) mice exhibited reduced lymph flow velocities and increased interstitial fluid pressure. Airways of LTß(-/-) mice infected with Mycoplasma pulmonis had significantly more lymphangiogenesis than wild type (WT) or LTα(-/-) mice, as did the skin draining immunization sites of LTß(-/-) mice. Macrophages, B cells, and T cells, known sources of LT and TNFα, were apparent in the skin surrounding the immunization sites as were LTα, LTß, and TNFα mRNAs. Ectopic expression of LTα led to the development of LYVE-1 and Prox1-positive lymphatic vessels within tertiary lymphoid organs (TLOs). Quantification of pancreatic lymphatic vessel density in RIPLTαLTß(-/-) and WT mice revealed that LTα was sufficient for inducing lymphangiogenesis and that LTß was not required for this process. Kidneys of inducible LTα transgenic mice developed lymphatic vessels before the appearance of obvious TLOs. These data indicate that LTα plays a significant role in lymphatic vessel function and in inflammation-associated lymphangiogenesis.


Assuntos
Linfangiogênese , Linfotoxina-alfa/fisiologia , Animais , Imunização , Inflamação , Rim/imunologia , Vasos Linfáticos , Linfotoxina-alfa/deficiência , Linfotoxina-alfa/genética , Linfotoxina-beta , Camundongos , Camundongos Knockout , Infecções por Mycoplasma/patologia , Mycoplasma pulmonis , Pele
12.
Cancer Res ; 70(6): 2213-23, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20197469

RESUMO

Inhibition of angiopoietin-2 (Ang2) can slow tumor growth, but the underlying mechanism is not fully understood. Because Ang2 is expressed in growing blood vessels and promotes angiogenesis driven by vascular endothelial growth factor (VEGF), we asked whether the antitumor effect of Ang2 inhibition results from reduced sprouting angiogenesis and whether the effect is augmented by inhibition of VEGF from tumor cells. Using Colo205 human colon carcinomas in nude mice as a model, we found that selective inhibition of Ang2 by the peptide-Fc fusion protein L1-7(N) reduced the number of vascular sprouts by 46% and tumor growth by 62% over 26 days. Strikingly, when the Ang2 inhibitor was combined with a function-blocking anti-VEGF antibody, the number of sprouts was reduced by 82%, tumor vascularity was reduced by 67%, and tumor growth slowed by 91% compared with controls. The reduction in tumor growth was accompanied by decreased cell proliferation and increased apoptosis. We conclude that inhibition of Ang2 slows tumor growth by limiting the expansion of the tumor vasculature by sprouting angiogenesis, in a manner that is complemented by concurrent inhibition of VEGF and leads to reduced proliferation and increased apoptosis of tumor cells.


Assuntos
Inibidores da Angiogênese/farmacologia , Angiopoietina-2/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Angiopoietina-2/biossíntese , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Processos de Crescimento Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Sinergismo Farmacológico , Humanos , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Ratos , Receptores Fc/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Am J Pathol ; 176(4): 2009-18, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20133818

RESUMO

Vascular endothelial growth factor (VEGF) is a key angiogenic factor in tumors, but less is known about what drives vascular remodeling in inflammation, where plasma leakage and leukocyte influx are prominent features. In chronic airway inflammation in mice infected by the bacterium Mycoplasma pulmonis (M. pulmonis), the segment of the microvasculature that supports leukocyte adhesion and migration expands through remodeling of capillaries into vessels with features of venules. Here, we report that the angiopoietin/Tie2 pathway is an essential driving force for capillary remodeling into venules in M. pulmonis-infected mouse airways. Similar to M. pulmonis infection, systemic overexpression of angiopoietin-1 (Ang1) resulted in remodeling of airway capillaries into venular-like vessels that expressed venous markers like P-selectin, ICAM-1, and EphB4 and were sites of leukocyte adhesion during lipopolysaccharide-induced acute inflammation. Ang1 and Ang2 protein increased in M. pulmonis-infected mouse airways but came from different cellular sources: Ang1 was expressed in infiltrating neutrophils and Ang2 in endothelial cells. Indeed, systemic administration of soluble Tie2 inhibited capillary remodeling, induction of venous markers, and leukocyte influx in M. pulmonis-infected mouse airways. Together, these findings suggest that blockade of the Ang/Tie2 pathway may represent a therapeutic approach in airway inflammation.


Assuntos
Angiopoietina-1/metabolismo , Capilares/metabolismo , Inflamação , Leucócitos/citologia , Receptor TIE-2/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adenoviridae/metabolismo , Animais , Transporte Biológico , Molécula 1 de Adesão Intercelular/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Mycoplasma pulmonis/metabolismo , Vênulas/metabolismo
14.
Am J Pathol ; 176(3): 1525-41, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20093490

RESUMO

Angiogenesis and lymphangiogenesis participate in many inflammatory diseases, and their reversal is thought to be beneficial. However, the extent of reversibility of vessel remodeling is poorly understood. We exploited the potent anti-inflammatory effects of the corticosteroid dexamethasone to test the preventability and reversibility of vessel remodeling in Mycoplasma pulmonis-infected mice using immunohistochemistry and quantitative RT-PCR. In this model robust immune responses drive rapid and sustained changes in blood vessels and lymphatics. In infected mice not treated with dexamethasone, capillaries enlarged into venules expressing leukocyte adhesion molecules, sprouting angiogenesis and lymphangiogenesis occurred, and the inflammatory cytokines tumor necrosis factor and interleukin-1 increased. Concurrent dexamethasone treatment largely prevented the remodeling of blood vessels and lymphatics. Dexamethasone also significantly reduced cytokine expression, bacterial burden, and leukocyte influx into airways and lungs over 4 weeks of infection. In contrast, when infection was allowed to proceed untreated for 2 weeks and then was treated with dexamethasone for 4 weeks, most blood vessel changes reversed but lymphangiogenesis did not, suggesting that different survival mechanisms apply. Furthermore, dexamethasone significantly reduced the bacterial burden and influx of lymphocytes but not of neutrophils or macrophages or cytokine expression. These findings show that lymphatic remodeling is more resistant than blood vessel remodeling to corticosteroid-induced reversal. We suggest that lymphatic remodeling that persists after the initial inflammatory response has resolved may influence subsequent inflammatory episodes in clinical situations.


Assuntos
Dexametasona/farmacologia , Inflamação/patologia , Vasos Linfáticos/efeitos dos fármacos , Vasos Linfáticos/patologia , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/patologia , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/microbiologia , Vasos Sanguíneos/patologia , Movimento Celular/efeitos dos fármacos , Doença Crônica , Citocinas/metabolismo , Feminino , Inflamação/complicações , Inflamação/microbiologia , Mediadores da Inflamação/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/patologia , Vasos Linfáticos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Infecções por Mycoplasma/complicações , Infecções por Mycoplasma/microbiologia , Infecções por Mycoplasma/patologia , Sistema Respiratório/microbiologia , Fatores de Tempo
15.
J Clin Invest ; 119(10): 2954-64, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19759514

RESUMO

Inflammation is associated with blood vessel and lymphatic vessel proliferation and remodeling. The microvasculature of the mouse trachea provides an ideal opportunity to study this process, as Mycoplasma pulmonis infection of mouse airways induces widespread and sustained vessel remodeling, including enlargement of capillaries into venules and lymphangiogenesis. Although the mediators responsible for these vascular changes in mice have not been identified, VEGF-A is known not to be involved. Here, we sought to determine whether TNF-alpha drives the changes in blood vessels and lymphatics in M. pulmonis-infected mice. The endothelial cells, but not pericytes, of blood vessels, but not lymphatics, were immunoreactive for TNF receptor 1 (TNF-R1) and lymphotoxin B receptors. Most TNF-R2 immunoreactivity was on leukocytes. Infection resulted in a large and sustained increase in TNF-alpha expression, as measured by real-time quantitative RT-PCR, and smaller increases in lymphotoxins and TNF receptors that preceded vessel remodeling. Substantially less vessel remodeling and lymphangiogenesis occurred when TNF-alpha signaling was inhibited by a blocking antibody or was silenced in Tnfr1-/- mice. When administered after infection was established, the TNF-alpha-specific antibody slowed but did not reverse blood vessel remodeling and lymphangiogenesis. The action of TNF-alpha on blood vessels is probably mediated through direct effects on endothelial cells, but its effects on lymphangiogenesis may require inflammatory mediators from recruited leukocytes. We conclude that TNF-alpha is a strong candidate for a mediator that drives blood vessel remodeling and lymphangiogenesis in inflammation.


Assuntos
Vasos Sanguíneos , Inflamação/imunologia , Linfangiogênese/imunologia , Vasos Linfáticos , Sistema Respiratório , Fator de Necrose Tumoral alfa/imunologia , Animais , Vasos Sanguíneos/anatomia & histologia , Vasos Sanguíneos/fisiologia , Perfilação da Expressão Gênica , Glicoproteínas/metabolismo , Vasos Linfáticos/anatomia & histologia , Vasos Linfáticos/fisiologia , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Mycoplasma/imunologia , Infecções por Mycoplasma/patologia , Mycoplasma pulmonis , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Sistema Respiratório/anatomia & histologia , Sistema Respiratório/imunologia , Transdução de Sinais/fisiologia
16.
Am J Pathol ; 174(6): 2388-99, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19443703

RESUMO

Both Eph receptors and ephrin ligands have been implicated in blood vessel and neuronal development. Recent studies suggested that EphA2 inhibition reduces tumor angiogenesis, but its role in blood vessel development and inflammation is unclear. We examined these issues using either airways of pathogen-free, EphA2-deficient mice at various ages or EphA2-deficient mice whose airways were inflamed by either Mycoplasma pulmonis infection or ovalbumin sensitization and challenge. EphA2-deficient mice had fewer capillaries, a greater number of endothelial sprouts, and greater capillary diameters than age-matched, wild-type control mice. Moreover, capillaries in EphA2-deficient mice had significantly less pericyte coverage, suggesting abnormal interactions between endothelial cells and pericytes. These differences were apparent in early postnatal life but decreased during progression into adulthood. In inflamed airways, significantly more angiogenesis and lymphangiogenesis, a greater number of infiltrating leukocytes, and higher expression levels of inflammatory cytokine mRNA were present in EphA2-deficient mice after M. pulmonis infection. Additionally, in allergic airway inflammation with ovalbumin sensitization and challenge, a greater number of lymphatic sprouts and infiltrating leukocytes, higher mRNA expression levels of TH2 cytokines and chemokines related to allergic airway inflammation, and enhanced airway hyper-responsiveness were present in EphA2-deficient mice. We conclude that defective pericyte coverage causes capillary defects, abundant endothelial sprouts, and thick capillary diameters in EphA2-deficient mice, indicating that these animals have exaggerated responses to airway inflammation.


Assuntos
Capilares/patologia , Linfangiogênese , Neovascularização Fisiológica , Pneumonia/patologia , Receptor EphA2/deficiência , Alérgenos/imunologia , Animais , Capilares/crescimento & desenvolvimento , Citocinas/biossíntese , Células Endoteliais/patologia , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Infecções por Mycoplasma/imunologia , Infecções por Mycoplasma/patologia , Mycoplasma pulmonis , Ovalbumina/imunologia , Pericitos/patologia , Pneumonia/genética , Pneumonia/metabolismo , Receptor EphA2/genética , Hipersensibilidade Respiratória/induzido quimicamente , Hipersensibilidade Respiratória/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Th2/imunologia
17.
Am J Pathol ; 174(6): 2378-87, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19443705

RESUMO

The integrin alpha5beta1 has been previously implicated in tumor angiogenesis, but its role in the remodeling of both blood vessels and lymphatics during inflammation is at an early stage of understanding. We examined this issue using a selective, small-molecule inhibitor of alpha5beta1 integrin, 2-aroylamino-3-{4-[(pyridin-2-ylaminomethyl)heterocyclyl]phenyl}propionic acid (JSM8757), in a model of sustained airway inflammation in mice with Mycoplasma pulmonis infection, which is known to be accompanied by robust blood vessel remodeling and lymphangiogenesis. The inhibitor significantly decreased the proliferation of lymphatic endothelial cells in culture and the number of lymphatic sprouts and new lymphatics in airways of mice infected for 2 weeks but did not reduce remodeling of blood vessels in the same airways. In inflamed airways, alpha5 integrin immunoreactivity was present on lymphatic sprouts, but not on collecting lymphatics or blood vessels, and was not found on any lymphatics of normal airways. Macrophages, potential targets of the inhibitor, did not have alpha5 integrin immunoreactivity in inflamed airways. In addition, macrophage recruitment, assessed in infected airways by quantitative reverse transcription-polymerase chain reaction measurements of expression of the marker protein ionized calcium-binding adapter molecule 1 (Iba1), was not reduced by JSM8757. We conclude that inhibition of the alpha5beta1 integrin reduces lymphangiogenesis in inflamed airways after M. pulmonis infection because expression of the integrin is selectively increased on lymphatic sprouts and plays an essential role in lymphatic growth.


Assuntos
Linfangiogênese/fisiologia , Pneumonia/metabolismo , Animais , Feminino , Imuno-Histoquímica , Inflamação/metabolismo , Inflamação/microbiologia , Linfangiogênese/efeitos dos fármacos , Vasos Linfáticos/efeitos dos fármacos , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Mycoplasma/metabolismo , Infecções por Mycoplasma/fisiopatologia , Mycoplasma pulmonis , Pneumonia/microbiologia , Propionatos/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Genes Dev ; 22(23): 3282-91, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19056883

RESUMO

The activity of the homeobox gene Prox1 is necessary and sufficient for venous blood endothelial cells (BECs) to acquire a lymphatic endothelial cell (LEC) fate. We determined that the differentiated LEC phenotype is a plastic, reprogrammable condition that depends on constant Prox1 activity for its maintenance. We show that conditional down-regulation of Prox1 during embryonic, postnatal, or adult stages is sufficient to reprogram LECs into BECs. Consequently, the identity of the mutant lymphatic vessels is also partially reprogrammed as they acquire some features typical of the blood vasculature. siRNA-mediated down-regulation of Prox1 in LECs in culture demonstrates that reprogramming of LECs into BECs is a Prox1-dependent, cell-autonomous process. We propose that Prox1 acts as a binary switch that suppresses BEC identity and promotes and maintains LEC identity; switching off Prox1 activity is sufficient to initiate a reprogramming cascade leading to the dedifferentiation of LECs into BECs. Therefore, LECs are one of the few differentiated cell types that require constant expression of a certain gene to maintain their phenotypic identity.


Assuntos
Desdiferenciação Celular/genética , Células Endoteliais/fisiologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Camundongos , Fenótipo , RNA Interferente Pequeno/farmacologia , Proteínas Supressoras de Tumor/genética
19.
Circ Res ; 103(10): 1092-9, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18832750

RESUMO

Vessel stabilization and the inhibition of side effects such as tissue edema are essential in angiogenic gene therapy. Thus, combination gene transfers stimulating both endothelial cell and pericyte proliferation have become of interest. However, there is currently little data to support combination gene transfer in large animal models. In this study, we evaluated the potential advantages of such a strategy by combining the transfer of adenoviral (Ad) vascular endothelial growth factor (VEGF)-A and platelet-derived growth factor (PDGF)-B into rabbit hindlimb skeletal muscle. AdLacZ alone or in combination with AdVEGF-A were used as controls. Contrast-enhanced ultrasound, modified Miles assay, and immunohistology were used to quantify perfusion, vascular permeability, and capillary size, respectively. Confocal microscopy was used in the assessment of pericyte-coverage. The transfer of AdPDGF-B alone and in combination with AdVEGF-A induced prominent proliferation of alpha-smooth muscle actin-, CD31-, RAM11-, HAM56-, and VEGF- positive cells. Although, pericyte recruitment to angiogenic vessels was not improved, combination gene transfer induced a longer-lasting increase in perfusion in both intact and ischemic muscles than AdVEGF-A gene transfer alone. In conclusion, intramuscular delivery of AdVEGF-A and AdPDGF-B, combined, resulted in a prolonged angiogenic response. However, the effects were most likely mediated via paracrine mechanisms rather than an increase in vascular pericyte coverage.


Assuntos
Movimento Celular , Terapia Genética/métodos , Neovascularização Fisiológica , Comunicação Parácrina , Pericitos/metabolismo , Proteínas Proto-Oncogênicas c-sis/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adenoviridae , Animais , Permeabilidade Capilar/genética , Movimento Celular/genética , Proliferação de Células , Humanos , Músculo Esquelético/citologia , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Neovascularização Fisiológica/genética , Comunicação Parácrina/genética , Pericitos/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Proteínas Proto-Oncogênicas c-sis/genética , Coelhos , Fatores de Tempo , Ultrassonografia , Fator A de Crescimento do Endotélio Vascular/genética
20.
Ann N Y Acad Sci ; 1131: 1-12, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18519955

RESUMO

Imaging of lymphangiogenesis and angiogenesis requires robust and unambiguous markers of lymphatic and blood vessels. Although much progress has been made in recent years in identifying molecules specifically expressed on lymphatic and blood vessels, no perfect marker has been found that works reliably in all species, tissues, vascular beds, and in all physiological and pathologic conditions. The heterogeneity of expression of markers in both blood and lymphatic vessels reflects underlying differences in the phenotype of endothelial cells. Use of only one marker can lead to misleading interpretations, but these pitfalls can usually be avoided by use of multiple markers and three-dimensional whole-mount preparations. LYVE-1, VEGFR-3, Prox1, and podoplanin are among the most useful markers for microscopic imaging of lymphatic vessels, but, depending on histologic location, each marker can be expressed by other cell types, including vascular endothelial cells. Other markers, including CD31, junctional proteins, and receptors, such as VEGF-2, are shared by lymphatic and blood vessels.


Assuntos
Biomarcadores/sangue , Imageamento Tridimensional , Linfangiogênese , Endotélio Linfático/metabolismo , Endotélio Linfático/fisiopatologia , Proteínas de Homeodomínio/fisiologia , Vasos Linfáticos/irrigação sanguínea , Vasos Linfáticos/fisiopatologia , Glicoproteínas de Membrana/fisiologia , Neovascularização Patológica , Proteínas Supressoras de Tumor/fisiologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Proteínas de Transporte Vesicular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA