Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Cell Physiol ; 236(6): 4580-4591, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33251612

RESUMO

Bone morphogenetic protein (BMP) signaling is well known in bone homeostasis. However, the physiological effects of BMP signaling on mandibles are largely unknown, as the mandible has distinct functions and characteristics from other bones. In this study, we investigated the roles of BMP signaling in bone homeostasis of the mandibles by deleting BMP type I receptor Acvr1 in osteoblast lineage cells with Osterix-Cre. We found mandibular bone loss in conditional knockout mice at the ages of postnatal day 21 and 42 in an age-dependent manner. The decreased bone mass was related to compromised osteoblast differentiation together with enhanced osteoclastogenesis, which was secondary to the changes in osteoblasts in vivo. In vitro study revealed that deletion of Acvr1 in the mandibular bone marrow stromal cells (BMSCs) significantly compromised osteoblast differentiation. When wild type bone marrow macrophages were cocultured with BMSCs lacking Acvr1 both directly and indirectly, both proliferation and differentiation of osteoclasts were induced as evidenced by an increase of multinucleated cells, compared with cocultured with control BMSCs. Furthermore, we demonstrated that the increased osteoclastogenesis in vitro was at least partially due to the secretion of soluble receptor activator of nuclear factor-κB ligand (sRANKL), which is probably the reason for the mandibular bone loss in vivo. Overall, our results proposed that ACVR1 played essential roles in maintaining mandibular bone homeostasis through osteoblast differentiation and osteoblast-osteoclast communication via sRANKL.


Assuntos
Receptores de Ativinas Tipo I/deficiência , Diferenciação Celular , Deleção de Genes , Mandíbula/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese , Ligante RANK/metabolismo , Receptores de Ativinas Tipo I/genética , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Reabsorção Óssea , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Feminino , Macrófagos/metabolismo , Masculino , Mandíbula/patologia , Células-Tronco Mesenquimais/patologia , Camundongos Knockout , Osteoblastos/patologia , Osteoclastos/patologia , Transdução de Sinais
2.
Free Radic Biol Med ; 129: 127-137, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30227271

RESUMO

The bone morphogenetic protein (BMP) type I receptors ALK2 and ALK3 are essential for expression of hepcidin, a key iron regulatory hormone. In mice, hepatocyte-specific Alk2 deficiency leads to moderate iron overload with periportal liver iron accumulation, while hepatocyte-specific Alk3 deficiency leads to severe iron overload with centrilobular liver iron accumulation and a more marked reduction of basal hepcidin levels. The objective of this study was to investigate whether the two receptors have additive roles in hepcidin regulation. Iron overload in mice with hepatocyte-specific Alk2 and Alk3 (Alk2/3) deficiency was characterized and compared to hepatocyte-specific Alk3 deficient mice. Co-immunoprecipitation studies were performed to detect the formation of ALK2 and ALK3 homodimer and heterodimer complexes in vitro in the presence and absence of ligands. The iron overload phenotype of hepatocyte-specific Alk2/3-deficient mice was more severe than that of hepatocyte-specific Alk3-deficient mice. In vitro co-immunoprecipitation studies in Huh7 cells showed that ALK3 can homodimerize in absence of BMP2 or BMP6. In contrast, ALK2 did not homodimerize in either the presence or absence of BMP ligands. However, ALK2 did form heterodimers with ALK3 in the presence of BMP2 or BMP6. ALK3-ALK3 and ALK2-ALK3 receptor complexes induced hepcidin expression in Huh7 cells. Our data indicate that: (I) ALK2 and ALK3 have additive functions in vivo, as Alk2/3 deficiency leads to a greater degree of iron overload than Alk3 deficiency; (II) ALK3, but not ALK2, undergoes ligand-independent homodimerization; (III) the formation of ALK2-ALK3 heterodimers is ligand-dependent and (IV) both receptor complexes functionally induce hepcidin expression in vitro.


Assuntos
Receptores de Ativinas Tipo I/genética , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 6/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Hepcidinas/genética , Sobrecarga de Ferro/genética , Ferro/metabolismo , Receptores de Ativinas Tipo I/deficiência , Animais , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 6/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepcidinas/metabolismo , Humanos , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Ligação Proteica , Multimerização Proteica , Índice de Gravidade de Doença , Transdução de Sinais
3.
Arterioscler Thromb Vasc Biol ; 38(8): 1821-1836, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29880487

RESUMO

Objective- Diabetic macular edema is a major cause of visual impairment. It is caused by blood-retinal barrier breakdown that leads to vascular hyperpermeability. Current therapeutic approaches consist of retinal photocoagulation or targeting VEGF (vascular endothelial growth factor) to limit vascular leakage. However, long-term intravitreal use of anti-VEGFs is associated with potential safety issues, and the identification of alternative regulators of vascular permeability may provide safer therapeutic options. The vascular specific BMP (bone morphogenetic protein) receptor ALK1 (activin-like kinase receptor type I) and its circulating ligand BMP9 have been shown to be potent vascular quiescence factors, but their role in the context of microvascular permeability associated with hyperglycemia has not been evaluated. Approach and Results- We investigated Alk1 signaling in hyperglycemic endothelial cells and assessed whether BMP9/Alk1 signaling could modulate vascular permeability. We show that high glucose concentrations impair Alk1 signaling, both in cultured endothelial cells and in a streptozotocin model of mouse diabetes mellitus. We observed that Alk1 signaling participates in the maintenance of vascular barrier function, as Alk1 haploinsufficiency worsens the vascular leakage observed in diabetic mice. Conversely, sustained delivery of BMP9 by adenoviral vectors significantly decreased the loss of retinal barrier function in diabetic mice. Mechanistically, we demonstrate that Alk1 signaling prevents VEGF-induced phosphorylation of VE-cadherin and induces the expression of occludin, thus strengthening vascular barrier functions. Conclusions- From these data, we suggest that by preventing retinal vascular permeability, BMP9 could serve as a novel therapeutic agent for diabetic macular edema.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Receptores de Ativinas Tipo I/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Diabetes Mellitus Experimental/terapia , Retinopatia Diabética/prevenção & controle , Células Endoteliais/efeitos dos fármacos , Terapia Genética/métodos , Fatores de Diferenciação de Crescimento , Hiperglicemia/terapia , Edema Macular/prevenção & controle , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II/genética , Animais , Glicemia/metabolismo , Linhagem Celular , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/induzido quimicamente , Retinopatia Diabética/genética , Retinopatia Diabética/metabolismo , Células Endoteliais/metabolismo , Fator 2 de Diferenciação de Crescimento , Fatores de Diferenciação de Crescimento/biossíntese , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/farmacologia , Haploinsuficiência , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hiperglicemia/induzido quimicamente , Hiperglicemia/genética , Hiperglicemia/metabolismo , Edema Macular/induzido quimicamente , Edema Macular/genética , Edema Macular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Estreptozocina , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 38(5): 1216-1229, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29449337

RESUMO

OBJECTIVE: ALK1 (activin-receptor like kinase 1) is an endothelial cell-restricted receptor with high affinity for BMP (bone morphogenetic protein) 9 TGF-ß (transforming growth factor-ß) family member. Loss-of-function mutations in ALK1 cause a subtype of hereditary hemorrhagic telangiectasia-a rare disease characterized by vasculature malformations. Therapeutic strategies are aimed at reducing potential complications because of vascular malformations, but currently, there is no curative treatment for hereditary hemorrhagic telangiectasia. APPROACH AND RESULTS: In this work, we report that a reduction in ALK1 gene dosage (heterozygous ALK1+/- mice) results in enhanced retinal endothelial cell proliferation and vascular hyperplasia at the sprouting front. We found that BMP9/ALK1 represses VEGF (vascular endothelial growth factor)-mediated PI3K (phosphatidylinositol 3-kinase) by promoting the activity of the PTEN (phosphatase and tensin homolog). Consequently, loss of ALK1 function in endothelial cells results in increased activity of the PI3K pathway. These results were confirmed in cutaneous telangiectasia biopsies of patients with hereditary hemorrhagic telangiectasia 2, in which we also detected an increase in endothelial cell proliferation linked to an increase on the PI3K pathway. In mice, genetic and pharmacological inhibition of PI3K is sufficient to abolish the vascular hyperplasia of ALK1+/- retinas and in turn normalize the vasculature. CONCLUSIONS: Overall, our results indicate that the BMP9/ALK1 hub critically mediates vascular quiescence by limiting PI3K signaling and suggest that PI3K inhibitors could be used as novel therapeutic agents to treat hereditary hemorrhagic telangiectasia.


Assuntos
Receptores de Activinas Tipo II/genética , Receptores de Ativinas Tipo I/genética , Células Endoteliais/enzimologia , Mutação , Neovascularização Patológica , Fosfatidilinositol 3-Quinase/metabolismo , Telangiectasia Retiniana/genética , Telangiectasia Hemorrágica Hereditária/genética , Receptores de Ativinas Tipo I/deficiência , Inibidores da Angiogênese/farmacologia , Animais , Estudos de Casos e Controles , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Ativação Enzimática , Deleção de Genes , Predisposição Genética para Doença , Fator 2 de Diferenciação de Crescimento/farmacologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Hiperplasia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Telangiectasia Retiniana/tratamento farmacológico , Telangiectasia Retiniana/enzimologia , Telangiectasia Retiniana/patologia , Transdução de Sinais , Telangiectasia Hemorrágica Hereditária/tratamento farmacológico , Telangiectasia Hemorrágica Hereditária/enzimologia , Telangiectasia Hemorrágica Hereditária/patologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
5.
Mol Ther ; 25(8): 1974-1987, 2017 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-28716575

RESUMO

Trauma-induced heterotopic ossification (tHO) is a condition of pathologic wound healing, defined by the progressive formation of ectopic bone in soft tissue following severe burns or trauma. Because previous studies have shown that genetic variants of HO, such as fibrodysplasia ossificans progressiva (FOP), are caused by hyperactivating mutations of the type I bone morphogenetic protein receptor (T1-BMPR) ACVR1/ALK2, studies evaluating therapies for HO have been directed primarily toward drugs for this specific receptor. However, patients with tHO do not carry known T1-BMPR mutations. Here we show that, although BMP signaling is required for tHO, no single T1-BMPR (ACVR1/ALK2, BMPR1a/ALK3, or BMPR1b/ALK6) alone is necessary for this disease, suggesting that these receptors have functional redundancy in the setting of tHO. By utilizing two different classes of BMP signaling inhibitors, we developed a translational approach to treatment, integrating treatment choice with existing diagnostic options. Our treatment paradigm balances either immediate therapy with reduced risk for adverse effects (Alk3-Fc) or delayed therapy with improved patient selection but greater risk for adverse effects (LDN-212854).


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/genética , Marcação de Genes , Ossificação Heterotópica/etiologia , Ossificação Heterotópica/patologia , Ferimentos e Lesões/complicações , Receptores de Ativinas Tipo I/deficiência , Animais , Anti-Inflamatórios/farmacologia , Biomarcadores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Técnicas de Inativação de Genes , Predisposição Genética para Doença , Humanos , Ligantes , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/metabolismo , Ossificação Heterotópica/prevenção & controle , Inibidores de Proteínas Quinases/farmacologia
6.
Oncotarget ; 7(51): 84314-84325, 2016 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-27741515

RESUMO

Angiogenesis occurs early in tumor development, sustains primary tumor growth and provides a route for metastatic escape. The TGF-ß family receptors modulate angiogenesis via endothelial-cell specific pathways. Here we investigate the interaction of two such receptors, ALK1 and endoglin, in pancreatic neuroendocrine tumors (PanNET). Independently, ALK1 and endoglin deficiencies exhibited genetically divergent phenotypes, while both highly correlate to an endothelial metagene in human and mouse PanNETs. A concurrent deficiency of both receptors synergistically decreased tumor burden to a greater extent than either individual knockdown. Furthermore, the knockout of Gdf2 (BMP9), the primary ligand for ALK1 and endoglin, exhibited a mixed phenotype from each of ALK1 and endoglin deficiencies; overall primary tumor burden decreased, but hepatic metastases increased. Tumors lacking BMP9 display a hyperbranching vasculature, and an increase in vascular mesenchymal-marker expression, which may be implicit in the increase in metastases. Taken together, our work cautions against singular blockade of BMP9 and instead demonstrates the utility of dual blockade of ALK1 and endoglin as a strategy for anti-angiogenic therapy in PanNET.


Assuntos
Receptores de Ativinas Tipo I/genética , Endoglina/genética , Neovascularização Patológica/genética , Tumores Neuroendócrinos/genética , Neoplasias Pancreáticas/genética , Fator de Crescimento Transformador beta/genética , Receptores de Ativinas Tipo I/deficiência , Receptores de Activinas Tipo II , Animais , Endoglina/deficiência , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Fator 2 de Diferenciação de Crescimento/deficiência , Fator 2 de Diferenciação de Crescimento/genética , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neovascularização Patológica/metabolismo , Tumores Neuroendócrinos/metabolismo , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/metabolismo , Carga Tumoral/genética
7.
Angiogenesis ; 19(4): 451-461, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27325285

RESUMO

An abnormally high number of macrophages are present in human brain arteriovenous malformations (bAVM) with or without evidence of prior hemorrhage, causing unresolved inflammation that may enhance abnormal vascular remodeling and exacerbate the bAVM phenotype. The reasons for macrophage accumulation at the bAVM sites are not known. We tested the hypothesis that persistent infiltration and pro-inflammatory differentiation of monocytes in angiogenic tissues increase the macrophage burden in bAVM using two mouse models and human monocytes. Mouse bAVM was induced through deletion of AVM causative genes, Endoglin (Eng) globally or Alk1 focally, plus brain focal angiogenic stimulation. An endothelial cell and vascular smooth muscle cell co-culture system was used to analyze monocyte differentiation in the angiogenic niche. After angiogenic stimulation, the Eng-deleted mice had fewer CD68(+) cells at 2 weeks (P = 0.02), similar numbers at 4 weeks (P = 0.97), and more at 8 weeks (P = 0.01) in the brain angiogenic region compared with wild-type (WT) mice. Alk1-deficient mice also had a trend toward more macrophages/microglia 8 weeks (P = 0.064) after angiogenic stimulation and more RFP(+) bone marrow-derived macrophages than WT mice (P = 0.01). More CD34(+) cells isolated from peripheral blood of patients with ENG or ALK1 gene mutation differentiated into macrophages than those from healthy controls (P < 0.001). These data indicate that persistent infiltration and pro-inflammatory differentiation of monocytes might contribute to macrophage accumulation in bAVM. Blocking macrophage homing to bAVM lesions should be tested as a strategy to reduce the severity of bAVM.


Assuntos
Malformações Arteriovenosas Intracranianas/patologia , Monócitos/patologia , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Diferenciação Celular , Técnicas de Cocultura , Modelos Animais de Doenças , Endoglina/deficiência , Endoglina/genética , Células Endoteliais/patologia , Humanos , Malformações Arteriovenosas Intracranianas/genética , Malformações Arteriovenosas Intracranianas/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miócitos de Músculo Liso/patologia , Neovascularização Patológica/genética
8.
J Endocrinol ; 229(3): 331-41, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27029473

RESUMO

Pituitary follicle-stimulating hormone (FSH) synthesis is regulated by transforming growth factorßsuperfamily ligands, most notably the activins and inhibins. Bone morphogenetic proteins (BMPs) also regulate FSHß subunit (Fshb) expression in immortalized murine gonadotrope-like LßT2 cells and in primary murine or ovine primary pituitary cultures. BMP2 signals preferentially via the BMP type I receptor, BMPR1A, to stimulate murine Fshb transcription in vitro Here, we used a Cre-lox approach to assess BMPR1A's role in FSH synthesis in mice in vivo Gonadotrope-specific Bmpr1a knockout animals developed normally and had reproductive organ weights comparable with those of controls. Knockouts were fertile, with normal serum gonadotropins and pituitary gonadotropin subunit mRNA expression. Cre-mediated recombination of the floxed Bmpr1a allele was efficient and specific, as indicated by PCR analysis of diverse tissues and isolated gonadotrope cells. Furthermore, BMP2 stimulation of inhibitor of DNA binding 3 expression was impaired in gonadotropes isolated from Bmpr1a knockout mice, confirming the loss of functional receptor protein in these cells. Treatment of purified gonadotropes with small-molecule inhibitors of BMPR1A (and the related receptors BMPR1B and ACVR1) suppressed Fshb mRNA expression, suggesting that an autocrine BMP-like molecule might regulate FSH synthesis. However, deletion of Bmpr1a and Acvr1 in cultured pituitary cells did not alter Fshb expression, indicating that the inhibitors had off-target effects. In sum, BMPs or related ligands acting via BMPR1A or ACVR1 are unlikely to play direct physiological roles in FSH synthesis by murine gonadotrope cells.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Fertilidade/fisiologia , Gonadotrofos/fisiologia , Gonadotropinas Hipofisárias/biossíntese , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/fisiologia , Animais , Proteína Morfogenética Óssea 2/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/antagonistas & inibidores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Células Cultivadas , Feminino , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Subunidade beta do Hormônio Folículoestimulante/genética , Gonadotrofos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
Elife ; 3: e03245, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-25161195

RESUMO

Obesity is associated with blunted ß-adrenoreceptor (ß-AR)-mediated lipolysis and lipid oxidation in adipose tissue, but the mechanisms linking nutrient overload to catecholamine resistance are poorly understood. We report that targeted disruption of TGF-ß superfamily receptor ALK7 alleviates diet-induced catecholamine resistance in adipose tissue, thereby reducing obesity in mice. Global and fat-specific Alk7 knock-out enhanced adipose ß-AR expression, ß-adrenergic signaling, mitochondrial biogenesis, lipid oxidation, and lipolysis under a high fat diet, leading to elevated energy expenditure, decreased fat mass, and resistance to diet-induced obesity. Conversely, activation of ALK7 reduced ß-AR-mediated signaling and lipolysis cell-autonomously in both mouse and human adipocytes. Acute inhibition of ALK7 in adult mice by a chemical-genetic approach reduced diet-induced weight gain, fat accumulation, and adipocyte size, and enhanced adipocyte lipolysis and ß-adrenergic signaling. We propose that ALK7 signaling contributes to diet-induced catecholamine resistance in adipose tissue, and suggest that ALK7 inhibitors may have therapeutic value in human obesity.


Assuntos
Receptores de Ativinas Tipo I/genética , Tecido Adiposo/metabolismo , Obesidade/genética , Receptores Adrenérgicos beta/genética , Receptores de Ativinas Tipo I/antagonistas & inibidores , Receptores de Ativinas Tipo I/deficiência , Trifosfato de Adenosina/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Adipócitos/metabolismo , Adipócitos/patologia , Tecido Adiposo/patologia , Animais , Catecolaminas/metabolismo , Dieta Hiperlipídica , Gorduras na Dieta/efeitos adversos , Regulação da Expressão Gênica , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Lipólise/efeitos dos fármacos , Camundongos , Camundongos Knockout , Renovação Mitocondrial/efeitos dos fármacos , Obesidade/etiologia , Obesidade/patologia , Obesidade/prevenção & controle , Cultura Primária de Células , Pirazóis/farmacologia , Pirimidinas/farmacologia , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 33(2): 305-10, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23241407

RESUMO

OBJECTIVE: Vessels in brain arteriovenous malformations are prone to rupture. The underlying pathogenesis is not clear. Hereditary hemorrhagic telangiectasia type 2 patients with activin receptor-like kinase 1 (Alk1) mutation have a higher incidence of brain arteriovenous malformation than the general population. We tested the hypothesis that vascular endothelial growth factor impairs vascular integrity in the Alk1-deficient brain through reduction of mural cell coverage. METHODS AND RESULTS: Adult Alk1(1f/2f) mice (loxP sites flanking exons 4-6) and wild-type mice were injected with 2×10(7) PFU adenovious-cre recombinase and 2×10(9) genome copies of adeno-associated virus-vascular endothelial growth factor to induce focal homozygous Alk1 deletion (in Alk1(1f/2f) mice) and angiogenesis. Brain vessels were analyzed 8 weeks later. Compared with wild-type mice, the Alk1-deficient brain had more fibrin (99±30×10(3) pixels/mm(2) versus 40±13×10(3); P=0.001), iron deposition (508±506 pixels/mm(2) versus 6±49; P=0.04), and Iba1(+) microglia/macrophage infiltration (888±420 Iba1(+) cells/mm(2) versus 240±104 Iba1(+); P=0.001) after vascular endothelial growth factor stimulation. In the angiogenic foci, the Alk1-deficient brain had more α-smooth muscle actin negative vessels (52±9% versus 12±7%, P<0.001), fewer vascular-associated pericytes (503±179/mm(2) versus 931±115, P<0.001), and reduced platelet-derived growth factor receptor-ß expression. CONCLUSIONS: Reduction of mural cell coverage in response to vascular endothelial growth factor stimulation is a potential mechanism for the impairment of vessel wall integrity in hereditary hemorrhagic telangiectasia type 2-associated brain arteriovenous malformation.


Assuntos
Receptores de Ativinas Tipo I/deficiência , Vasos Sanguíneos/enzimologia , Encéfalo/irrigação sanguínea , Neovascularização Patológica , Pericitos/enzimologia , Telangiectasia Hemorrágica Hereditária/enzimologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Actinas/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Becaplermina , Vasos Sanguíneos/patologia , Dependovirus/genética , Modelos Animais de Doenças , Fibrina/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Ferro/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Microglia/patologia , Pericitos/patologia , Proteínas Proto-Oncogênicas c-sis/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Telangiectasia Hemorrágica Hereditária/genética , Telangiectasia Hemorrágica Hereditária/patologia , Fator A de Crescimento do Endotélio Vascular/genética
11.
Am J Physiol Heart Circ Physiol ; 295(6): H2250-6, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18835925

RESUMO

Loss-of-function activin receptor-like kinase 1 gene mutation (ALK1+/-) is associated with brain arteriovenous malformations (AVM) in hereditary hemorrhagic telangiectasia type 2. Other determinants of the lesional phenotype are unknown. In the present study, we investigated the influence of high vascular flow rates on ALK1+/- mice by manipulating cerebral blood flow (CBF) using vasodilators. Adult male ALK1+/- mice underwent adeno-associated viral-mediated vascular endothelial growth factor (AAVVEGF) or lacZ (AAVlacZ as a control) gene transfer into the brain. Two weeks after vector injection, hydralazine or nicardipine was infused intraventricularly for another 14 days. CBF was measured to evaluate relative tissue perfusion. We analyzed the number and morphology of capillaries. Results demonstrated that hydralazine or nicardipine infusion increased focal brain perfusion in all mice. It was noted that focal CBF increased most in AAVVEGF-injected ALK1+/- mice following hydralazine or nicardipine infusion (145+/-23% or 150+/-11%; P<0.05). There were more detectable dilated and dysplastic capillaries (2.4+/-0.3 or 2.0+/-0.4 dysplasia index; P<0.01) in the brains of ALK1+/- mice treated with AAVVEGF and hydralazine or nicardipine compared with the mice treated with them individually. We concluded that increased focal tissue perfusion and angiogenic factor VEGF stimulation could have a synergistic effect to promote capillary dysplasia in a genetic deficit animal model, which may have relevance to further studies of AVMs.


Assuntos
Receptores de Ativinas Tipo I/deficiência , Encéfalo/irrigação sanguínea , Circulação Cerebrovascular , Malformações Arteriovenosas Intracranianas/fisiopatologia , Microcirculação , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Capilares/enzimologia , Capilares/patologia , Capilares/fisiopatologia , Circulação Cerebrovascular/efeitos dos fármacos , Dependovirus/genética , Modelos Animais de Doenças , Vetores Genéticos , Hidralazina/administração & dosagem , Imageamento Tridimensional , Infusões Parenterais , Malformações Arteriovenosas Intracranianas/enzimologia , Malformações Arteriovenosas Intracranianas/patologia , Masculino , Camundongos , Camundongos Knockout , Microcirculação/efeitos dos fármacos , Microscopia Confocal , Neovascularização Fisiológica , Nicardipino/administração & dosagem , Transdução Genética , Fator A de Crescimento do Endotélio Vascular/genética , Vasodilatadores/administração & dosagem
12.
Cell Cycle ; 6(11): 1360-6, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17534148

RESUMO

We generated a mouse model with a conditional deletion of TGF-beta signaling in the neurons by crossing TGF-beta receptor I (TbetaRI) floxed mice with neurofilament-H (NF-H) Cre mice. 35% of F1 conditional knockout (COKO) mice developed spontaneous squamous cell carcinomas (SCCs) in periorbital and/or perianal regions. Transplantation of these tumors into athymic nude mice resulted in 62% tumorigenicity. To determine whether evasion of the immune response plays any role in this tumorigenesis, we analyzed the expression levels of receptors for interleukin-13 (mIL-13R), a key negative regulator of tumor immunosurveillance, and found that 33% of COKO tumors expressed the IL-13R alpha2 chain. Primary cultures of the SCCs expressing IL-13R alpha2 were sensitive to the cytotoxic effect of IL-13R-directed cytotoxin treatment. This is the first demonstration that loss of TbetaRI can lead to spontaneous tumor formation. These mice can serve as a unique mouse model of SCC to evaluate the tumorigenicity and effect of anti-cancer therapeutics.


Assuntos
Receptores de Ativinas Tipo I/fisiologia , Neoplasias do Ânus/genética , Carcinoma de Células Escamosas/genética , Neoplasias de Cabeça e Pescoço/genética , Síndromes Neoplásicas Hereditárias/genética , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Animais , Antineoplásicos/farmacologia , Neoplasias do Ânus/imunologia , Neoplasias do Ânus/metabolismo , Encéfalo/embriologia , Encéfalo/metabolismo , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Cruzamentos Genéticos , Ensaios de Seleção de Medicamentos Antitumorais , Exotoxinas/farmacologia , Feminino , Genes Sintéticos , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/metabolismo , Integrases/genética , Interleucina-13/farmacologia , Subunidade alfa2 de Receptor de Interleucina-13/biossíntese , Subunidade alfa2 de Receptor de Interleucina-13/genética , Masculino , Camundongos , Camundongos Knockout , Camundongos Nus , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Proteínas de Neurofilamentos/genética , Especificidade de Órgãos , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteínas Recombinantes de Fusão/farmacologia
13.
Mol Cell ; 12(4): 817-28, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14580334

RESUMO

Transforming growth factor-beta (TGFbeta) regulates the activation state of the endothelium via two opposing type I receptor/Smad pathways. Activin receptor-like kinase-1 (ALK1) induces Smad1/5 phosphorylation, leading to an increase in endothelial cell proliferation and migration, while ALK5 promotes Smad2/3 activation and inhibits both processes. Here, we report that ALK5 is important for TGFbeta/ALK1 signaling; endothelial cells lacking ALK5 are deficient in TGFbeta/ALK1-induced responses. More specifically, we show that ALK5 mediates a TGFbeta-dependent recruitment of ALK1 into a TGFbeta receptor complex and that the ALK5 kinase activity is required for optimal ALK1 activation. TGFbeta type II receptor is also required for ALK1 activation by TGFbeta. Interestingly, ALK1 not only induces a biological response opposite to that of ALK5 but also directly antagonizes ALK5/Smad signaling.


Assuntos
Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/metabolismo , Células Endoteliais/enzimologia , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Proteínas Repressoras , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Células COS , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Humanos , Proteína 1 Inibidora de Diferenciação , Substâncias Macromoleculares , Mutação/genética , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Smad , Proteína Smad1 , Proteína Smad5 , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
14.
Dev Biol ; 261(1): 235-50, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12941632

RESUMO

Several characteristic morphological and functional differences distinguish arteries from veins. It was thought that hemodynamic forces shaped these differences; however, increasing evidence suggests that morphogenetic programs play a central role in blood vessel differentiation. Hereditary hemorrhagic telangiectasia (HHT) is a vascular dysplasia characterized by the inappropriate fusion of arterioles with venules. The genes implicated in this disease, ALK1 and endoglin, may be involved in defining the fundamental boundaries between arteries and veins. We previously showed that mice lacking Alk1 lost structural, molecular, and functional distinctions between arteries and veins. Here, we report that mice lacking endoglin develop arterial-venous malformations and fail to confine intraembryonic hematopoiesis to arteries. In contrast to Alk1 mutants, endoglin mutants do not show profound vessel dilation or downregulation of arterial ephrinB2. Finally, our data indicate that a failure in cardiac cushion formation observed in both strains may be secondary to the peripheral vasculature defect. The phenotypic similarities, yet reduced severity, implicates endoglin as an accessory coreceptor that specifically modulates Alk1 signaling. We propose that endoglin and Alk1 are necessary for the maintenance of distinct arterial-venous vascular beds and that attenuation of the Alk1 signaling pathway is the precipitating event in the etiology of HHT.


Assuntos
Receptores de Ativinas Tipo I/fisiologia , Vasos Sanguíneos/embriologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Molécula 1 de Adesão de Célula Vascular/fisiologia , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Antígenos CD , Artérias/embriologia , Malformações Arteriovenosas/embriologia , Malformações Arteriovenosas/genética , Sequência de Bases , DNA/genética , Comunicação Atrioventricular/embriologia , Comunicação Atrioventricular/genética , Endoglina , Efrina-B2/genética , Hematopoese/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Receptores de Superfície Celular , Telangiectasia Hemorrágica Hereditária/embriologia , Telangiectasia Hemorrágica Hereditária/genética , Molécula 1 de Adesão de Célula Vascular/genética , Veias/embriologia
15.
Blood ; 102(9): 3129-35, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12842983

RESUMO

Studies in vitro implicate transforming growth factor beta (TGF-beta) as a key regulator of hematopoiesis with potent inhibitory effects on progenitor and stem cell proliferation. In vivo studies have been hampered by early lethality of knock-out mice for TGF-beta isoforms and the receptors. To directly assess the role of TGF-beta signaling for hematopoiesis and hematopoietic stem cell (HSC) function in vivo, we generated a conditional knock-out model in which a disruption of the TGF-beta type I receptor (T beta RI) gene was induced in adult mice. HSCs from induced mice showed increased proliferation recruitment when cultured as single cells under low stimulatory conditions in vitro, consistent with an inhibitory role of TGF-beta in HSC proliferation. However, induced T beta RI null mice show normal in vivo hematopoiesis with normal numbers and differentiation ability of hematopoietic progenitor cells. Furthermore HSCs from T beta RI null mice exhibit a normal cell cycle distribution and do not differ in their ability long term to repopulate primary and secondary recipient mice following bone marrow transplantation. These findings challenge the classical view that TGF-beta is an essential negative regulator of hematopoietic stem cells under physiologic conditions in vivo.


Assuntos
Receptores de Ativinas Tipo I/fisiologia , Células-Tronco Hematopoéticas/citologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Animais , Transplante de Medula Óssea , Ciclo Celular , Diferenciação Celular , Divisão Celular , Células Cultivadas , Hematopoese , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA