Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Bone Miner Res ; 39(4): 382-398, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38477818

RESUMO

Single case studies of extraordinary disease resilience may provide therapeutic insight into conditions for which no definitive treatments exist. An otherwise healthy 35-year-old man (patient-R) with the canonical pathogenic ACVR1R206H variant and the classic congenital great toe malformation of fibrodysplasia ossificans progressiva (FOP) had extreme paucity of post-natal heterotopic ossification (HO) and nearly normal mobility. We hypothesized that patient-R lacked a sufficient post-natal inflammatory trigger for HO. A plasma biomarker survey revealed a reduction in total matrix metalloproteinase-9 (MMP-9) compared to healthy controls and individuals with quiescent FOP. Whole exome sequencing identified compound heterozygous variants in MMP-9 (c.59C > T, p.A20V and c.493G > A, p.D165N). Structural analysis of the D165N variant predicted both decreased MMP-9 secretion and activity that were confirmed by enzyme-linked immunosorbent assay and gelatin zymography. Further, human proinflammatory M1-like macrophages expressing either MMP-9 variant produced significantly less Activin A, an obligate ligand for HO in FOP, compared to wildtype controls. Importantly, MMP-9 inhibition by genetic, biologic, or pharmacologic means in multiple FOP mouse models abrogated trauma-induced HO, sequestered Activin A in the extracellular matrix (ECM), and induced regeneration of injured skeletal muscle. Our data suggest that MMP-9 is a druggable node linking inflammation to HO, orchestrates an existential role in the pathogenesis of FOP, and illustrates that a single patient's clinical phenotype can reveal critical molecular mechanisms of disease that unveil novel treatment strategies.


A healthy 35-year-old man (patient-R) with the classic fibrodysplasia ossificans progressiva (FOP) mutation and the congenital great toe malformation of FOP had extreme lack of heterotopic ossification (HO) and nearly normal mobility. We hypothesized that patient-R lacked a sufficient inflammatory trigger for HO. Blood tests revealed a reduction in the level of an inflammatory protein called matrix metalloproteinase-9 (MMP-9) compared to other individuals with FOP as well as healthy controls. DNA analysis in patient-R identified mutations in MMP-9, one of which predicted decreased activity of MMP-9 which was confirmed by further testing. Inflammatory cells (macrophages) expressing the MMP-9 mutations identified in patient-R produced significantly less Activin A, an obligate stimulus for HO in FOP. In order to determine if MMP-9 deficiency was a cause of HO prevention in FOP, we inhibited MMP-9 activity by genetic, biologic, or pharmacologic means in FOP mouse models and showed that MMP-9 inhibition prevented or dramatically decreased trauma-induced HO in FOP, locked-up Activin A in the extracellular matrix, and induced regeneration of injured skeletal muscle. Our data show that MMP-9 links inflammation to HO and illustrate that one patient's clinical picture can reveal critical molecular mechanisms of disease that unveil new treatment strategies.


Assuntos
Receptores de Ativinas Tipo I , Metaloproteinase 9 da Matriz , Miosite Ossificante , Adulto , Animais , Humanos , Masculino , Camundongos , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Receptores de Ativinas Tipo I/deficiência , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Miosite Ossificante/genética , Miosite Ossificante/patologia , Miosite Ossificante/metabolismo , Ossificação Heterotópica/patologia , Ossificação Heterotópica/genética , Ossificação Heterotópica/metabolismo
2.
FASEB J ; 35(8): e21759, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34245608

RESUMO

Life-style change and anti-inflammatory interventions have only transient effects in obesity. It is not clear how benefits obtained by these treatments can be maintained longer term, especially during sustained high caloric intake. Constitutive ablation of the activin receptor ALK7 in adipose tissue enhances catecholamine signaling and lipolysis in adipocytes, and protects mice from diet-induced obesity. Here, we investigated the consequences of conditional ALK7 ablation in adipocytes of adult mice with pre-existing obesity. Although ALK7 deletion had little effect on its own, it synergized strongly with a transient switch to low-fat diet (life-style change) or anti-inflammatory treatment (Na-salicylate), resulting in enhanced lipolysis, increased energy expenditure, and reduced adipose tissue mass and body weight gain, even under sustained high caloric intake. By themselves, diet-switch and salicylate had only a temporary effect on weight gain. Mechanistically, combination of ALK7 ablation with either treatment strongly enhanced the levels of ß3-AR, the main adrenergic receptor for catecholamine stimulation of lipolysis, and C/EBPα, an upstream regulator of ß3-AR expression. These results suggest that inhibition of ALK7 can be combined with simple interventions to produce longer-lasting benefits in obesity.


Assuntos
Receptores de Ativinas Tipo I/deficiência , Adipócitos/metabolismo , Ingestão de Alimentos , Lipólise , Obesidade/metabolismo , Receptores de Ativinas Tipo I/metabolismo , Adipócitos/patologia , Animais , Camundongos , Camundongos Transgênicos , Obesidade/genética , Obesidade/patologia , Salicilatos/farmacologia
3.
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
4.
Clin Exp Pharmacol Physiol ; 47(2): 313-321, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31608496

RESUMO

Diabetic retinopathy (DR) is one of the diabetic complications associated with hyperglycaemia-mediated oxidative stress. Activin receptor-like kinase 7 (ALK7) has been proven to be a potential therapeutic approach for diabetic cardiomyopathy, which is another diabetic complication. However, the role of ALK7 in DR remains unclear. In the current study, ALK7 was found to be up-regulated in clinical samples from DR patients and high glucose (HG)-induced human retinal pigment epithelial cells (ARPE-19). In vitro studies demonstrated that knockdown of ALK7 in ARPE-19 cells through transfection with siRNA-ALK7 (si-ALK7) improved cell viability in HG-induced ARPE-19 cells. Knockdown of ALK7 suppressed HG-induced reactive oxygen species (ROS) production, as well elevating the activities of superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) in ARPE-19 cells. The number of apoptotic cells was significantly decreased after transfection with si-ALK7. ALK7 knockdown also caused a significant decrease in bax expression and an increase in bcl-2 expression in HG-induced ARPE-19 cells. In addition, ALK7 knockdown resulted in remarkable increase in the expressions of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and heme oxygenase-1 (HO-1) in ARPE-19 cells in response to HG induction. Taken together, knockdown of ALK7 protected ARPE-19 cells from HG-induced oxidative injury, which might be mediated by the activation of the Nrf2/HO-1 signalling pathway.


Assuntos
Receptores de Ativinas Tipo I/deficiência , Apoptose/efeitos dos fármacos , Glucose/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Apoptose/fisiologia , Linhagem Celular , Sobrevivência Celular , Técnicas de Silenciamento de Genes/métodos , Humanos , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo
5.
Dev Dyn ; 248(5): 396-403, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30854720

RESUMO

PURPOSE: This study analyzes Prx1-specific conditional knockout of Acvr1 aiming to elucidate the endogenous role of Acvr1 during limb formation in early embryonic development. ACVR1 can exhibit activating and inhibiting function in BMP signaling. ACVR1 gain-of-function mutations can cause the rare disease fibrodysplasia ossificans progressiva (FOP), where patients develop ectopic bone replacing soft tissue, tendons and ligaments. METHODS: Whole-mount in situ hybridization and skeletal preparations revealed that following limb-specific conditional knockout of Acvr1, metacarpals and proximal phalanges were shortened and additional cartilage and bone elements were formed. RESULTS: The analysis of a set of marker genes including ligands and receptors of BMP signaling as well as genes involved in patterning and tendon and cartilage formation, revealed temporal disturbances with distinct spatial patterns. The most striking result was that in the absence of Acvr1 in mesoderm precursor cells, first digits were drastically malformed. CONCLUSION: In FOP, malformation of big toes can serve as a first soft marker in diagnostics. The surprising similarities in phenotype between the described conditional knockout of Acvr1 and the FOP mouse model, indicates a natural inhibitory function of ACVR1. This represents a further step towards better understanding the role of Acvr1 and developing treatment options for FOP.


Assuntos
Receptores de Ativinas Tipo I/genética , Desenvolvimento Embrionário/genética , Extremidades/embriologia , Miosite Ossificante/etiologia , Dedos do Pé/anormalidades , Receptores de Ativinas Tipo I/deficiência , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Embrião de Mamíferos , Humanos , Camundongos , Camundongos Knockout , Transdução de Sinais
6.
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
7.
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
8.
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
9.
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
10.
Arterioscler Thromb Vasc Biol ; 37(4): 657-663, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28232325

RESUMO

OBJECTIVE: Increasing evidence suggests that bone morphogenetic protein (BMP) signaling regulates angiogenesis. Here, we aimed to define the function of BMP receptors in regulating early postnatal angiogenesis by analysis of inducible, endothelial-specific deletion of the BMP receptor components Bmpr2 (BMP type 2 receptor), Alk1 (activin receptor-like kinase 1), Alk2, and Alk3 in mouse retinal vessels. APPROACH AND RESULTS: Expression analysis of several BMP ligands showed that proangiogenic BMP ligands are highly expressed in postnatal retinas. Consistently, BMP receptors are also strongly expressed in retina with a distinct pattern. To assess the function of BMP signaling in retinal angiogenesis, we first generated mice carrying an endothelial-specific inducible deletion of Bmpr2. Postnatal deletion of Bmpr2 in endothelial cells substantially decreased the number of angiogenic sprouts at the vascular front and branch points behind the front, leading to attenuated radial expansion. To identify critical BMPR1s (BMP type 1 receptors) associated with BMPR2 in retinal angiogenesis, we generated endothelial-specific inducible deletion of 3 BMPR1s abundantly expressed in endothelial cells and analyzed the respective phenotypes. Among these, endothelial-specific deletion of either Alk2/acvr1 or Alk3/Bmpr1a caused a delay in radial expansion, reminiscent of vascular defects associated with postnatal endothelial-specific deletion of BMPR2, suggesting that ALK2/ACVR1 and ALK3/BMPR1A are likely to be the critical BMPR1s necessary for proangiogenic BMP signaling in retinal vessels. CONCLUSIONS: Our data identify BMP signaling mediated by coordination of ALK2/ACVR1, ALK3/BMPR1A, and BMPR2 as an essential proangiogenic cue for retinal vessels.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Células Endoteliais/efeitos dos fármacos , Artéria Retiniana/efeitos dos fármacos , Neovascularização Retiniana , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Ligantes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Artéria Retiniana/metabolismo , Transdução de Sinais
11.
J Mol Cell Cardiol ; 105: 1-11, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28214509

RESUMO

Cardiac fibrosis (CF), a repairing process following myocardial infarction (MI), is characterized by abnormal proliferation of cardiac fibroblasts and excessive deposition of extracellular matrix (ECM) resulting in inevitable resultant heart failure. TGF-ß (transforming growth factor-ß)/ALK5 (Activin receptor-like kinase 5)/Smad2/3/4 pathways have been reported to be involved in the process. Recent studies have implicated both activin and its specific downstream component ALK4 in stimulating fibrosis in non-cardiac organs. We recently reported that ALK4 is upregulated in the pressure-overloaded heart and its partial inhibition attenuated the pressure overload-induced CF and cardiac dysfunction. However, the role of ALK4 in the pathogenesis of MI-induced CF, which is usually more severe than that induced by pressure-overload, remains unknown. Here we report: 1) In a wild-type mouse model of MI, ALK4 upregulation was restricted in the fibroblasts of the infarct border zone; 2) In contrast, ALK4+/- mice with a haplodeficiency of ALK4 gene, showed a significantly attenuated CF in the border zone, with a smaller scar size, a preserved cardiac function and an improved survival rate post-MI; 3) Similarly to pressure-overloaded heart, these beneficial effects might be through a partial inactivation of the Smad3/4 pathway but not MAPK cascades; 4) The apoptotic rate of the cardiomyocytes were indistinguishable in the border zone of the wild-type control and ALK4+/- mice; 5) Cardiac fibroblasts isolated from ALK4+/- mice showed reduced migration, proliferation and ECM synthesis in response to hypoxia. These results indicate that partial inhibition of ALK4 may reduce MI-induced CF, suggesting ALK4 as a novel target for inhibition of unfavorable CF and for preservation of LV systolic function induced by not only pressure-overload but also MI.


Assuntos
Receptores de Ativinas Tipo I/genética , Haploinsuficiência , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/patologia , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/metabolismo , Animais , Movimento Celular/genética , Proliferação de Células , Modelos Animais de Doenças , Ecocardiografia , Matriz Extracelular , Fibrose , Regulação da Expressão Gênica , Genótipo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Mortalidade , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Miofibroblastos/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo , Proteína Smad4/metabolismo , Função Ventricular
12.
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
13.
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
14.
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
15.
PLoS One ; 11(2): e0149205, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26882027

RESUMO

We aimed to investigate the role of activin receptor-like kinase (ALK7) in regulating cardiac electrophysiology. Here, we showed that Alk7-/- mice exhibited prolonged QT intervals in telemetry ECG recordings. Furthermore, Langendorff-perfused Alk7-/- hearts had significantly longer action potential duration (APD) and greater incidence of ventricular arrhythmia (AV) induced by burst pacing. Using whole-cell patch clamp, we found that the densities of repolarizing K+ currents Ito and IK1 were profoundly reduced in Alk7-/- ventricular cardiomyocytes. Mechanistically, the expression of Kv4.2 (a major subunit of Ito carrying channel) and KCHIP2 (a key accessory subunit of Ito carrying channel), was markedly decreased in Alk7-/- hearts. These findings suggest that endogenous expression of ALK7 is necessary to maintain repolarizing K+ currents in ventricular cardiomyocytes, and finally prevent action potential prolongation and ventricular arrhythmia.


Assuntos
Potenciais de Ação , Receptores de Ativinas Tipo I/deficiência , Arritmias Cardíacas/fisiopatologia , Ventrículos do Coração/fisiopatologia , Receptores de Ativinas Tipo I/metabolismo , Animais , Arritmias Cardíacas/diagnóstico por imagem , Arritmias Cardíacas/metabolismo , Separação Celular , Suscetibilidade a Doenças , Eletrocardiografia , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/patologia , Técnicas In Vitro , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Perfusão , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Subunidades Proteicas/metabolismo , Telemetria , Ultrassonografia
16.
Am J Physiol Heart Circ Physiol ; 310(8): H984-94, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26873969

RESUMO

Bone morphogenetic protein (BMP) signaling contributes to the development of cardiac hypertrophy. However, the identity of the BMP type I receptor involved in cardiac hypertrophy and the underlying molecular mechanisms are poorly understood. By using quantitative PCR and immunoblotting, we demonstrated that BMP signaling increased during phenylephrine-induced hypertrophy in cultured neonatal rat cardiomyocytes (NRCs), as evidenced by increased phosphorylation of Smads 1 and 5 and induction of Id1 gene expression. Inhibition of BMP signaling with LDN193189 or noggin, and silencing of Smad 1 or 4 using small interfering RNA diminished the ability of phenylephrine to induce hypertrophy in NRCs. Conversely, activation of BMP signaling with BMP2 or BMP4 induced hypertrophy in NRCs. Luciferase reporter assay further showed that BMP2 or BMP4 treatment of NRCs repressed atrogin-1 gene expression concomitant with an increase in calcineurin protein levels and enhanced activity of nuclear factor of activated T cells, providing a mechanism by which BMP signaling contributes to cardiac hypertrophy. In a model of cardiac hypertrophy, C57BL/6 mice treated with angiotensin II (A2) had increased BMP signaling in the left ventricle. Treatment with LDN193189 attenuated A2-induced cardiac hypertrophy and collagen deposition in left ventricles. Cardiomyocyte-specific deletion of BMP type I receptor ALK2 (activin-like kinase 2), but not ALK1 or ALK3, inhibited BMP signaling and mitigated A2-induced cardiac hypertrophy and left ventricular fibrosis in mice. The results suggest that BMP signaling upregulates the calcineurin/nuclear factor of activated T cell pathway via BMP type I receptor ALK2, contributing to cardiac hypertrophy and fibrosis.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Angiotensina II , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 4/farmacologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Cardiomegalia/enzimologia , Miócitos Cardíacos/enzimologia , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/prevenção & controle , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fibrose , Proteína 1 Inibidora de Diferenciação/genética , Proteína 1 Inibidora de Diferenciação/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fatores de Transcrição NFATC/metabolismo , Fenilefrina/farmacologia , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , Interferência de RNA , Ratos Sprague-Dawley , Transdução de Sinais , Proteínas Smad/genética , Proteínas Smad/metabolismo , Fatores de Tempo , Transfecção
17.
Dis Model Mech ; 8(11): 1427-39, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26398936

RESUMO

The activin receptor-like kinase 1 (ALK-1) is a type I cell-surface receptor for the transforming growth factor-ß (TGF-ß) family of proteins. Hypertension is related to TGF-ß1, because increased TGF-ß1 expression is correlated with an elevation in arterial pressure (AP) and TGF-ß expression is upregulated by the renin-angiotensin-aldosterone system. The purpose of this study was to assess the role of ALK-1 in regulation of AP using Alk1 haploinsufficient mice (Alk1(+/-)). We observed that systolic and diastolic AP were significantly higher in Alk1(+/-) than in Alk1(+/+) mice, and all functional and structural cardiac parameters (echocardiography and electrocardiography) were similar in both groups. Alk1(+/-) mice showed alterations in the circadian rhythm of AP, with higher AP than Alk1(+/+) mice during most of the light period. Higher AP in Alk1(+/-) mice is not a result of a reduction in the NO-dependent vasodilator response or of overactivation of the peripheral renin-angiotensin system. However, intracerebroventricular administration of losartan had a hypotensive effect in Alk1(+/-) and not in Alk1(+/+) mice. Alk1(+/-) mice showed a greater hypotensive response to the ß-adrenergic antagonist atenolol and higher concentrations of epinephrine and norepinephrine in plasma than Alk1(+/+) mice. The number of brain cholinergic neurons in the anterior basal forebrain was reduced in Alk1(+/-) mice. Thus, we concluded that the ALK-1 receptor is involved in the control of AP, and the high AP of Alk1(+/-) mice is explained mainly by the sympathetic overactivation shown by these animals, which is probably related to the decreased number of cholinergic neurons.


Assuntos
Receptores de Ativinas Tipo I/deficiência , Pressão Arterial , Heterozigoto , Hipertensão/enzimologia , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Pressão Arterial/efeitos dos fármacos , Pressão Arterial/genética , Sistema Nervoso Central/patologia , Sistema Nervoso Central/fisiopatologia , Neurônios Colinérgicos/patologia , Ritmo Circadiano , Relação Dose-Resposta a Droga , Predisposição Genética para Doença , Haploinsuficiência , Hipertensão/tratamento farmacológico , Hipertensão/genética , Hipertensão/patologia , Hipertensão/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Nervoso Simpático/enzimologia , Sistema Nervoso Simpático/fisiopatologia , Fatores de Tempo , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
18.
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
19.
Arterioscler Thromb Vasc Biol ; 34(10): 2232-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25082229

RESUMO

OBJECTIVE: Hereditary hemorrhagic telangiectasia is a genetic disorder characterized by visceral and mucocutaneous arteriovenous malformations (AVMs). Clinically indistinguishable hereditary hemorrhagic telangiectasia 1 and hereditary hemorrhagic telangiectasia 2 are caused by mutations in ENG and ALK1, respectively. In this study, we have compared the development of visceral and mucocutaneous AVMs in adult stages between Eng- and Alk1-inducible knockout (iKO) models. APPROACH AND RESULTS: Eng or Alk1 were deleted from either vascular endothelial cells (ECs) or smooth muscle cells in adult stages using Scl-CreER and Myh11-CreER lines, respectively. Latex perfusion and intravital spectral imaging in a dorsal skinfold window chamber system were used to visualize remodeling vasculature during AVM formation. Global Eng deletion resulted in lethality with visceral AVMs and wound-induced skin AVMs. Deletion of Alk1 or Eng in ECs, but not in smooth muscle cells, resulted in wound-induced skin AVMs. Visceral AVMs were observed in EC-specific Alk1-iKO but not in Eng-iKO. Intravital spectral imaging revealed that Eng-iKO model exhibited more dynamic processes for AVM development when compared with Alk1-iKO model. CONCLUSIONS: Both Alk1- and Eng-deficient models require a secondary insult, such as wounding, and ECs are the primary cell type responsible for the pathogenesis. However, Alk1 but not Eng deletion in ECs results in visceral AVMs.


Assuntos
Malformações Arteriovenosas/patologia , Telangiectasia Hemorrágica Hereditária/patologia , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Malformações Arteriovenosas/genética , Malformações Arteriovenosas/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Endoglina , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Telangiectasia Hemorrágica Hereditária/genética , Telangiectasia Hemorrágica Hereditária/metabolismo , Fatores de Tempo , Cicatrização
20.
Kidney Int ; 85(2): 319-32, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23945497

RESUMO

Tubulointerstitial fibrosis is characterized by an accumulation of extracellular matrix in the renal interstitium, myofibroblast activation, cell infiltration, and tubular cell apoptosis, leading to chronic renal failure. Activin receptor-like kinase 1 (ALK1) is a transforming growth factor-ß1 type I receptor with a pivotal role in endothelial proliferation and migration, but its role in the development of renal fibrosis is unknown. To assess this we used the unilateral ureteral obstruction model of tubulointerstitial fibrosis in ALK1 haploinsufficient (ALK1(+/-)) and wild-type mice. After 15 days, there was an increase in extracellular matrix protein expression in the obstructed kidneys from both ALK1(+/+) and ALK1(+/-) mice, but obstructed kidneys from ALK1(+/-) mice showed significantly higher expression of type I collagen than those from wild-type mice. Ureteral obstruction increased kidney myofibroblasts markers (α-smooth muscle actin and S100A4), without differences between mouse genotypes. ALK1 expression was increased after ureteral obstruction, and this increased expression was located in myofibroblasts. Moreover, cultured renal fibroblasts from ALK1(+/-) mice expressed more collagen type I and fibronectin than fibroblasts derived from wild-type mice. Thus, ALK1 modulates obstruction-induced renal fibrosis by increased extracellular matrix synthesis in myofibroblasts, but without differences in myofibroblast number.


Assuntos
Receptores de Ativinas Tipo I/deficiência , Heterozigoto , Nefropatias/enzimologia , Rim/enzimologia , Obstrução Ureteral/complicações , Actinas/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II , Animais , Biomarcadores/metabolismo , Proliferação de Células , Células Cultivadas , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Fibrose , Haploinsuficiência , Rim/patologia , Nefropatias/genética , Nefropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miofibroblastos/enzimologia , Miofibroblastos/patologia , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Obstrução Ureteral/enzimologia , Obstrução Ureteral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA