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










Base de dados
Intervalo de ano de publicação
1.
Metabolism ; 105: 154189, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32105664

RESUMO

BACKGROUND: Sprouty (SPRY) proteins play critical roles in controlling cell proliferation, differentiation, and survival by inhibiting receptor tyrosine kinase (RTK)-mediated extracellular signal-regulated kinase (ERK) signaling. Recent studies have demonstrated that SPRY4 negatively regulates angiogenesis and tumor growth. However, whether SPRY4 regulates osteogenic and/or adipogenic differentiation of mesenchymal stem cells remains to be explored. RESULTS: In this study, we investigated the expression pattern of Spry4 and found that its expression was regulated during the differentiation of mouse marrow stromal progenitor cells and increased in the metaphysis of ovariectomized mice. In vitro loss-of-function and gain-of-function studies demonstrated that SPRY4 inhibited osteogenic differentiation and stimulated adipogenic differentiation of progenitor cells. In vivo experiments showed that silencing of Spry4 in the marrow of C57BL/6 mice blocked fat accumulation and promoted osteoblast differentiation in ovariectomized mice. Mechanistic investigations revealed the inhibitory effect of SPRY4 on canonical wingless-type MMTV integration site (Wnt) signaling and ERK pathway. ERK1/2 was shown to interact with low-density lipoprotein receptor-related protein 6 (LRP6) and activate the canonical Wnt signaling pathway. Inactivation of Wnt signaling attenuated the inhibition of adipogenic differentiation and stimulation of osteogenic differentiation by Spry4 small interfering RNA (siRNA). Finally, promoter study revealed that ß-catenin transcriptionally inhibited the expression of Spry4. CONCLUSIONS: Our study for the first time suggests that a novel SPRY4-ERK1/2-Wnt/ß-catenin regulatory loop exists in marrow stromal progenitor cells and plays a key role in cell fate determination. It also highlights the potential of SPRY4 as a novel therapeutic target for the treatment of metabolic bone disorders such as osteoporosis.


Assuntos
Adipogenia/genética , Adipogenia/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Células-Tronco Mesenquimais/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Osteogênese/genética , Osteogênese/fisiologia , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/fisiologia , beta Catenina/genética , beta Catenina/fisiologia , Animais , Medula Óssea/metabolismo , Feminino , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , RNA Interferente Pequeno/farmacologia
2.
J Surg Res ; 235: 44-51, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30691826

RESUMO

BACKGROUND: Calcific aortic valve disease (CAVD) is an age-related and slowly progressive valvular disorder. Overexpression of matrix metalloproteinase 12 (MMP-12) has been found in atherosclerosis, stiffed vascular tissue, and calcified aortic valves. We hypothesized that MMP-12 may induce the pro-osteogenic responses in human aortic valve interstitial cells (AVICs). METHODS: Human AVICs were isolated from normal and calcified aortic valves. Cells were treated with MMP-12. The pro-osteogenic marker Runt-related transcription factor 2 (RUNX-2), bone morphogenetic protein 2 (BMP-2), and alkaline phosphatase (ALP), as well as MMP-12-associated signaling molecules, were analyzed. RESULTS: Human calcified aortic valves expressed significantly higher MMP-12 than normal human aortic valves. MMP-12-induced the expression of RUNX-2, BMP-2, ALP, and calcium deposit formation. Suppression of MMP-12 by its inhibitor decreased the expression of RUNX-2, BMP-2, and ALP. MMP-12-induced osteogenic responses were associated with higher levels of phosphorylation of p38 mitogen-activated protein kinases (MAPK), low density lipoprotein-related protein 6 (LRP-6), and ß-catenin signaling molecules. Calcified aortic valves exhibited markedly higher levels of LRP-6 and ß-catenin levels. Inhibition of either p38 MAPK or LRP-6 attenuated MMP-12-induced expression of RUNX-2, BMP-2, and ALP. Suppression of p38 MAPK abrogated MMP-12-induced activation of LRP-6 and ß-catenin signaling pathways. CONCLUSIONS: MMP-12 induces pro-osteogenic responses in AVICs by activation of p38 MAPK-mediated LRP-6 and ß-catenin signaling pathways. The study revealed that the potential role of MMP-12 in the pathogenesis of CAVD and therapeutically targeting MMP-12 may suppress the development of CAVD.


Assuntos
Valva Aórtica/citologia , Metaloproteinase 12 da Matriz/fisiologia , Osteogênese/fisiologia , Idoso , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Estenose da Valva Aórtica/etiologia , Estenose da Valva Aórtica/metabolismo , Proteína Morfogenética Óssea 2/fisiologia , Calcinose/etiologia , Calcinose/metabolismo , Células Cultivadas , Feminino , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Masculino , Pessoa de Meia-Idade , Transdução de Sinais , beta Catenina/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
3.
Reprod Sci ; 26(1): 35-43, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29439617

RESUMO

Remodeling of maternal spiral artery after the implantation of embryo relies on tightly regulated trophoblast functions and is pivotal to ensure the success of pregnancy. Low-density lipoprotein receptor-related protein 6 (LRP6) is implicated in angiogenesis and several vascular diseases, but its function in trophoblast regulation is still not fully understood. We aimed to investigate the involvement of LRP6 in trophoblast functions during maternal spiral artery remodeling. HTR-8/SVneo cells, a trophoblast cell line, were employed to examine the impact of LRP6 knockdown on proliferation, apoptosis, and migration, as well as invasion and tube formation. Expression of matrix metalloproteinases (MMPs), angiogenic factors placental growth factor (PlGF), and vascular endothelial growth factor (VEGF), as well as activation of canonical Wnt/ß-catenin signaling pathway, which was involved in artery remodeling process, was also analyzed. Lipoprotein receptor-related protein 6 knockdown suppressed proliferation, migration, invasion, and tube formation, as well as induced apoptosis in HTR-8/SVneo cells. In addition, LRP6 knockdown also significantly decreased expression of MMPs, PlGF, and VEGF and activation of Wnt/ß-catenin signaling pathway. Lipoprotein receptor-related protein 6 might positively regulate the remodeling of the maternal artery through both endovascular formation and trophoblast invasion.


Assuntos
Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Trofoblastos/fisiologia , Apoptose , Linhagem Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Humanos , Neovascularização Fisiológica , Transdução de Sinais
4.
J Endod ; 43(9S): S25-S30, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28778505

RESUMO

The aim of this study was to evaluate the effects of Wnt signaling through lipoprotein receptor-related protein 6 (LRP6) and Frizzled6 on the endothelial differentiation of dental pulp stem cells (DPSCs). DPSCs were stably transduced with enhanced green fluorescent protein (EGFP)-tagged lentiviral vectors (short hairpin RNA-LRP6, short hairpin RNA-Frizzled6, or empty vector controls). We evaluated the effects of LRP6 and Frizzled6 on expression of endothelial markers and on capillary tube formation mediated by DPSCs induced with recombinant human Wnt1 (rhWnt1) and/or recombinant human vascular endothelial growth factor165 (rhVEGF165). In vivo, tooth slices/scaffolds were seeded with LRP6-silenced, Frizzled6-silenced, or vector control DPSC cells and transplanted into immunodeficient mice. The density of blood vessels generated by DPSCs differentiated into vascular endothelial cells was analyzed by immunohistochemistry for EGFP. The rhWnt1 and rhVEGF165 induced expression of active ß-catenin in control DPSCs and in Frizzled6-silenced DPSCs, but not in LRP6-silenced DPSCs. Furthermore, VEGF and interleukin-8 were downregulated in LRP6-silenced DPSCs, but not in control DPSCs or in Frizzled6-silenced DPSCs (P < .05). Likewise, rhWnt1 and rhVEGF165 induced expression of the endothelial marker VEGF receptor-2 in control DPSCs and in Frizzled6-silenced DPSCs, but not in LRP6-silenced DPSCs. These data correlated with a trend for lower density of capillary sprouts generated by LRP6-silenced DPSCs when compared with control DPSCs in Matrigel. In vivo, tooth slice/scaffolds seeded with DPSC-short hairpinRNA-LRP6 cells showed lower density of human blood vessels (ie, EGFP-positive blood vessels), when compared with tooth slice/scaffolds seeded with vector control cells (P < .05). Collectively, these data demonstrated that LRP6 signaling is necessary for the vasculogenic differentiation of human DPSCs.


Assuntos
Diferenciação Celular/fisiologia , Polpa Dentária/citologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Transdução de Sinais , Células-Tronco/fisiologia , Animais , Células Cultivadas , Humanos , Camundongos , Fator A de Crescimento do Endotélio Vascular , Via de Sinalização Wnt
5.
FEBS J ; 284(11): 1657-1671, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28425175

RESUMO

Snake venom metalloproteases (SVMPs) are members of the a disintegrin and metalloprotease (ADAM) family of proteins, as they possess similar domains. SVMPs are known to elicit snake venom-induced haemorrhage; however, the target proteins and cleavage sites are not known. In this work, we identified a target protein of vascular apoptosis-inducing protein 1 (VAP1), an SVMP, relevant to its ability to induce haemorrhage. VAP1 disrupted cell-cell adhesions by relocating VE-cadherin and γ-catenin from the cell-cell junction to the cytosol, without inducing proteolysis of VE-cadherin. The Wnt receptors low-density lipoprotein receptor-related proteins 5 and 6 (LRP5/6) are known to promote catenin relocation, and are rendered constitutively active in Wnt signalling by truncation. Thus, we examined whether VAP1 cleaves LRP5/6 to induce catenin relocation. Indeed, we found that VAP1 cleaved the extracellular region of LRP6 and LRP5. This cleavage removes four inhibitory ß-propeller structures, resulting in activation of LRP5/6. Recombinant human ADAM8 and ADAM12 also cleaved LRP6 at the same site. An antibody against a peptide including the LRP6-cleavage site inhibited VAP1-induced VE-cadherin relocation and disruption of cell-cell adhesions in cultured cells, and blocked haemorrhage in mice in vivo. Intriguingly, animals resistant to the effects of haemorrhagic snake venom express variants of LRP5/6 that lack the VAP1-cleavage site, or low-density lipoprotein receptor domain class A domains involved in formation of the constitutively active form. The results validate LRP5/6 as physiological targets of ADAMs. Furthermore, they indicate that SVMP-induced cleavage of LRP5/6 causes disruption of cell-cell adhesion and haemorrhage, potentially opening new avenues for the treatment of snake bites.


Assuntos
Proteínas ADAM/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Venenos de Crotalídeos/metabolismo , Hemorragia/induzido quimicamente , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Metaloendopeptidases/metabolismo , Proteínas ADAM/farmacologia , Proteína ADAM12/metabolismo , Proteína ADAM12/farmacologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/farmacologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Resistência a Medicamentos , Fibrinogênio/metabolismo , Fibronectinas/metabolismo , Células HeLa , Células Endoteliais da Veia Umbilical Humana , Humanos , Interações Hidrofóbicas e Hidrofílicas , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/química , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/química , Masculino , Proteínas de Membrana/metabolismo , Proteínas de Membrana/farmacologia , Camundongos , Modelos Moleculares , Simulação de Acoplamento Molecular , Domínios Proteicos , Estrutura Secundária de Proteína/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Vertebrados/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/fisiologia
6.
J Am Soc Nephrol ; 28(6): 1769-1782, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28130402

RESUMO

Connective tissue growth factor (CTGF), a matrix-associated protein with four distinct cytokine binding domains, has roles in vasculogenesis, wound healing responses, and fibrogenesis and is upregulated in fibroblasts and myofibroblasts in disease. Here, we investigated the role of CTGF in fibrogenic cells. In mice, tissue-specific inducible overexpression of CTGF by kidney pericytes and fibroblasts had no bearing on nephrogenesis or kidney homeostasis but exacerbated inflammation and fibrosis after ureteral obstruction. These effects required the WNT receptor LDL receptor-related protein 6 (LRP6). Additionally, pericytes isolated from these mice became hypermigratory and hyperproliferative on overexpression of CTGF. CTGF is cleaved in vivo into distinct domains. Treatment with recombinant domain 1, 1+2 (N terminus), or 4 (C terminus) independently activated myofibroblast differentiation and wound healing responses in cultured pericytes, but domain 4 showed the broadest profibrotic activity. Domain 4 exhibited low-affinity binding to LRP6 in in vitro binding assays, and inhibition of LRP6 or critical signaling cascades downstream of LRP6, including JNK and WNT/ß-catenin, inhibited the biologic activity of domain 4. Administration of blocking antibodies specifically against CTGF domain 4 or recombinant Dickkopf-related protein-1, an endogenous inhibitor of LRP6, effectively inhibited inflammation and fibrosis associated with ureteral obstruction in vivo Therefore, domain 4 of CTGF and the WNT signaling pathway are important new targets in fibrosis.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/fisiologia , Nefropatias/etiologia , Rim/patologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Animais , Fator de Crescimento do Tecido Conjuntivo/antagonistas & inibidores , Fibroblastos , Fibrose/etiologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pericitos
7.
Cancer Lett ; 389: 41-48, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28043913

RESUMO

Activation of Wnt/ß-catenin signaling is associated with pancreatic and colorectal cancer, among others. To-date, there are no FDA-approved small molecule Wnt/ß-catenin inhibitors and many past efforts resulted in compounds with undesirable off-target effects. We recently identified a series of benzimidazole analogs as potent inhibitors of Wnt/ß-catenin signaling. Here, we show that the lead compound SRI36160 displayed selective Wnt inhibition and potent antiproliferative activity in pancreatic and colorectal cancer cells. Moreover, SRI36160 had no effect on STAT3 and mTORC1 signaling in pancreatic and colorectal cancer cells, and was not effective in inhibiting proliferation of non-cancerous cells. Our findings suggest that this series of benzimidazole analogs presents a novel approach for the treatment of Wnt-dependent cancers such as colorectal and pancreatic cancer.


Assuntos
Benzimidazóis/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Wnt/antagonistas & inibidores , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/antagonistas & inibidores , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Neoplasias Colorretais/patologia , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Camundongos , Mutação , Neoplasias Pancreáticas/patologia , Fosforilação , Proteína Wnt3A/fisiologia , beta Catenina/genética
8.
Oncogene ; 35(33): 4321-34, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-26751775

RESUMO

Most tumors initially respond to cytotoxic treatments, but acquired resistance often follows. The tumor microenvironment (TME) is a major barrier to clinical success by compromising therapeutic efficacy, and pathological relevance of multiple soluble factors released by a therapeutically remodeled TME remains largely unexplored. Here we show that the secreted frizzled-related protein 2 (SFRP2), a Wnt pathway modulator, is produced by human primary fibroblasts after genotoxic treatments. SFRP2 induction is remarkable in tumor stroma, with transcription mainly modulated by the nuclear factor-κB (NF-κB) complex, a property shared by several effectors of the DNA damage secretory program. Instead of directly altering canonical Wnt signaling, SFRP2 augments ß-catenin activities initiated by WNT16B, another soluble factor from DNA-damaged stroma. WNT16B recognizes cancer cell surface receptors including frizzled (FZD) 3/4/6, a process enhanced by SFRP2, coordinated by the co-receptor LRP6 but subject to abrogation by DKK1. Importantly, we found WNT16B plays a central role in promoting advanced malignancies particularly acquired resistance by counteracting cell death, an effect that can be minimized by a neutralizing antibody co-administered with classical chemotherapy. Furthermore, DNA damage-triggered expression of WNT16B is systemic, imaged by significant induction among diverse solid organs and circulation in peripheral blood, thereby holding promise as not only a TME-derived anticancer target but also a novel biomarker for clinical evaluation of treatment efficacy. Overall, our study substantiates the biological complexity and pathological implication of a therapy-activated TME, and provides the proof of principle of co-targeting tumor and the TME to prevent acquired resistance, with the aim of improving intervention outcome in an era of precision medicine.


Assuntos
Proteínas de Membrana/fisiologia , Neoplasias/tratamento farmacológico , Transdução de Sinais/fisiologia , Microambiente Tumoral , Proteínas Wnt/fisiologia , Apoptose , Linhagem Celular Tumoral , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , NF-kappa B/fisiologia , Microambiente Tumoral/fisiologia , Proteínas Wnt/antagonistas & inibidores , beta Catenina/fisiologia
9.
J Am Soc Nephrol ; 27(2): 417-27, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26047795

RESUMO

Hypoplastic and/or cystic kidneys have been found in both LDL receptor-related protein 6 (Lrp6)- and ß-catenin-mutant mouse embryos, and these proteins are key molecules for Wnt signaling. However, the underlying mechanisms of Lrp6/ß-catenin signaling in renal development and cystic formation remain poorly understood. In this study, we found evidence that diminished cell proliferation and increased apoptosis occur before cystic dysplasia in the renal primordia of Lrp6-deficient mouse embryos. The expression of Ret proto-oncogene (Ret), a critical receptor for the growth factor glial cell line-derived neurotrophic factor (GDNF), which is required for early nephrogenesis, was dramatically diminished in the mutant renal primordia. The activities of other representative nephrogenic genes, including Lim1, Pax2, Pax8, GDNF, and Wnt11, were subsequently diminished in the mutant renal primordia. Molecular biology experiments demonstrated that Ret is a novel transcriptional target of Wnt/ß-catenin signaling. Wnt agonist lithium promoted Ret expression in vitro and in vivo. Furthermore, Lrp6-knockdown or lithium treatment in vitro led to downregulation or upregulation, respectively, of the phosphorylated mitogen-activated protein kinases 1 and 3, which act downstream of GDNF/Ret signaling. Mice with single and double mutations of Lrp6 and Ret were perinatal lethal and demonstrated gene dosage-dependent effects on the severity of renal hypoplasia during embryogenesis. Taken together, these results suggest that Lrp6-mediated Wnt/ß-catenin signaling modulates or interacts with a signaling network consisting of Ret cascades and related nephrogenic factors for renal development, and the disruption of these genes or signaling activities may cause a spectrum of hypoplastic and cystic kidney disorders.


Assuntos
Rim/crescimento & desenvolvimento , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Rim Displásico Multicístico/etiologia , Proteínas Proto-Oncogênicas c-ret/fisiologia , Transdução de Sinais , Animais , Camundongos , Camundongos Knockout , Rim Displásico Multicístico/genética
10.
Circ Res ; 117(2): 142-56, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-26034040

RESUMO

RATIONALE: Wnt signaling regulates key aspects of diabetic vascular disease. OBJECTIVE: We generated SM22-Cre;LRP6(fl/fl);LDLR(-/-) mice to determine contributions of Wnt coreceptor low-density lipoprotein receptor-related protein 6 (LRP6) in the vascular smooth muscle lineage of male low-density lipoprotein receptor-null mice, a background susceptible to diet (high-fat diet)-induced diabetic arteriosclerosis. METHODS AND RESULTS: As compared with LRP6(fl/fl);LDLR(-/-) controls, SM22-Cre;LRP6(fl/fl);LDLR(-/-) (LRP6-VKO) siblings exhibited increased aortic calcification on high-fat diet without changes in fasting glucose, lipids, or body composition. Pulse wave velocity (index of arterial stiffness) was also increased. Vascular calcification paralleled enhanced aortic osteochondrogenic programs and circulating osteopontin (OPN), a matricellular regulator of arteriosclerosis. Survey of ligands and Frizzled (Fzd) receptor profiles in LRP6-VKO revealed upregulation of canonical and noncanonical Wnts alongside Fzd10. Fzd10 stimulated noncanonical signaling and OPN promoter activity via an upstream stimulatory factor (USF)-activated cognate inhibited by LRP6. RNA interference revealed that USF1 but not USF2 supports OPN expression in LRP6-VKO vascular smooth muscle lineage, and immunoprecipitation confirmed increased USF1 association with OPN chromatin. ML141, an antagonist of cdc42/Rac1 noncanonical signaling, inhibited USF1 activation, osteochondrogenic programs, alkaline phosphatase, and vascular smooth muscle lineage calcification. Mass spectrometry identified LRP6 binding to protein arginine methyltransferase (PRMT)-1, and nuclear asymmetrical dimethylarginine modification was increased with LRP6-VKO. RNA interference demonstrated that PRMT1 inhibits OPN and TNAP, whereas PRMT4 supports expression. USF1 complexes containing the histone H3 asymmetrically dimethylated on Arg-17 signature of PRMT4 are increased with LRP6-VKO. Jmjd6, a demethylase downregulated with LRP6 deficiency, inhibits OPN and TNAP expression, USF1: histone H3 asymmetrically dimethylated on Arg-17 complex formation, and transactivation. CONCLUSIONS: LRP6 restrains vascular smooth muscle lineage noncanonical signals that promote osteochondrogenic differentiation, mediated in part via USF1- and arginine methylation-dependent relays.


Assuntos
Arteriosclerose/prevenção & controle , Calcinose/prevenção & controle , Diabetes Mellitus Experimental/complicações , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/metabolismo , Receptores de LDL/deficiência , Via de Sinalização Wnt , Animais , Arginina/análogos & derivados , Arginina/metabolismo , Arteriosclerose/etiologia , Arteriosclerose/metabolismo , Calcinose/etiologia , Calcinose/metabolismo , Diabetes Mellitus Experimental/patologia , Gorduras na Dieta/efeitos adversos , Receptores Frizzled/fisiologia , Regulação da Expressão Gênica/fisiologia , Histonas/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Masculino , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/patologia , Osteopontina/biossíntese , Osteopontina/genética , Comunicação Parácrina , Mapeamento de Interação de Proteínas , Proteína-Arginina N-Metiltransferases/metabolismo , Receptores de Superfície Celular , Receptores de LDL/genética , Fatores Estimuladores Upstream/fisiologia , Rigidez Vascular/fisiologia
11.
J Clin Invest ; 124(9): 3825-46, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25083995

RESUMO

Canonical WNT signaling is required for proper vascularization of the CNS during embryonic development. Here, we used mice with targeted mutations in genes encoding canonical WNT pathway members to evaluate the exact contribution of these components in CNS vascular development and in specification of the blood-brain barrier (BBB) and blood-retina barrier (BRB). We determined that vasculature in various CNS regions is differentially sensitive to perturbations in canonical WNT signaling. The closely related WNT signaling coreceptors LDL receptor-related protein 5 (LRP5) and LRP6 had redundant functions in brain vascular development and barrier maintenance; however, loss of LRP5 alone dramatically altered development of the retinal vasculature. The BBB in the cerebellum and pons/interpeduncular nuclei was highly sensitive to decrements in canonical WNT signaling, and WNT signaling was required to maintain plasticity of barrier properties in mature CNS vasculature. Brain and retinal vascular defects resulting from ablation of Norrin/Frizzled4 signaling were ameliorated by stabilizing ß-catenin, while inhibition of ß-catenin-dependent transcription recapitulated the vascular development and barrier defects associated with loss of receptor, coreceptor, or ligand, indicating that Norrin/Frizzled4 signaling acts predominantly through ß-catenin-dependent transcriptional regulation. Together, these data strongly support a model in which identical or nearly identical canonical WNT signaling mechanisms mediate neural tube and retinal vascularization and maintain the BBB and BRB.


Assuntos
Barreira Hematoencefálica/fisiologia , Barreira Hematorretiniana/fisiologia , Neovascularização Fisiológica/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Proteínas do Olho/genética , Receptores Frizzled/fisiologia , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Camundongos , Proteínas do Tecido Nervoso/genética , Retina/fisiologia , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , beta Catenina/fisiologia
12.
J Bone Miner Res ; 29(1): 29-42, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23901037

RESUMO

The bone formation inhibitor sclerostin encoded by SOST binds in vitro to low-density lipoprotein receptor-related protein (LRP) 5/6 Wnt co-receptors, thereby inhibiting Wnt/ß-catenin signaling, a central pathway of skeletal homeostasis. Lrp5/LRP5 deficiency results in osteoporosis-pseudoglioma (OPPG), whereas Sost/SOST deficiency induces lifelong bone gain in mice and humans. Here, we analyzed the bone phenotype of mice lacking Sost (Sost(-/-) ), Lrp5 (Lrp5(-/-) ), or both (Sost(-/-) ;Lrp5(-/-) ) to elucidate the mechanism of action of Sost in vivo. Sost deficiency-induced bone gain was significantly blunted in Sost(-/-) ;Lrp5(-/-) mice. Yet the Lrp5 OPPG phenotype was fully rescued in Sost(-/-) ;Lrp5(-/-) mice and most bone parameters were elevated relative to wild-type. To test whether the remaining bone increases in Sost(-/-) ;Lrp5(-/-) animals depend on Lrp6, we treated wild-type, Sost(-/-) , and Sost(-/-) ;Lrp5(-/-) mice with distinct Lrp6 function blocking antibodies. Selective blockage of Wnt1 class-mediated Lrp6 signaling reduced cancellous bone mass and density in wild-type mice. Surprisingly, it reversed the abnormal bone gain in Sost(-/-) and Sost(-/-) ;Lrp5(-/-) mice to wild-type levels irrespective of enhancement or blockage of Wnt3a class-mediated Lrp6 activity. Thus, whereas Sost deficiency-induced bone anabolism partially requires Lrp5, it fully depends on Wnt1 class-induced Lrp6 activity. These findings indicate: first, that OPPG syndrome patients suffering from LRP5 loss-of-function should benefit from principles antagonizing SOST/sclerostin action; and second, that therapeutic WNT signaling inhibitors may stop the debilitating bone overgrowth in sclerosing disorders related to SOST deficiency, such as sclerosteosis, van Buchem disease, and autosomal dominant craniodiaphyseal dysplasia, which are rare disorders without viable treatment options.


Assuntos
Glicoproteínas/deficiência , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Via de Sinalização Wnt/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Densidade Óssea , Desenvolvimento Ósseo/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/imunologia , Camundongos , Osteogênese Imperfeita/tratamento farmacológico , Tíbia/química , Via de Sinalização Wnt/efeitos dos fármacos , Microtomografia por Raio-X
14.
Osteoarthritis Cartilage ; 21(10): 1576-85, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23756208

RESUMO

OBJECTIVE: Wnt/ß-catenin signaling plays an integral and complex role in cartilage development and maintenance. ß-catenin signaling has been linked to osteoarthritis (OA), but the role of Lrp6-mediated Wnt/ß-catenin signaling during OA remains unexplored. Mutations in the Wnt/ß-catenin co-receptors LRP5 and LRP6 (low-density lipoprotein-related receptors 5 and 6) result in skeletal abnormalities, which tend to be more severe in Lrp6 mutant mice. We examined OA development, chondrocyte and osteoblast behavior, and ß-catenin signaling after ligament and meniscus damage in mice with global heterozygous deletion of Lrp6. DESIGN: Ligament and meniscus damage was surgically induced in Lrp6(+/-) and wild-type (WT) mice, and evidence of joint disease was assessed by Microcomputed tomography (micro-CT) and histology. Wnt/ß-catenin signaling, proliferation, apoptosis, chondrogenesis, osteogenesis, and catabolic enzyme activity were measured. RESULTS: Relative to WT mice, Lrp6(+/-) mice had lower nuclear ß-catenin signaling within articular cartilage. After surgery, osteophytes and reduced articular cartilage were apparent in WT mice, but more severe in Lrp6(+/-) animals. Impairments to trabecular bone geometry occurred for WT and Lrp6(+/-) mice after surgery. Relative to WT mice, Lrp6(+/-) mice had reduced trabecular BMD and thickness, and Cyclin D1 and Lrp6 gene expression after surgery. There was an increase in apoptotic cells and serum matrix metalloproteinase-9 (MMP9) for Lrp6(+/-) mice after surgery, but no differences in cell proliferation occurred. CONCLUSIONS: Heterozygous loss-of-function mutation in Lrp6 leads to less ß-catenin signaling within articular cartilage and to increased degenerative joint disease after ligament and meniscus injury. Modulation of Lrp6 function could attenuate joint disease after damage to ligaments and the meniscus.


Assuntos
Artrite Experimental/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Mutação , Osteoartrite/genética , Animais , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Cartilagem Articular/metabolismo , Heterozigoto , Ligamentos Articulares/lesões , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Masculino , Camundongos , Camundongos Mutantes , Osteoartrite/metabolismo , Osteoartrite/patologia , Tíbia/patologia , Lesões do Menisco Tibial , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo
15.
Stroke ; 44(8): 2284-2291, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23743975

RESUMO

BACKGROUND AND PURPOSE: Loss-of-function mutations of the lipoprotein receptor-related protein-6 (LRP6), a coreceptor in the Wingless-related integration site-ß-catenin prosurvival pathway, have been implicated in myocardial ischemia and neurodegeneration. However, it remains to be established whether LRP6 is also involved in ischemic brain injury. We used LRP6+/- mice to examine the role of this receptor in the mechanisms of focal cerebral ischemia. METHODS: Focal cerebral ischemia was induced by transient occlusion of the middle cerebral artery. Motor deficits and infarct volume were assessed 3 days later. Glycogen-synthase-kinase-3ß (GSK-3ß) phosphorylation was examined by Western blotting with phosphospecific antibodies, and the mitochondrial membrane potential changes induced by Ca2+ were also assessed. RESULTS: LRP6+/- mice have larger stroke and more severe motor deficits, effects that were independent of intraischemic cerebral blood flow, vascular factors, or cytosolic ß-catenin levels. Rather, LRP6 haploinsufficiency increased the activating phosphorylation and decreased the inhibitory phosphorylation of GSK-3ß, a kinase involved in proinflammatory signaling and mitochondrial dysfunction. Accordingly, postischemic inflammatory gene expression was enhanced in LRP6+/- mice. Furthermore, the association of mitochondria with activated GSK-3ß was increased in LRP6+/- mice, resulting in a reduction in the Ca2+ handling ability of mitochondria. The mitochondrial dysfunction was reversed by pharmacological inhibition of GSK-3ß. CONCLUSIONS: LRP6 activates an endogenous neuroprotective pathway that acts independently of ß-catenin by controlling GSK-3ß activity and preventing its deleterious mitochondrial and proinflammatory effects. The findings raise the possibility that emerging treatment strategies for diseases attributable to LRP6 loss-of-function mutations could also lead to new therapeutic avenues for ischemic stroke.


Assuntos
Isquemia Encefálica/prevenção & controle , Encéfalo/metabolismo , Encéfalo/patologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Animais , Comportamento Animal/fisiologia , Encéfalo/fisiopatologia , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Inflamação/genética , Inflamação/metabolismo , Inflamação/prevenção & controle , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Camundongos , Mitocôndrias/genética , Atividade Motora/genética , Fosforilação/genética , Transdução de Sinais/genética , beta Catenina/genética , beta Catenina/metabolismo , beta Catenina/fisiologia
16.
Hum Mol Genet ; 22(21): 4267-81, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23773994

RESUMO

Low-density lipoprotein receptor related protein 6 (Lrp6) mutational effects on neurulation were examined using gain (Crooked tail, Lrp6(Cd)) and loss (Lrp6(-)) of function mouse lines. Two features often associated with canonical Wnt signaling, dorsal-ventral patterning and proliferation, were no different from wild-type (WT) in the Lrp6(Cd/Cd) neural tube. Lrp6(-/-) embryos showed reduced proliferation and subtle patterning changes in the neural folds. Cell polarity defects in both Lrp6(Cd/Cd) and Lrp6(-/-) cranial folds were indicated by cell shape, centrosome displacement and failure of F-actin and GTP-RhoA accumulation at the apical surface. Mouse embryonic fibroblasts (MEFs) derived from Lrp6(Cd/Cd) or Lrp6(-/-) embryos exhibited elevated and decreased RhoA basal activity levels, respectively. While ligand-independent activation of canonical Wnt signaling, bypassing Lrp-Frizzled receptors, did not activate RhoA, non-canonical Wnt5a stimulation of RhoA activity was impaired in Lrp6(-/-) MEFs. RhoA inhibition exacerbated NTDs in cultured Lrp6 knockout embryos compared with WT littermates. In contrast, a ROCK inhibitor rescued Lrp6(Cd/Cd) embryos from NTDs. Lrp6 co-immunoprecipitated with Disheveled-associated activator of morphogenesis 1 (DAAM1), a formin promoting GEF activity in Wnt signaling. Biochemical and cell biological data revealed intracellular accumulation of Lrp6(Cd) protein where interaction with DAAM1 could account for observed elevated RhoA activity. Conversely, null mutation that eliminates Lrp6 interaction with DAAM1 led to lower basal RhoA activity in Lrp6(-/-) embryos. These results indicate that Lrp6 mediates not only canonical Wnt signaling, but can also modulate non-canonical pathways involving RhoA-dependent mechanisms to impact neurulation, possibly through intracellular complexes with DAAM1.


Assuntos
Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Tubo Neural/embriologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Alelos , Animais , Polaridade Celular , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Células NIH 3T3 , Crista Neural/metabolismo , Tubo Neural/fisiologia , Neurulação/genética , Gravidez , Proteínas Wnt/genética , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
17.
Contrib Nephrol ; 180: 124-37, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23652555

RESUMO

Deregulated phosphate homeostasis can lead to a wide range of disorders, including myopathy, cardiac dysfunction, and skeletal abnormalities. Therefore, characterization of the molecular regulation of phosphate metabolism is of pathophysiological and clinical significance. Hyp mouse is the model for human X-linked hypophosphatemia which is due to mutations that inactivate the endopeptidases of the X chromosome (PHEX). PHEX inactivation leads to increased serum levels of fibroblast growth factor 23 (FGF23), a phosphaturic hormone that induces excessive renal phosphate excretion and severe hypophosphatemia. The expression of WNT signaling components is increased in Hyp mice. To determine the potential role of WNT signaling in FGF23-mediated hypophosphatemia, we cross-bred Hyp mice with mice deficient in the WNT coreceptor low-density lipoprotein receptor-related protein 6 (Lrp6) to generate Hyp and Lrp6 double mutant mice (Hyp/Lrp6). Like Hyp mice, Hyp/Lrp6 double mutants maintained high serum levels of FGF23, and accordingly exhibited hypophosphatemia to the same degree as the Hyp mice did, indicating that genetically reducing WNT signaling does not impact FGF23-induced phosphaturia. Moreover, similar to Hyp mice, the Hyp/Lrp6 double mutants also exhibited reduced mineralization of the bone, further supporting that reduced WNT signaling does not affect the chronic phosphate wasting caused by excess FGF23 in these mice. In further support of our finding, injection of bioactive FGF23 protein into Lrp6 mutant mice reduced serum phosphate levels to a similar degree as FGF23 injection into wild-type mice. Our in vivo studies provide genetic and pharmacological evidence for a WNT-independent function of FGF23 in the regulation of phosphate homeostasis.


Assuntos
Modelos Animais de Doenças , Raquitismo Hipofosfatêmico Familiar/fisiopatologia , Fatores de Crescimento de Fibroblastos/fisiologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Endopeptidase Neutra Reguladora de Fosfato PHEX/fisiologia , Via de Sinalização Wnt , Animais , Raquitismo Hipofosfatêmico Familiar/diagnóstico por imagem , Raquitismo Hipofosfatêmico Familiar/etiologia , Raquitismo Hipofosfatêmico Familiar/genética , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/toxicidade , Homeostase , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/metabolismo , Rim/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Masculino , Camundongos , Camundongos Knockout , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Fosfatos/metabolismo , Radiografia , Proteínas Recombinantes/toxicidade , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/biossíntese , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética
18.
Nat Med ; 19(2): 179-92, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23389618

RESUMO

Low bone mass and strength lead to fragility fractures, for example, in elderly individuals affected by osteoporosis or children with osteogenesis imperfecta. A decade ago, rare human mutations affecting bone negatively (osteoporosis-pseudoglioma syndrome) or positively (high-bone mass phenotype, sclerosteosis and Van Buchem disease) have been identified and found to all reside in components of the canonical WNT signaling machinery. Mouse genetics confirmed the importance of canonical Wnt signaling in the regulation of bone homeostasis, with activation of the pathway leading to increased, and inhibition leading to decreased, bone mass and strength. The importance of WNT signaling for bone has also been highlighted since then in the general population in numerous genome-wide association studies. The pathway is now the target for therapeutic intervention to restore bone strength in millions of patients at risk for fracture. This paper reviews our current understanding of the mechanisms by which WNT signalng regulates bone homeostasis.


Assuntos
Osso e Ossos/metabolismo , Homeostase , Mutação , Via de Sinalização Wnt/fisiologia , Animais , Densidade Óssea , Desenvolvimento Ósseo , Proteínas Morfogenéticas Ósseas/fisiologia , Hematopoese , Humanos , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Camundongos , beta Catenina/fisiologia
19.
PLoS One ; 7(5): e36520, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22606268

RESUMO

Aberrant activation of the Wnt pathway contributes to human cancer progression. Antagonists that interfere with Wnt ligand/receptor interactions can be useful in cancer treatments. In this study, we evaluated the therapeutic potential of a soluble Wnt receptor decoy in cancer gene therapy. We designed a Wnt antagonist sLRP6E1E2, and generated a replication-incompetent adenovirus (Ad), dE1-k35/sLRP6E1E2, and a replication-competent oncolytic Ad, RdB-k35/sLRP6E1E2, both expressing sLRP6E1E2. sLRP6E1E2 prevented Wnt-mediated stabilization of cytoplasmic ß-catenin, decreased Wnt/ß-catenin signaling and cell proliferation via the mitogen-activated protein kinase, and phosphatidylinositol 3-kinase pathways. sLRP6E1E2 induced apoptosis, cytochrome c release, and increased cleavage of PARP and caspase-3. sLRP6E1E2 suppressed growth of the human lung tumor xenograft, and reduced motility and invasion of cancer cells. In addition, sLRP6E1E2 upregulated expression of epithelial marker genes, while sLRP6E1E2 downregulated mesenchymal marker genes. Taken together, sLRP6E1E2, by inhibiting interaction between Wnt and its receptor, suppressed Wnt-induced cell proliferation and epithelial-to-mesenchymal transition.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Neoplasias Pulmonares/terapia , Via de Sinalização Wnt/fisiologia , Adenovírus Humanos/genética , Animais , Apoptose/genética , Apoptose/fisiologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/fisiopatologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Linhagem Celular Tumoral , Proliferação de Células , Transição Epitelial-Mesenquimal/genética , Terapia Genética/métodos , Vetores Genéticos , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/antagonistas & inibidores , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/fisiopatologia , Masculino , Camundongos , Camundongos Nus , Mitocôndrias/genética , Mitocôndrias/fisiologia , Invasividade Neoplásica/prevenção & controle , RNA Interferente Pequeno/genética , Receptores Wnt/metabolismo , Fatores de Transcrição TCF/metabolismo , Proteína Wnt3A/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/metabolismo
20.
FEBS J ; 278(24): 4704-16, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21902810

RESUMO

Tissue transglutaminase (TG2) is a ubiquitously expressed member of the transglutaminase family of Ca(2+)-dependent crosslinking enzymes. Unlike other family members, TG2 is a multifunctional protein, which has several other well documented enzymatic and non-enzymatic functions. A significant body of evidence accumulated over the last decade reveals multiple and complex activities of this protein on the cell surface and in the extracellular matrix (ECM), including its role in the regulation of cell-ECM interactions and outside-in signaling by several types of transmembrane receptors. Moreover, recent findings indicate a dynamic regulation of the levels and functions of extracellular TG2 by several complementary mechanisms. This review summarizes and assesses recent research into the emerging functions and regulation of extracellular TG2.


Assuntos
Matriz Extracelular/metabolismo , Proteínas de Ligação ao GTP/fisiologia , Transglutaminases/fisiologia , Adesão Celular/fisiologia , Reagentes de Ligações Cruzadas/metabolismo , Cisteína/metabolismo , Ativação Enzimática , Fibronectinas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Humanos , Integrinas/metabolismo , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Macrófagos/fisiologia , Metaloproteinases da Matriz Associadas à Membrana/metabolismo , Transplante de Células-Tronco Mesenquimais , Conformação Proteica/efeitos dos fármacos , Proteína 2 Glutamina gama-Glutamiltransferase , Transporte Proteico/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Transdução de Sinais/fisiologia , Sindecana-4/fisiologia , Transglutaminases/metabolismo , beta Catenina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...