Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Blood ; 136(17): 1907-1918, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-32573726

RESUMO

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant vascular dysplasia. Care delivery for HHT patients is impeded by the need for laborious, repeated phenotyping and gaps in knowledge regarding the relationships between causal DNA variants in ENG, ACVRL1, SMAD4 and GDF2, and clinical manifestations. To address this, we analyzed DNA samples from 183 previously uncharacterized, unrelated HHT and suspected HHT cases using the ThromboGenomics high-throughput sequencing platform. We identified 127 rare variants across 168 heterozygous genotypes. Applying modified American College of Medical Genetics and Genomics Guidelines, 106 variants were classified as pathogenic/likely pathogenic and 21 as nonpathogenic (variant of uncertain significance/benign). Unlike the protein products of ACVRL1 and SMAD4, the extracellular ENG amino acids are not strongly conserved. Our inferences of the functional consequences of causal variants in ENG were therefore informed by the crystal structure of endoglin. We then compared the accuracy of predictions of the causal gene blinded to the genetic data using 2 approaches: subjective clinical predictions and statistical predictions based on 8 Human Phenotype Ontology terms. Both approaches had some predictive power, but they were insufficiently accurate to be used clinically, without genetic testing. The distributions of red cell indices differed by causal gene but not sufficiently for clinical use in isolation from genetic data. We conclude that parallel sequencing of the 4 known HHT genes, multidisciplinary team review of variant calls in the context of detailed clinical information, and statistical and structural modeling improve the prognostication and treatment of HHT.


Assuntos
Estudos de Associação Genética , Mutação , Telangiectasia Hemorrágica Hereditária/genética , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Estudos de Coortes , Análise Mutacional de DNA/métodos , Endoglina/química , Endoglina/genética , Feminino , Estudos de Associação Genética/métodos , Predisposição Genética para Doença , Testes Genéticos/métodos , Genômica/métodos , Fator 2 de Diferenciação de Crescimento/química , Fator 2 de Diferenciação de Crescimento/genética , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Modelos Moleculares , Fenótipo , Estudos Retrospectivos , Análise de Sequência de DNA/métodos , Proteína Smad4/química , Proteína Smad4/genética , Telangiectasia Hemorrágica Hereditária/epidemiologia , Telangiectasia Hemorrágica Hereditária/patologia
2.
Nat Commun ; 11(1): 1621, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32238803

RESUMO

Activin receptor-like kinase 1 (ALK1)-mediated endothelial cell signalling in response to bone morphogenetic protein 9 (BMP9) and BMP10 is of significant importance in cardiovascular disease and cancer. However, detailed molecular mechanisms of ALK1-mediated signalling remain unclear. Here, we report crystal structures of the BMP10:ALK1 complex at 2.3 Å and the prodomain-bound BMP9:ALK1 complex at 3.3 Å. Structural analyses reveal a tripartite recognition mechanism that defines BMP9 and BMP10 specificity for ALK1, and predict that crossveinless 2 is not an inhibitor of BMP9, which is confirmed by experimental evidence. Introduction of BMP10-specific residues into BMP9 yields BMP10-like ligands with diminished signalling activity in C2C12 cells, validating the tripartite mechanism. The loss of osteogenic signalling in C2C12 does not translate into non-osteogenic activity in vivo and BMP10 also induces bone-formation. Collectively, these data provide insight into ALK1-mediated BMP9 and BMP10 signalling, facilitating therapeutic targeting of this important pathway.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Fator 2 de Diferenciação de Crescimento/metabolismo , Transdução de Sinais/fisiologia , Receptores de Activinas Tipo II/química , Animais , Sítios de Ligação , Proteínas Morfogenéticas Ósseas/química , Osso e Ossos/química , Osso e Ossos/metabolismo , Linhagem Celular , Cristalografia por Raios X , Células Endoteliais/metabolismo , Fator 2 de Diferenciação de Crescimento/química , Humanos , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Conformação Proteica , Domínios Proteicos , Fator de Crescimento Transformador beta/metabolismo
3.
Sci Rep ; 9(1): 13446, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31530856

RESUMO

Bone morphogenetic proteins (BMPs) are multifunctional cytokines of the transforming growth factor ß (TGFß) superfamily with potential therapeutic applications due to their broad biological functionality. Designing BMP mimetics with specific activity will contribute to the translational potential of BMP-based therapies. Here, we report a BMP9 peptide mimetic, P3, designed from the type I receptor binding site, which showed millimolar binding affinities for the type I receptor activin receptor like kinase 1 (ALK1), ALK2 and ALK3. Although showing no baseline activity, P3 significantly enhanced BMP9-induced Smad1/5 phosphorylation as well as ID1, BMPR2, HEY1 and HEY2 gene expression in pulmonary artery endothelial cells (hPAECs), and this activity is dependent on its alpha helix propensity. However, in human dermal microvascular endothelial cells, P3 did not affect BMP9-induced Smad1/5 phosphorylation, but potently inhibited ALK3-dependent BMP4-induced Smad1/5 phosphorylation and gene expression. In C2C12 mouse myoblast cells, P3 had no effect on BMP9-induced osteogenic signalling, which is primarily mediated by ALK2. Interestingly, a previously published peptide from the knuckle region of BMP9 was found to inhibit BMP4-induced Smad1/5 phosphorylation. Together, our data identify a BMP9-derived peptide that can selectively enhance ALK1-mediated BMP9 signalling in hPAECs and modulate BMP9 and BMP4 signalling in a cell type-specific manner.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Animais , Sítios de Ligação , Linhagem Celular , Células Endoteliais , Fator 2 de Diferenciação de Crescimento/química , Fator 2 de Diferenciação de Crescimento/metabolismo , Humanos , Camundongos , Mimetismo Molecular , Peptídeos/metabolismo , Fosforilação , Transdução de Sinais , Proteína Smad1/metabolismo , Proteína Smad8/metabolismo , Relação Estrutura-Atividade
5.
J Med Chem ; 60(4): 1495-1508, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28103025

RESUMO

We synthesized 1 (San78-130), a reversible version of L-783277, as a selective and potent ALK1 inhibitor. Our study showed that 1 possesses great kinase selectivity against a panel of 342 kinases and more potent activity against ALK1 than L-783277. Among the six ALK isotypes (ALK1-6), ALK1 is most significantly inhibited by compound 1. Compound 1 suppresses the BMP9-induced Smad1/5 pathway by mainly inhibiting ALK1 in C2C12 cells. Our molecular dynamics simulations suggest that H-bonding interaction between the C-4' hydroxyl group of 1 and Arg334 of ALK1 substantially contributes to the ALK1 inhibition. To the best of our knowledge, 1 is the first selective ALK1 inhibitor. Furthermore, compound 1 promoted angiogenesis in both endothelial tube formation and microfluidic chip based 3D angiogenesis assays, suggesting that 1 could be a lead compound for therapeutic angiogenesis agents. Our study may provide an insight into designing selective and potent inhibitors against ALK1.


Assuntos
Receptores de Activinas Tipo II/antagonistas & inibidores , Lactonas/química , Lactonas/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Resorcinóis/química , Resorcinóis/farmacologia , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Sequência de Aminoácidos , Indutores da Angiogênese/química , Indutores da Angiogênese/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Alinhamento de Sequência , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo
6.
Cancer Sci ; 108(1): 151-155, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28133920

RESUMO

Clinical development of anti-angiogenic agents has been a major landmark in cancer therapy for several types of cancers. Signals mediated by both vascular endothelial growth factor (VEGF) and bone morphogenetic protein (BMP)-9 and 10 have been implicated in tumor angiogenesis. However, previous studies have shown that targeting the individual signals was not sufficiently effective in retarding tumor growth in certain preclinical and clinical conditions. In the present study, we developed a novel decoy chimeric receptor that traps both VEGF and BMP-9/10. Single targeting of either VEGF or BMP-9/10 signals significantly reduced the formation of tumor vessels in a mouse xenograft model of human pancreatic cancer; however, it did not show significant therapeutic effects on tumor growth. In contrast, dual targeting of the angiogenic signals resulted in more significant inhibition of tumor angiogenesis, leading to delay of tumor growth. Our findings suggest that simultaneous blockade of VEGF and BMP-9/10 signals is a promising therapeutic strategy for the cancers that are resistant to anti-VEGF and BMP-9/10 therapies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Neovascularização Patológica/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/farmacologia , Receptores de Activinas Tipo II/uso terapêutico , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Proliferação de Células , Feminino , Fator 2 de Diferenciação de Crescimento/antagonistas & inibidores , Fator 2 de Diferenciação de Crescimento/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/farmacologia , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/irrigação sanguínea , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/química , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
7.
PLoS One ; 11(11): e0166803, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27870893

RESUMO

Spinal muscular atrophy (SMA) is a devastating neurodegenerative disorder that causes progressive muscle atrophy and weakness. Using adeno-associated virus-mediated gene transfer, we evaluated the potential to improve skeletal muscle weakness via systemic, postnatal inhibition of either myostatin or all signaling via the activin receptor type IIB (ActRIIB). After demonstrating elevated p-SMAD3 content and differential content of ActRIIB ligands, 4-week-old male C/C SMA model mice were treated intraperitoneally with 1x1012 genome copies of pseudotype 2/8 virus encoding a soluble form of the ActRIIB extracellular domain (sActRIIB) or protease-resistant myostatin propeptide (dnMstn) driven by a liver specific promoter. At 12 weeks of age, muscle mass and function were improved in treated C/C mice by both treatments, compared to controls. The fast fiber type muscles had a greater response to treatment than did slow muscles, and the greatest therapeutic effects were found with sActRIIB treatment. Myostatin/activin inhibition, however, did not rescue C/C mice from the reduction in motor unit numbers of the tibialis anterior muscle. Collectively, this study indicates that myostatin/activin inhibition represents a potential therapeutic strategy to increase muscle mass and strength, but not neuromuscular junction defects, in less severe forms of SMA.


Assuntos
Receptores de Activinas Tipo II/genética , Músculo Esquelético/patologia , Atrofia Muscular Espinal/terapia , Miostatina/antagonistas & inibidores , Peptídeos/genética , Receptores de Activinas Tipo II/antagonistas & inibidores , Receptores de Activinas Tipo II/química , Animais , Dependovirus/genética , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Masculino , Camundongos , Contração Muscular , Músculo Esquelético/fisiopatologia , Atrofia Muscular Espinal/patologia , Atrofia Muscular Espinal/fisiopatologia , Miostatina/genética , Tamanho do Órgão , Peptídeos/farmacologia , Fenótipo
8.
Am J Hematol ; 90(1): 8-14, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25236856

RESUMO

Over expression of hepcidin antimicrobial peptide is a common feature of iron-restricted anemia in humans. We investigated the erythroid response to either erythropoietin or RAP-011, a "murinized" ortholog of sotatercept, in C57BL/6 mice and in hepcidin antimicrobial peptide 1 over expressing mice. Sotatercept, a soluble, activin receptor type IIA ligand trap, is currently being evaluated for the treatment of anemias associated with chronic renal disease, myelodysplastic syndrome, ß-thalassemia, and Diamond Blackfan anemia and acts by inhibiting signaling downstream of activin and other Transforming Growth Factor-ß superfamily members. We found that erythropoietin and RAP-011 increased hemoglobin concentration in C57BL/6 mice and in hepcidin antimicrobial peptide 1 over expressing mice. While erythropoietin treatment depleted splenic iron stores in C57BL/6 mice, RAP-011 treatment did not deplete splenic iron stores in mice of either genotype. Bone marrow erythroid progenitors from erythropoietin-treated mice exhibited iron-restricted erythropoiesis, as indicated by increased median fluorescence intensity of transferrin receptor immunostaining by flow cytometry. In contrast, RAP-011-treated mice did not exhibit the same degree of iron-restricted erythropoiesis. In conclusion, we have demonstrated that RAP-011 can improve hemoglobin concentration in hepcidin antimicrobial peptide 1 transgenic mice. Our data support the hypothesis that RAP-011 has unique biologic effects which prevent or circumvent depletion of mouse splenic iron stores. RAP-011 may, therefore, be an appropriate therapeutic for trials in human anemias characterized by increased expression of hepcidin antimicrobial peptide and iron-restricted erythropoiesis.


Assuntos
Eritropoese/efeitos dos fármacos , Hemoglobinas/análise , Hepcidinas/genética , Ferro/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Receptores de Activinas Tipo II/química , Animais , Transporte Biológico , Contagem de Células Sanguíneas , Células Precursoras Eritroides/efeitos dos fármacos , Eritropoetina/farmacologia , Feminino , Imunoglobulina G/química , Ferro/sangue , Ligantes , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Baço/metabolismo
9.
J Biol Chem ; 290(6): 3390-404, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25368322

RESUMO

GDF8, or myostatin, is a member of the TGF-ß superfamily of secreted polypeptide growth factors. GDF8 is a potent negative regulator of myogenesis both in vivo and in vitro. We found that GDF8 signaling was inhibited by the small molecule ATP competitive inhibitors dorsomorphin and LDN-193189. These compounds were previously shown to be potent inhibitors of BMP signaling by binding to the BMP type I receptors ALK1/2/3/6. We present the crystal structure of the type II receptor ActRIIA with dorsomorphin and demonstrate that dorsomorphin or LDN-193189 target GDF8 induced Smad2/3 signaling and repression of myogenic transcription factors. As a result, both inhibitors rescued myogenesis in myoblasts treated with GDF8. As revealed by quantitative live cell microscopy, treatment with dorsomorphin or LDN-193189 promoted the contractile activity of myotubular networks in vitro. We therefore suggest these inhibitors as suitable tools to promote functional myogenesis.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Diferenciação Celular , Mioblastos/efeitos dos fármacos , Miostatina/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais , Receptores de Activinas Tipo II/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Humanos , Camundongos , Dados de Sequência Molecular , Mioblastos/citologia , Mioblastos/metabolismo , Ligação Proteica , Células Sf9 , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Spodoptera , Fatores de Transcrição/metabolismo
10.
Stem Cell Reports ; 3(6): 1103-17, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25458897

RESUMO

Embryonic stem cells (ESCs) enable rapid proliferation that also causes DNA damage. To maintain genomic stabilization during rapid proliferation, ESCs must have an efficient system to repress genotoxic stress. Here, we show that withdrawal of leukemia inhibitory factor (LIF), which maintains the self-renewal capability of mouse ESCs (mESCs), significantly inhibits the cell proliferation and DNA damage of mESCs and upregulates the expression of miR-590. miR-590 promotes single-strand break (SSB) and double-strand break (DSB) damage repair, thus slowing proliferation of mESCs without influencing stemness. miR-590 directly targets Activin receptor type 2a (Acvr2a) to mediate Activin signaling. We identified the homologous recombination-mediated repair (HRR) gene, Rad51b, as a downstream molecule of the miR-590/Acvr2a pathway regulating the SSB and DSB damage repair and cell cycle. Our study shows that a miR-590/Acvr2a/Rad51b signaling axis ensures the stabilization of mESCs by balancing DNA damage repair and rapid proliferation during self-renewal.


Assuntos
Receptores de Activinas Tipo II/genética , Dano ao DNA , Reparo do DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , MicroRNAs/genética , Rad51 Recombinase/genética , Regiões 3' não Traduzidas , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Proteínas de Ciclo Celular/genética , Diferenciação Celular/genética , Proliferação de Células , Quebras de DNA , Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Camundongos , MicroRNAs/química , Interferência de RNA , RNA Mensageiro/química , RNA Mensageiro/genética , Rad51 Recombinase/química , Transdução de Sinais
11.
PLoS One ; 8(7): e69535, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936038

RESUMO

TGF-ß family members play a relevant role in tumorigenic processes, including hepatocellular carcinoma (HCC), but a specific implication of the Bone Morphogenetic Protein (BMP) subfamily is still unknown. Although originally isolated from fetal liver, little is known about BMP9, a BMP family member, and its role in liver physiology and pathology. Our results show that BMP9 promotes growth in HCC cells, but not in immortalized human hepatocytes. In the liver cancer cell line HepG2, BMP9 triggers Smad1,5,8 phosphorylation and inhibitor of DNA binding 1 (Id1) expression up- regulation. Importantly, by using chemical inhibitors, ligand trap and gene silencing approaches we demonstrate that HepG2 cells autocrinely produce BMP9 that supports their proliferation and anchorage independent growth. Additionally, our data reveal that in HepG2 cells BMP9 triggers cell cycle progression, and strikingly, completely abolishes the increase in the percentage of apoptotic cells induced by long-term incubation in low serum. Collectively, our data unveil a dual role for BMP9, both promoting a proliferative response and exerting a remarkable anti-apoptotic function in HepG2 cells, which result in a robust BMP9 effect on liver cancer cell growth. Finally, we show that BMP9 expression is increased in 40% of human HCC tissues compared with normal human liver as revealed by immunohistochemistry analysis, suggesting that BMP9 signaling may be relevant during hepatocarcinogenesis in vivo. Our findings provide new clues for a better understanding of BMPs contribution, and in particular BMP9, in HCC pathogenesis that may result in the development of effective and targeted therapeutic interventions.


Assuntos
Carcinoma Hepatocelular/metabolismo , Fatores de Diferenciação de Crescimento/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Apoptose/efeitos dos fármacos , Comunicação Autócrina/efeitos dos fármacos , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Carcinoma Hepatocelular/patologia , Adesão Celular/efeitos dos fármacos , Contagem de Células , Linhagem Celular Transformada , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fator 2 de Diferenciação de Crescimento , Fatores de Diferenciação de Crescimento/farmacologia , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Neoplasias Hepáticas/patologia , Estrutura Terciária de Proteína , Soro , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo
12.
PLoS One ; 8(8): e72407, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967299

RESUMO

Mortality from prostate cancer (PCa) is due to the formation of metastatic disease. Understanding how that process is regulated is therefore critical. We previously demonstrated that endoglin, a type III transforming growth factor ß (TGFß) superfamily receptor, suppresses human PCa cell invasion and metastasis. Endoglin-mediated suppression of invasion was also shown by us to be dependent upon the type I TGFß receptor, activin receptor-like kinase 2 (ALK2), and the downstream effector, Smad1. In this study we demonstrate for the first time that two type II TGFß receptors are required for endoglin-mediated suppression of invasion: activin A receptor type IIA (ActRIIA) and bone morphogenetic protein receptor type II (BMPRII). Downstream signaling through these receptors is predominantly mediated by Smad1. ActRIIA stimulates Smad1 activation in a kinase-dependent manner, and this is required for suppression of invasion. In contrast BMPRII regulates Smad1 in a biphasic manner, promoting Smad1 signaling through its kinase domain but suppressing it through its cytoplasmic tail. BMPRII's Smad1-regulatory effects are dependent upon its expression level. Further, its ability to suppress invasion is independent of either kinase function or tail domain. We demonstrate that ActRIIA and BMPRII physically interact, and that each also interacts with endoglin. The current findings demonstrate that both BMPRII and ActRIIA are necessary for endoglin-mediated suppression of human PCa cell invasion, that they have differential effects on Smad1 signaling, that they make separate contributions to regulation of invasion, and that they functionally and physically interact.


Assuntos
Ativinas/metabolismo , Antígenos CD/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores de Superfície Celular/metabolismo , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/química , Linhagem Celular Tumoral , Endoglina , Ativação Enzimática , Humanos , Masculino , Invasividade Neoplásica , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transdução de Sinais , Proteína Smad1/metabolismo
13.
Protein Sci ; 22(8): 1060-70, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23776011

RESUMO

A plethora of both experimental and computational methods have been proposed in the past 20 years for the identification of hot spots at a protein-protein interface. The experimental determination of a protein-protein complex followed by alanine scanning mutagenesis, though able to determine hot spots with much precision, is expensive and has no guarantee of success while the accuracy of the current computational methods for hot-spot identification remains low. Here, we present a novel structure-based computational approach that accurately determines hot spots through docking into a set of proteins homologous to only one of the two interacting partners of a compound capable of disrupting the protein-protein interaction (PPI). This approach has been applied to identify the hot spots of human activin receptor type II (ActRII) critical for its binding toward Cripto-I. The subsequent experimental confirmation of the computationally identified hot spots portends a potentially accurate method for hot-spot determination in silico given a compound capable of disrupting the PPI in question. The hot spots of human ActRII first reported here may well become the focal points for the design of small molecule drugs that target the PPI. The determination of their interface may have significant biological implications in that it suggests that Cripto-I plays an important role in both activin and nodal signal pathways.


Assuntos
Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Simulação de Acoplamento Molecular/métodos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Mapeamento de Interação de Proteínas/métodos , Receptores de Activinas Tipo II/genética , Ativinas/genética , Ativinas/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Bases de Dados de Proteínas , Proteínas Ligadas por GPI/genética , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Modelos Moleculares , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Ligação Proteica , Conformação Proteica , Alinhamento de Sequência , Transdução de Sinais
14.
Biochim Biophys Acta ; 1832(10): 1492-510, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23707512

RESUMO

Activin receptor-like kinase-1 or ALK-1 is a type I cell surface receptor for the transforming growth factor-ß (TGF-ß) family of proteins. The role of ALK-1 in endothelial cells biology and in angiogenesis has been thoroughly studied by many authors. However, it has been recently suggested a possible role of ALK-1 in cardiovascular homeostasis. ALK-1 is not only expressed in endothelial cells but also in smooth muscle cells, myofibroblast, hepatic stellate cells, chondrocytes, monocytes, myoblasts, macrophages or fibroblasts, but its role in these cells have not been deeply analyzed. Due to the function of ALK-1 in these cells, this receptor plays a role in several cardiovascular diseases. Animals with ALK-1 haploinsufficiency and patients with mutations in Acvrl1 (the gene that codifies for ALK-1) develop type-2 Hereditary Hemorrhagic Telangiectasia. Moreover, ALK-1 heterozygous mice develop pulmonary hypertension. Higher levels of ALK-1 have been observed in atherosclerotic plaques, suggesting a possible protector role of this receptor. ALK-1 deficiency is also related to the development of arteriovenous malformations (AVMs). Besides, due to the ability of ALK-1 to regulate cell proliferation and migration, and to modulate extracellular matrix (ECM) protein expression in several cell types, ALK-1 has been now demonstrated to play an important role in cardiovascular remodeling. In this review, we would like to offer a complete vision of the role of ALK-1 in many process related to cardiovascular homeostasis, and the involvement of this protein in the development of cardiovascular diseases, suggesting the possibility of using the ALK-1/smad-1 pathway as a powerful therapeutic target.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Doenças Cardiovasculares/metabolismo , Proteína Smad1/metabolismo , Receptores de Activinas Tipo II/química , Homeostase , Humanos , Transdução de Sinais , Proteína Smad1/química , Fator de Crescimento Transformador beta/metabolismo
15.
J Biol Chem ; 287(33): 27313-25, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22718755

RESUMO

Activin receptor-like kinase 1 (ALK1), an endothelial cell-specific type I receptor of the TGF-ß superfamily, is an important regulator of normal blood vessel development as well as pathological tumor angiogenesis. As such, ALK1 is an important therapeutic target. Thus, several ALK1-directed agents are currently in clinical trials as anti-angiogenic cancer therapeutics. Given the biological and clinical importance of the ALK1 signaling pathway, we sought to elucidate the biophysical and structural basis underlying ALK1 signaling. The TGF-ß family ligands BMP9 and BMP10 as well as the three type II TGF-ß family receptors ActRIIA, ActRIIB, and BMPRII have been implicated in ALK1 signaling. Here, we provide a kinetic and thermodynamic analysis of BMP9 and BMP10 interactions with ALK1 and type II receptors. Our data show that BMP9 displays a significant discrimination in type II receptor binding, whereas BMP10 does not. We also report the crystal structure of a fully assembled ternary complex of BMP9 with the extracellular domains of ALK1 and ActRIIB. The structure reveals that the high specificity of ALK1 for BMP9/10 is determined by a novel orientation of ALK1 with respect to BMP9, which leads to a unique set of receptor-ligand interactions. In addition, the structure explains how BMP9 discriminates between low and high affinity type II receptors. Taken together, our findings provide structural and mechanistic insights into ALK1 signaling that could serve as a basis for novel anti-angiogenic therapies.


Assuntos
Receptores de Activinas Tipo II/química , Proteínas Morfogenéticas Ósseas/química , Fatores de Diferenciação de Crescimento/química , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Inibidores da Angiogênese/química , Inibidores da Angiogênese/uso terapêutico , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Cristalografia por Raios X , Fator 2 de Diferenciação de Crescimento , Fatores de Diferenciação de Crescimento/genética , Fatores de Diferenciação de Crescimento/metabolismo , Células HEK293 , Humanos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
16.
Mol Cell Endocrinol ; 359(1-2): 2-12, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21763751

RESUMO

Like other members of the transforming growth factor-ß (TGF-ß) superfamily, activins are synthesised as precursor molecules comprising an N-terminal prodomain and C-terminal mature region. During synthesis, the prodomain interacts non-covalently with mature activin, maintaining the molecule in a conformation competent for dimerisation. Dimeric precursors are cleaved by proprotein convertases and activin is secreted from the cell non-covalently associated with its propeptide. Extracellularly, the propeptide interacts with heparan sulfate proteoglycans to regulate activin localization within tissues. The mature activin dimer exhibits the classic 'open-hand' structure of TGF-ß ligands with 'finger-like' domains projecting outward from the cysteine knot core of the molecule. These finger domains form the binding epitopes for type I and II serine/threonine kinase receptors. Activins ability to access its signalling receptors is regulated by the extracellular binding proteins, follistatin, follistatin-like-3, and by inhibins, which, in the presence of betaglycan, sequester type II receptors.


Assuntos
Ativinas/fisiologia , Precursores de Proteínas/fisiologia , Receptores de Ativinas Tipo I/metabolismo , Receptores de Ativinas Tipo I/fisiologia , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Receptores de Activinas Tipo II/fisiologia , Ativinas/química , Ativinas/metabolismo , Motivos de Aminoácidos , Animais , Humanos , Inibinas/metabolismo , Inibinas/fisiologia , Ligação Proteica , Precursores de Proteínas/metabolismo , Estrutura Terciária de Proteína , Proteoglicanas/metabolismo , Proteoglicanas/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Transdução de Sinais
17.
PLoS One ; 6(10): e26431, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22028876

RESUMO

Activin A receptor, type II-like kinase 1 (also called ALK1), is a serine-threonine kinase predominantly expressed on endothelial cells surface. Mutations in its ACVRL1 encoding gene (12q11-14) cause type 2 Hereditary Haemorrhagic Telangiectasia (HHT2), an autosomal dominant multisystem vascular dysplasia. The study of the structural effects of mutations is crucial to understand their pathogenic mechanism. However, while an X-ray structure of ALK1 intracellular domain has recently become available (PDB ID: 3MY0), structure determination of ALK1 ectodomain (ALK1(EC)) has been elusive so far. We here describe the building of a homology model for ALK1(EC), followed by an extensive bioinformatic analysis, based on a set of 38 methods, of the effect of missense mutations at the sequence and structural level. ALK1(EC) potential interaction mode with its ligand BMP9 was then predicted combining modelling and docking data. The calculated model of the ALK1(EC) allowed mapping and a preliminary characterization of HHT2 associated mutations. Major structural changes and loss of stability of the protein were predicted for several mutations, while others were found to interfere mainly with binding to BMP9 or other interactors, like Endoglin (CD105), whose encoding ENG gene (9q34) mutations are known to cause type 1 HHT. This study gives a preliminary insight into the potential structure of ALK1(EC) and into the structural effects of HHT2 associated mutations, which can be useful to predict the potential effect of each single mutation, to devise new biological experiments and to interpret the biological significance of new mutations, private mutations, or non-synonymous polymorphisms.


Assuntos
Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Biologia Computacional , Mutação de Sentido Incorreto , Receptores de Activinas Tipo II/metabolismo , Sequência de Aminoácidos , Sequência Conservada , Análise Mutacional de DNA , Estabilidade Enzimática , Fator 2 de Diferenciação de Crescimento , Fatores de Diferenciação de Crescimento/química , Fatores de Diferenciação de Crescimento/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Telangiectasia Hemorrágica Hereditária/enzimologia , Telangiectasia Hemorrágica Hereditária/genética , Telangiectasia Hemorrágica Hereditária/patologia
18.
EMBO J ; 22(7): 1555-66, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12660162

RESUMO

The TGF-beta superfamily of ligands and receptors stimulate cellular events in diverse processes ranging from cell fate specification in development to immune suppression. Activins define a major subgroup of TGF-beta ligands that regulate cellular differentiation, proliferation, activation and apoptosis. Activins signal through complexes formed with type I and type II serine/threonine kinase receptors. We have solved the crystal structure of activin A bound to the extracellular domain of a type II receptor, ActRIIB, revealing the details of this interaction. ActRIIB binds to the outer edges of the activin finger regions, with the two receptors juxtaposed in close proximity, in a mode that differs from TGF-beta3 binding to type II receptors. The dimeric activin A structure differs from other known TGF-beta ligand structures, adopting a compact folded-back conformation. The crystal structure of the complex is consistent with recruitment of two type I receptors into a close packed arrangement at the cell surface and suggests that diversity in the conformational arrangements of TGF-beta ligand dimers could influence cellular signaling processes.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Ativinas/metabolismo , Subunidades beta de Inibinas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Receptores de Activinas Tipo II/química , Ativinas/química , Sítios de Ligação , Cristalografia por Raios X , Subunidades beta de Inibinas/química , Ligantes , Modelos Moleculares , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA