Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Blood ; 136(17): 1907-1918, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32573726

RESUMEN

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.


Asunto(s)
Estudios de Asociación Genética , Mutación , Telangiectasia Hemorrágica Hereditaria/genética , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Estudios de Cohortes , Análisis Mutacional de ADN/métodos , Endoglina/química , Endoglina/genética , Femenino , Estudios de Asociación Genética/métodos , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Genómica/métodos , Factor 2 de Diferenciación de Crecimiento/química , Factor 2 de Diferenciación de Crecimiento/genética , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Modelos Moleculares , Fenotipo , Estudios Retrospectivos , Análisis de Secuencia de ADN/métodos , Proteína Smad4/química , Proteína Smad4/genética , Telangiectasia Hemorrágica Hereditaria/epidemiología , Telangiectasia Hemorrágica Hereditaria/patología
2.
Endocrinology ; 161(7)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32333774

RESUMEN

Anti-Müllerian hormone (AMH) or Müllerian inhibiting substance is a unique member of the TGF-ß family responsible for development and differentiation of the reproductive system. AMH signals through its own dedicated type II receptor, anti-Müllerian hormone receptor type II (AMHR2), providing an exclusive ligand-receptor pair within the broader TGF-ß family. In this study, we used previous structural information to derive a model of AMH bound to AMHR2 to guide mutagenesis studies to identify receptor residues important for AMH signaling. Nonconserved mutations were introduced in AMHR2 and characterized in an AMH-responsive cell-based luciferase assay and native PAGE. Collectively, our results identified several residues important for AMH signaling within the putative ligand binding interface of AMHR2. Our results show that AMH engages AMHR2 at a similar interface to how activin and BMP class ligands bind the type II receptor, ACVR2B; however, there are significant molecular differences at the ligand interface of these 2 receptors, where ACVR2B is mostly hydrophobic and AMHR2 is predominately charged. Overall, this study shows that although the location of ligand binding on the receptor is similar to ACVR2A, ACVR2B, and BMPR2; AMHR2 uses unique ligand-receptor interactions to impart specificity for AMH.


Asunto(s)
Hormona Antimülleriana/metabolismo , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Trastorno del Desarrollo Sexual 46,XY/genética , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Receptores de Péptidos/química , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/genética
3.
Nat Commun ; 11(1): 1621, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32238803

RESUMEN

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.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Factor 2 de Diferenciación de Crecimiento/metabolismo , Transducción de Señal/fisiología , Receptores de Activinas Tipo II/química , Animales , Sitios de Unión , Proteínas Morfogenéticas Óseas/química , Huesos/química , Huesos/metabolismo , Línea Celular , Cristalografía por Rayos X , Células Endoteliales/metabolismo , Factor 2 de Diferenciación de Crecimiento/química , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Conformación Proteica , Dominios Proteicos , Factor de Crecimiento Transformador beta/metabolismo
4.
Sci Rep ; 9(1): 13446, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31530856

RESUMEN

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.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Péptidos/química , Péptidos/farmacología , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Animales , Sitios de Unión , Línea Celular , Células Endoteliales , Factor 2 de Diferenciación de Crecimiento/química , Factor 2 de Diferenciación de Crecimiento/metabolismo , Humanos , Ratones , Imitación Molecular , Péptidos/metabolismo , Fosforilación , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad8/metabolismo , Relación Estructura-Actividad
5.
J Biol Chem ; 294(16): 6333-6343, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30814254

RESUMEN

Growth differentiation factor 8 (GDF8; also known as myostatin) and GDF11 are closely related members of the transforming growth factor ß (TGF-ß) family. GDF8 strongly and negatively regulates skeletal muscle growth, and GDF11 has been implicated in various age-related pathologies such as cardiac hypertrophy. GDF8 and GDF11 signaling activities are controlled by the extracellular protein antagonists follistatin; follistatin-like 3 (FSTL3); and WAP, follistatin/kazal, immunoglobulin, Kunitz, and netrin domain-containing (WFIKKN). All of these proteins contain a follistatin domain (FSD) important for ligand binding and antagonism. Here, we investigated the structure and function of the FSD from murine WFIKKN2 and compared it with the FSDs of follistatin and FSTL3. Using native gel shift and surface plasmon resonance analyses, we determined that the WFIKKN2 FSD can interact with both GDF8 and GDF11 and block their interactions with the type II receptor activin A receptor type 2B (ActRIIB). Further, we solved the crystal structure of the WFIKKN2 FSD to 1.39 Å resolution and identified surface-exposed residues that, when substituted with alanine, reduce antagonism of GDF8 in full-length WFIKKN2. Comparison of the WFIKKN2 FSD with those of follistatin and FSTL3 revealed differences in both the FSD structure and position of residues within the domain that are important for ligand antagonism. Taken together, our results indicate that both WFIKKN and follistatin utilize their FSDs to block the type II receptor but do so via different binding interactions.


Asunto(s)
Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Portadoras/química , Factores de Diferenciación de Crecimiento/antagonistas & inhibidores , Miostatina/antagonistas & inhibidores , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Animales , Proteínas Morfogenéticas Óseas/química , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras/metabolismo , Cristalografía por Rayos X , Proteínas Relacionadas con la Folistatina/química , Proteínas Relacionadas con la Folistatina/metabolismo , Factores de Diferenciación de Crecimiento/química , Factores de Diferenciación de Crecimiento/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Miostatina/química , Miostatina/metabolismo , Resonancia por Plasmón de Superficie
7.
Drug Test Anal ; 9(11-12): 1721-1730, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28929587

RESUMEN

Luspatercept (ACE-536, ACVR2B-Fc), a fusion protein consisting of the extracellular domain of ActRIIB receptor and the Fc-part of human immunoglobulin G1 (IgG1), is currently under clinical development (Phase III). It stimulates the formation of red blood cells and hence may be misused by athletes for doping purposes in the future. Several antibody-based strategies for the detection of Luspatercept and other ACVR2B-Fc fusion proteins in human serum were evaluated (ELISA; IEF-, SDS-, and SAR-PAGE followed by Western blotting; immunoprecipitation). Two methods led to useful results: a commercial "soluble" ACTR-IIB ELISA, which also detected Luspatercept and other ACVR2B-Fc's, but showed no cross-reactivity with Sotatercept/ACVR2A-Fc's. The ELISA might be applied as fast screening tool (100 µL serum; limit of detection (LOD) ca 15.6 ng/mL). The second method uses a polyclonal ACVR2B-antibody for immunoprecipitation followed by SAR-PAGE and Western blotting with a monoclonal detection antibody (50 µL serum; LOD ca 1.0 ng/mL). It can be used for initial as well as for confirmatory testing. Due to the high doses (mg/kg) and long serum half-life of Luspatercept, both strategies may be useful in anti-doping control in the future. Copyright © 2017 John Wiley & Sons, Ltd.


Asunto(s)
Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Activinas/análisis , Anticuerpos Monoclonales/química , Fragmentos Fc de Inmunoglobulinas/análisis , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/química , Activinas/química , Activinas/metabolismo , Western Blotting , Doping en los Deportes , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/metabolismo , Límite de Detección , Proteínas Recombinantes de Fusión/metabolismo
8.
J Biol Chem ; 292(33): 13809-13822, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28659341

RESUMEN

Bone and skeletal muscle mass are highly correlated in mammals, suggesting the existence of common anabolic signaling networks that coordinate the development of these two anatomically adjacent tissues. The activin signaling pathway is an attractive candidate to fulfill such a role. Here, we generated mice with conditional deletion of activin receptor (ACVR) type 2A, ACVR2B, or both, in osteoblasts, to determine the contribution of activin receptor signaling in regulating bone mass. Immunohistochemistry localized ACVR2A and ACVR2B to osteoblasts and osteocytes. Primary osteoblasts expressed activin signaling components, including ACVR2A, ACVR2B, and ACVR1B (ALK4) and demonstrated increased levels of phosphorylated Smad2/3 upon exposure to activin ligands. Osteoblasts lacking ACVR2B did not show significant changes in vitro However, osteoblasts deficient in ACVR2A exhibited enhanced differentiation indicated by alkaline phosphatase activity, mineral deposition, and transcriptional expression of osterix, osteocalcin, and dentin matrix acidic phosphoprotein 1. To investigate activin signaling in osteoblasts in vivo, we analyzed the skeletal phenotypes of mice lacking these receptors in osteoblasts and osteocytes (osteocalcin-Cre). Similar to the lack of effect in vitro, ACVR2B-deficient mice demonstrated no significant change in any bone parameter. By contrast, mice lacking ACVR2A had significantly increased femoral trabecular bone volume at 6 weeks of age. Moreover, mutant mice lacking both ACVR2A and ACVR2B demonstrated sustained increases in trabecular bone volume, similar to those in ACVR2A single mutants, at 6 and 12 weeks of age. Taken together, these results indicate that activin receptor signaling, predominantly through ACVR2A, directly and negatively regulates bone mass in osteoblasts.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Osteoblastos/metabolismo , Osteocitos/metabolismo , Osteogénesis , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Animales , Animales Recién Nacidos , Desarrollo Óseo , Proliferación Celular , Células Cultivadas , Cruzamientos Genéticos , Femenino , Fémur , Eliminación de Gen , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Desarrollo de Músculos , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Mutación , Especificidad de Órganos , Osteoblastos/citología , Osteocitos/citología , Cráneo
9.
J Med Chem ; 60(4): 1495-1508, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28103025

RESUMEN

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.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Lactonas/química , Lactonas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Resorcinoles/química , Resorcinoles/farmacología , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Secuencia de Aminoácidos , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Alineación de Secuencia , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo
10.
Cancer Sci ; 108(1): 151-155, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28133920

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/farmacología , Receptores de Activinas Tipo II/uso terapéutico , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Femenino , Factor 2 de Diferenciación de Crecimiento/antagonistas & inhibidores , Factor 2 de Diferenciación de Crecimiento/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/farmacología , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Pancreáticas/irrigación sanguínea , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/química , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Artículo en Inglés | MEDLINE | ID: mdl-27575753

RESUMEN

Activin receptor type IIA (ActRIIA), a transmembrane serine/threonine kinase receptor is an important regulator of physiological traits, viz., reproduction and body growth in vertebrates including teleosts. However, existing knowledge of its role in regulating fish physiology is limited. To address this, we have cloned and characterized the ActRIIA cDNA of Labeo rohita (rohu), an economically important fish species of the Indian subcontinent. Comparative expression profiling of the receptor gene at various reproductive and growth stages supports to its role in promoting oocyte maturation, spermatogenesis and skeletal muscle development via interaction with multiple ligands of transforming growth factor-ß (TGF-ß) family. The full-length cDNA of rohu ActRIIA was found to be of 1587bp length encoding 528 amino acids. The three-dimensional structure of the intracellular kinase domain of rohu ActRIIA has also been predicted. Phylogenetic relationship studies showed that the gene is evolutionarily conserved across the vertebrate lineage implicating that the functioning of the receptor is more or less similar in vertebrates. Taken together, these findings could be an initial step towards the use of ActRIIA as a potential candidate gene marker for understanding the complex regulatory mechanism of fish reproduction and growth.


Asunto(s)
Receptores de Activinas Tipo II/genética , Carpas/crecimiento & desarrollo , Carpas/genética , Proteínas de Peces/genética , Regulación del Desarrollo de la Expresión Génica , Ovario/crecimiento & desarrollo , Testículo/crecimiento & desarrollo , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Evolución Molecular , Femenino , Proteínas de Peces/química , Proteínas de Peces/metabolismo , Humanos , Masculino , Ratones , Modelos Moleculares , Ovario/metabolismo , Filogenia , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Análisis de Secuencia de ADN , Testículo/metabolismo
12.
PLoS One ; 11(11): e0166803, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27870893

RESUMEN

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.


Asunto(s)
Receptores de Activinas Tipo II/genética , Músculo Esquelético/patología , Atrofia Muscular Espinal/terapia , Miostatina/antagonistas & inhibidores , Péptidos/genética , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/química , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Masculino , Ratones , Contracción Muscular , Músculo Esquelético/fisiopatología , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/fisiopatología , Miostatina/genética , Tamaño de los Órganos , Péptidos/farmacología , Fenotipo
13.
Drug Test Anal ; 8(11-12): 1152-1161, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27649383

RESUMEN

Sotatercept (formerly ACE-011) is a glycosylated, dimeric fusion protein composed of the extracellular domain of the human activin receptor type IIA (ActRIIA) and the Fc region of human IgG1. The protein-based drug candidate acts as a ligand trap which competitively binds to activin A and other members of the transforming growth factor beta superfamily, thus blocking signalling through ActRIIA. Since the inhibition of activin A was found to significantly increase bone formation and quality, Sotatercept was originally developed for the treatment of diseases involving bone loss. But as the protein therapeutic also stimulates erythropoiesis by a mechanism independent of the EPO receptor, it has been evaluated for the treatment of anaemia in rare blood diseases such as beta thalassemia. Due to its positive effects on erythropoiesis and bone formation, Sotatercept may also be misused as performance-enhancing agent in sports. Within this study, two complementary detection assays for Sotatercept and related ActRIIA-Fc fusion proteins in serum samples were developed. While the first assay combines affinity purification and Western blotting to generically detect ActRIIA-Fc fusion proteins irrespective of their amino acid sequence, the liquid chromatography-high resolution mass spectrometry (LC-HRMS) method is highly specific for proteolytic peptides originating from the receptor and Fc domain of Sotatercept. Both approaches can readily be modified to include other pharmaceutical proteins such as therapeutic antibodies, and serve as proof-of-concept for the capability of the approach to detect TGF-ß inhibitors and Fc fusion proteins in doping control serum samples. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Receptores de Activinas Tipo II/química , Western Blotting/métodos , Eritropoyesis/efectos de los fármacos , Cromatografía de Gases y Espectrometría de Masas/métodos , Proteínas Recombinantes de Fusión/química , Factor de Crecimiento Transformador beta/análisis , Doping en los Deportes , Humanos , Proteínas Recombinantes de Fusión/análisis , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/química
14.
J Biol Chem ; 291(7): 3395-410, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26677222

RESUMEN

By non-covalent association after proteolytic cleavage, the pro-domains modulate the activities of the mature growth factor domains across the transforming growth factor-ß family. In the case of bone morphogenic protein 9 (BMP9), however, the pro-domains do not inhibit the bioactivity of the growth factor, and the BMP9·pro-domain complexes have equivalent biological activities as the BMP9 mature ligand dimers. By using real-time surface plasmon resonance, we could demonstrate that either binding of pro-domain-complexed BMP9 to type I receptor activin receptor-like kinase 1 (ALK1), type II receptors, co-receptor endoglin, or to mature BMP9 domain targeting antibodies leads to immediate and complete displacement of the pro-domains from the complex. Vice versa, pro-domain binding by an anti-pro-domain antibody results in release of the mature BMP9 growth factor. Based on these findings, we adjusted ELISA assays to measure the protein levels of different BMP9 variants. Although mature BMP9 and inactive precursor BMP9 protein were directly detectable by ELISA, BMP9·pro-domain complex could only be measured indirectly as dissociated fragments due to displacement of mature growth factor and pro-domains after antibody binding. Our studies provide a model in which BMP9 can be readily activated upon getting into contact with its receptors. This increases the understanding of the underlying biology of BMP9 activation and also provides guidance for ELISA development for the detection of circulating BMP9 variants.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Antígenos CD/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Modelos Moleculares , Receptores de Superficie Celular/metabolismo , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Animales , Antígenos CD/química , Antígenos CD/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/química , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Células Cultivadas , Dimerización , Endoglina , Femenino , Factor 2 de Diferenciación de Crecimiento/sangre , Factor 2 de Diferenciación de Crecimiento/aislamiento & purificación , Factor 2 de Diferenciación de Crecimiento/metabolismo , Factores de Diferenciación de Crecimiento/sangre , Factores de Diferenciación de Crecimiento/química , Factores de Diferenciación de Crecimiento/genética , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones Endogámicos BALB C , Fragmentos de Péptidos/agonistas , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/aislamiento & purificación , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Precursores de Proteínas/sangre , Precursores de Proteínas/química , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transducción de Señal , Organismos Libres de Patógenos Específicos
15.
Placenta ; 36(4): 345-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25659497

RESUMEN

INTRODUCTION: Pre-eclampsia is a pregnancy-specific disorder and characterized by reduced trophoblast invasion and reduced spiral artery remodeling in the first trimester placenta. A polymorphism located in the promoter region of ACVR2A (rs1424954 (A > G)) has previously been shown to be significantly associated with pre-eclampsia. METHODS: The effects of this variant on ACVR2A expression and its function in the Activin-A signaling pathway were studied by transfections in SGHPL-5 extravillous trophoblasts followed by qRT-PCR. RESULTS: Here we show that the ACVR2A promoter susceptibility variant causes a downregulation of ACVR2A expression. We also provide evidence for transcription of a so-called PROMPT (PROMoter-uPstream-Transcript) in the opposite direction of ACVR2A, containing the polymorphism, and downregulated when the susceptibility allele is carried, which either shares the same promoter as ACVR2A or is a non-coding RNA that is able to enhance ACVR2A transcription. Furthermore, when the effect of the susceptibility variant is mimicked by knockdown of ACVR2A, physiologic concentrations of Activin-A cause a reduction in NODAL mRNA expression in the SGHPL-5 trophoblasts, indicative of a protective effect as reduction in NODAL expression is associated with an increase in trophoblast invasion. However, at pathologic levels of Activin-A, as found in pre-eclampsia, this effect is no longer seen, and we show this is potentially caused by a lack of downregulation of ACVR2B. DISCUSSION: The combined data suggest a double hit phenomenon in which the first hit, the promoter variant, together with the second hit, pathological levels of Activin-A, lead to high levels of NODAL, associated with reduced trophoblast invasion and observed in pre-eclamptic placentas.


Asunto(s)
Receptores de Activinas Tipo II/genética , Activinas/metabolismo , Regulación hacia Abajo , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Transducción de Señal , Trofoblastos/metabolismo , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Alelos , Línea Celular , Exones , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteína Nodal/antagonistas & inhibidores , Proteína Nodal/genética , Proteína Nodal/metabolismo , Fragmentos de Péptidos/agonistas , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Preeclampsia/genética , Preeclampsia/metabolismo , Embarazo , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes/metabolismo
16.
Am J Hematol ; 90(1): 8-14, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25236856

RESUMEN

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.


Asunto(s)
Eritropoyesis/efectos de los fármacos , Hemoglobinas/análisis , Hepcidinas/genética , Hierro/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Receptores de Activinas Tipo II/química , Animales , Transporte Biológico , Recuento de Células Sanguíneas , Células Precursoras Eritroides/efectos de los fármacos , Eritropoyetina/farmacología , Femenino , Inmunoglobulina G/química , Hierro/sangre , Ligandos , Ratones Endogámicos C57BL , Ratones Transgénicos , Bazo/metabolismo
17.
J Biol Chem ; 290(6): 3390-404, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25368322

RESUMEN

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.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Diferenciación Celular , Mioblastos/efectos de los fármacos , Miostatina/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal , Receptores de Activinas Tipo II/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Humanos , Ratones , Datos de Secuencia Molecular , Mioblastos/citología , Mioblastos/metabolismo , Unión Proteica , Células Sf9 , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Spodoptera , Factores de Transcripción/metabolismo
18.
Stem Cell Reports ; 3(6): 1103-17, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25458897

RESUMEN

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.


Asunto(s)
Receptores de Activinas Tipo II/genética , Daño del ADN , Reparación del ADN , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , MicroARNs/genética , Recombinasa Rad51/genética , Regiones no Traducidas 3' , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Proteínas de Ciclo Celular/genética , Diferenciación Celular/genética , Proliferación Celular , Roturas del ADN , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Ratones , MicroARNs/química , Interferencia de ARN , ARN Mensajero/química , ARN Mensajero/genética , Recombinasa Rad51/química , Transducción de Señal
19.
Cell Mol Life Sci ; 71(16): 3165-72, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24337809

RESUMEN

The bone morphogenetic protein (BMP) signaling pathway is essential for normal development and tissue homeostasis. BMP signal transduction occurs when ligands interact with a complex of type 1 and type 2 receptors to activate downstream transcription factors. It is well established that a single BMP receptor may bind multiple BMP ligands with varying affinity, and this has been largely attributed to conformation at the amino acid level. However, all three type 2 BMP receptors (BMPR2, ACVR2A/B) contain consensus N-glycosylation sites in their extracellular domains (ECDs), which could play a role in modulating interaction with ligand. Here, we show a differential pattern of N-glycosylation between BMPR2 and ACVR2A/B. Site-directed mutagenesis reveals that BMPR2 is uniquely glycosylated near its ligand binding domain and at a position that is mutated in patients with heritable pulmonary arterial hypertension. We further demonstrate using a cell-free pulldown assay that N-glycosylation of the BMPR2-ECD enhances its ability to bind BMP2 ligand but has no impact on binding by the closely-related ACVR2B. Our results illuminate a novel aspect of BMP signaling pathway mechanics and demonstrate a functional difference resulting from post-translational modification of type 2 BMP receptors. Additionally, since BMPR2 is required for several aspects of normal development and defects in its function are strongly implicated in human disease, our findings are likely to be relevant in several biological contexts in normal and abnormal human physiology.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/química , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Línea Celular , Hipertensión Pulmonar Primaria Familiar , Glicosilación , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Terciaria de Proteína
20.
PLoS One ; 8(7): e69535, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936038

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/metabolismo , Apoptosis/efectos de los fármacos , Comunicación Autocrina/efectos de los fármacos , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Carcinoma Hepatocelular/patología , Adhesión Celular/efectos de los fármacos , Recuento de Células , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factor 2 de Diferenciación de Crecimiento , Factores de Diferenciación de Crecimiento/farmacología , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Neoplasias Hepáticas/patología , Estructura Terciaria de Proteína , Suero , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...