Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 255
Filtrar
1.
Int J Mol Sci ; 22(18)2021 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-34576257

RESUMEN

Although anti-Müllerian hormone (AMH) has classically been correlated with the regression of Müllerian ducts in male mammals, involvement of this growth factor in other reproductive processes only recently come to light. Teleost is the only gnathostomes that lack Müllerian ducts despite having amh orthologous genes. In adult teleost gonads, Amh exerts a role in the early stages of germ cell development in both males and females. Mechanisms involving the interaction of Amh with gonadotropin- and growth factor-induced functions have been proposed, but our overall knowledge regarding Amh function in fish gonads remains modest. In this study, we report on Amh actions in the European sea bass ovary. Amh and type 2 Amh receptor (Amhr2) are present in granulosa and theca cells of both early and late-vitellogenic follicles and cannot be detected in previtellogenic ovaries. Using the Pichia pastoris system a recombinant sea bass Amh has been produced that is endogenously processed to generate a 12-15 kDa bioactive mature protein. Contrary to previous evidence in lower vertebrates, in explants of previtellogenic sea bass ovaries, mature Amh has a synergistic effect on steroidogenesis induced by the follicle-stimulating hormone (Fsh), increasing E2 and cyp19a1a levels.


Asunto(s)
Hormona Antimülleriana/química , Hormona Folículo Estimulante/metabolismo , Ovario/metabolismo , Receptores de Péptidos/química , Receptores de Factores de Crecimiento Transformadores beta/química , Proteínas Recombinantes/química , Animales , Hormona Antimülleriana/metabolismo , Lubina , Células COS , Chlorocebus aethiops , Estradiol/metabolismo , Femenino , Gonadotropinas/metabolismo , Gónadas/metabolismo , Células de la Granulosa/metabolismo , Inmunoensayo , Folículo Ovárico/metabolismo , Plásmidos/metabolismo , Esteroides/metabolismo , Células Tecales/metabolismo , Vitelogénesis
2.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34155118

RESUMEN

Anti-Müllerian hormone (AMH), or Müllerian-inhibiting substance, is a protein hormone that promotes Müllerian duct regression during male fetal sexual differentiation and regulation of folliculogenesis in women. AMH is a member of the transforming growth factor beta (TGF-ß) family, which has evolved to signal through its own dedicated type II receptor, AMH receptor type II (AMHR2). Structures of other TGF-ß family members have revealed how ligands infer specificity for their cognate receptors; however, it is unknown how AMH binds AMHR2 at the molecular level. Therefore, in this study, we solved the X-ray crystal structure of AMH bound to the extracellular domain of AMHR2 to a resolution of 2.6Å. The structure reveals that while AMH binds AMHR2 in a similar location to Activin and BMP ligand binding to their type II receptors, differences in both AMH and AMHR2 account for a highly specific interaction. Furthermore, using an AMH responsive cell-based luciferase assay, we show that a conformation in finger 1 of AMHR2 and a salt bridge formed by K534 on AMH and D81/E84 of AMHR2 are key to the AMH/AMHR2 interaction. Overall, our study highlights how AMH engages AMHR2 using a modified paradigm of receptor binding facilitated by modifications to the three-finger toxin fold of AMHR2. Furthermore, understanding these elements contributing to the specificity of binding will help in the design of agonists or antagonists or the selection of antibody therapies.


Asunto(s)
Hormona Antimülleriana/química , Hormona Antimülleriana/metabolismo , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Activinas/química , Secuencia de Aminoácidos , Proteínas Morfogenéticas Óseas/química , Cristalografía por Rayos X , Modelos Moleculares , Receptores de Péptidos/química , Receptores de Factores de Crecimiento Transformadores beta/química , Homología Estructural de Proteína
3.
J Mater Sci Mater Med ; 31(8): 62, 2020 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-32696084

RESUMEN

The aim of this study was to evaluate the effect of biofunctionalization with two TGF-ß1 inhibitor peptides, P17 and P144, on osseointegration of CP-Ti dental implants. A total of 36 implants (VEGA, Klockner®) with 3.5 × 8 mm internal connection were used in this study, divided in three groups: (1) control group (n = 12), (2) implants which surfaces were biofunctionalized with P17 peptide inhibitor (n = 12), (3) implants with surfaces biofunctionalized by P144 peptide (n = 12). Three implants, one from each group, were inserted in both hemimandibles of 6 beagle dogs, 2 months after tooth extraction. Two animals were sacrificed at 2, 4 and 8 weeks post implant insertion, respectively. The samples were analyzed by Backscattering Scanning Electron Microscopy (BS-SEM) and histological analysis. Histomorphometric analysis of bone to implant contact (BIC), peri-implant bone fraction (BF) and interthread bone (IB) were carried out. Bone formation around implants measured by quantitative analysis, BS-SEM, was significantly higher in the P17-biofunctionalized implants, 4 and 8 weeks after the implantation. Histomorphometric analysis of BIC, BF and IB showed higher values in the P17-biofunctionalized group at initial stages of healing (2 weeks) and early osseointegration both at 4 and 8 weeks. For P144 biofunctionalized implants, the histomorphometric values obtained are also higher than control group. Accordingly, better results in the experimental groups were proven both by the quantitative and the qualitative analysis. Surface biofunctionalization with TGF-ß1 inhibitor peptides, P17 and P144, resulted in better quantitative and qualitative parameters relative to implant osseointegration.


Asunto(s)
Materiales Biocompatibles Revestidos/farmacología , Implantes Dentales , Oseointegración/efectos de los fármacos , Fragmentos de Péptidos/química , Péptidos/química , Receptores de Factores de Crecimiento Transformadores beta/química , Animales , Materiales Biocompatibles Revestidos/química , Implantación Dental Endoósea , Diseño de Prótesis Dental , Perros , Implantes Experimentales , Masculino , Mandíbula/patología , Mandíbula/cirugía , Ensayo de Materiales , Osteogénesis/efectos de los fármacos , Propiedades de Superficie/efectos de los fármacos , Factor de Crecimiento Transformador beta1/química
4.
Fish Shellfish Immunol ; 105: 41-52, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32629101

RESUMEN

Transforming growth factor-ß type III receptor (TßR3), as a co-receptor of TGF-ß superfamily, plays critical roles in development and growth as well as some disease pathogeneses by presenting ligands to other receptors in vertebrates. However, the identification and functional characterization of TßR3 had not been reported yet in invertebrates. In the present study, TßR3 was first identified and characterized in mud crab Scylla paramamosain. The obtained cDNA length of SpTßR3 was 2, 424 bp with a 1, 854 bp open reading frame, which encoded a putative peptide of 617 amino acids containing a typical transmembrane region and a Zona pellucida (ZP) domain. Real-time PCR results showed that SpTßR3 was predominantly expressed at early embryonic development stage and early postmolt stage, suggesting its participation in development and growth. We report, for the first time in invertebrates, the challenge of both Vibro alginolyticus and Poly (I:C) could alter the expression patterns of SpTßR3. Notably, the expression levels of SpIKK, two NF-κB members (SpRelish and SpDorsal), and five antimicrobial peptide genes (SpCrustin and SpALF1-4) were significantly suppressed when SpTßR3 was interfered in vivo. Secondly, the overexpression of SpTßR3 in vitro could activate NF-κB signaling through the dual-luciferase reporter assays. Furthermore, the bacterial clearance assay after SpTßR3 was silenced in vivo highlighted the potential of SpTßR3 in activating the innate immune responses. These results implied the involvement of SpTßR3 in the innate immune responses by regulating the NF-κB pathway. This study first indicated that TßR3 was present in invertebrate, and it participated in not only the development and growth but also the innate immunity of S. paramamosain. It also provided new insights into the origin or evolution of TGF-ß receptors in crustacean species and even in invertebrates.


Asunto(s)
Braquiuros/genética , Braquiuros/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Transducción de Señal/genética , Secuencia de Aminoácidos , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Proteínas de Artrópodos/inmunología , Secuencia de Bases , Perfilación de la Expresión Génica , Filogenia , Poli I-C/farmacología , Receptores de Factores de Crecimiento Transformadores beta/química , Alineación de Secuencia , Vibrio alginolyticus/fisiología
5.
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
7.
Proc Natl Acad Sci U S A ; 116(31): 15505-15513, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31315975

RESUMEN

TGFß family ligands, which include the TGFßs, BMPs, and activins, signal by forming a ternary complex with type I and type II receptors. For TGFßs and BMPs, structures of ternary complexes have revealed differences in receptor assembly. However, structural information for how activins assemble a ternary receptor complex is lacking. We report the structure of an activin class member, GDF11, in complex with the type II receptor ActRIIB and the type I receptor Alk5. The structure reveals that receptor positioning is similar to the BMP class, with no interreceptor contacts; however, the type I receptor interactions are shifted toward the ligand fingertips and away from the dimer interface. Mutational analysis shows that ligand type I specificity is derived from differences in the fingertips of the ligands that interact with an extended loop specific to Alk4 and Alk5. The study also reveals differences for how TGFß and GDF11 bind to the same type I receptor, Alk5. For GDF11, additional contacts at the fingertip region substitute for the interreceptor interactions that are seen for TGFß, indicating that Alk5 binding to GDF11 is more dependent on direct contacts. In support, we show that a single residue of Alk5 (Phe84), when mutated, abolishes GDF11 signaling, but has little impact on TGFß signaling. The structure of GDF11/ActRIIB/Alk5 shows that, across the TGFß family, different mechanisms regulate type I receptor binding and specificity, providing a molecular explanation for how the activin class accommodates low-affinity type I interactions without the requirement of cooperative receptor interactions.


Asunto(s)
Activinas/química , Activinas/metabolismo , Complejos Multiproteicos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Humanos , Ratones , Modelos Moleculares , Complejos Multiproteicos/química , Ratas , Factor de Crecimiento Transformador beta/metabolismo
8.
PLoS One ; 14(5): e0216628, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31071172

RESUMEN

The transforming growth factor-ß (TGFß) family plays an important role in many developmental processes and when mutated often contributes to various diseases. Marfan syndrome is a genetic disease with an occurrence of approximately 1 in 5,000. The disease is caused by mutations in fibrillin, which lead to an increase in TGFß ligand activity, resulting in abnormalities of connective tissues which can be life-threatening. Mutations in other components of TGFß signaling (receptors, Smads, Schnurri) lead to similar diseases with attenuated phenotypes relative to Marfan syndrome. In particular, mutations in TGFß receptors, most of which are clustered at the C-terminal end, result in Marfan-like (MFS-like) syndromes. Even though it was assumed that many of these receptor mutations would reduce or eliminate signaling, in many cases signaling is active. From our previous studies on receptor trafficking in C. elegans, we noticed that many of these receptor mutations that lead to Marfan-like syndromes overlap with mutations that cause mis-trafficking of the receptor, suggesting a link between Marfan-like syndromes and TGFß receptor trafficking. To test this hypothesis, we introduced three of these key MFS and MFS-like mutations into the C. elegans TGFß receptor and asked if receptor trafficking is altered. We find that in every case studied, mutated receptors mislocalize to the apical surface rather than basolateral surface of the polarized intestinal cells. Further, we find that these mutations result in longer animals, a phenotype due to over-stimulation of the nematode TGFß pathway and, importantly, indicating that function of the receptor is not abrogated in these mutants. Our nematode models of Marfan syndrome suggest that MFS and MFS-like mutations in the type II receptor lead to mis-trafficking of the receptor and possibly provides an explanation for the disease, a phenomenon which might also occur in some cancers that possess the same mutations within the type II receptor (e.g. colon cancer).


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Mutación Missense , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Animales Modificados Genéticamente , Proteínas de Caenorhabditis elegans/química , Modelos Animales de Enfermedad , Humanos , Dominios Proteicos , Receptor Tipo II de Factor de Crecimiento Transformador beta/química , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad de la Especie
9.
J Biol Chem ; 294(9): 3065-3080, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30598510

RESUMEN

Betaglycan (BG) is a membrane-bound co-receptor of the TGF-ß family that selectively binds transforming growth factor-ß (TGF-ß) isoforms and inhibin A (InhA) to enable temporal-spatial patterns of signaling essential for their functions in vivo Here, using NMR titrations of methyl-labeled TGF-ß2 with BG's C-terminal binding domain, BGZP-C, and surface plasmon resonance binding measurements with TGF-ß2 variants, we found that the BGZP-C-binding site on TGF-ß2 is located on the inner surface of its extended finger region. Included in this binding site are Ile-92, Lys-97, and Glu-99, which are entirely or mostly specific to the TGF-ß isoforms and the InhA α-subunit, but they are unconserved in other TGF-ß family growth factors (GFs). In accord with the proposed specificity-determining role of these residues, BG bound bone morphogenetic protein 2 (BMP-2) weakly or not at all, and TGF-ß2 variants with the corresponding residues from BMP-2 bound BGZP-C more weakly than corresponding alanine variants. The BGZP-C-binding site on InhA previously was reported to be located on the outside of the extended finger region, yet at the same time to include Ser-112 and Lys-119, homologous to TGF-ß2 Ile-92 and Lys-97, on the inside of the fingers. Therefore, it is likely that both TGF-ß2 and InhA bind BGZP-C through a site on the inside of their extended finger regions. Overall, these results identify the BGZP-C-binding site on TGF-ß2 and shed light on the specificity of BG for select TGF-ß-type GFs and the mechanisms by which BG influences their signaling.


Asunto(s)
Inhibinas/metabolismo , Proteoglicanos/química , Proteoglicanos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta2/química , Factor de Crecimiento Transformador beta2/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Humanos , Concentración de Iones de Hidrógeno , Ratones , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína , Ratas , Especificidad por Sustrato
10.
Comput Biol Chem ; 76: 327-337, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30145406

RESUMEN

By using the molecular docking and 4D-QSAR analysis, it is aimed to find the interaction points in the receptor binding site of transforming growth factor-beta (TGF-beta) used to inhibit invasion and metastasis. To elucidate the interaction points of receptor, different types of local reactive descriptor (LRD) of ligands have been used. Activity values related to interaction energy between the ligand-receptor (L-R) were determined by nonlinear least squares (NLLS) using the Levenberg-Marquardt (LM) algorithm. Using the Molecule Comparative Electron Topology (MCET) method, the 3D pharmacophore model (3D-PhaM) was obtained after alignment and superimposition of the molecules, and also confirmed by molecular docking method. With the leave one out-cross validation (LOO-CV) method, the best predictions are q2 or rCV2 = 0.789 for the 51 compounds in the internal training set and r2 = 0.785 for the 13 compounds in the external test set. Furthermore, the predictive capability of the advanced QSAR model is more precisely calculated with the rm2 metric (rm2 = 0.769).


Asunto(s)
Antineoplásicos/química , Proteínas Serina-Treonina Quinasas/química , Receptores de Factores de Crecimiento Transformadores beta/química , Tiazoles/química , Algoritmos , Sitios de Unión , Humanos , Ligandos , Conformación Molecular , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad Cuantitativa , Receptor Tipo I de Factor de Crecimiento Transformador beta
11.
Comput Biol Chem ; 76: 139-150, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30015175

RESUMEN

The discovery of drugs relevant to transforming growth factor ß (TGF-ß) receptor inhibitors have been considered as a considerable challenge during therapy idiopathic pulmonary fibrosis diseases. For the first time, herein we illustrate a field-based quantitative structure-activity relationship (QSAR) model and molecular dynamics (MD) simulations for novel 7-substituted-pyrazolo [4, 3-b] pyridine derivatives with biological activity for the TGF-ß receptor, with an attempt of elucidating the 3D structural features that are essential for the activity. Results demonstrate that the field-based model (Q2 = 0.548, R2training = 0.840, R2test = 0.750) are acceptable with good predictive capabilities. In addition, MD studies were also carried out on the training set with the aim of exploring their binding modes in the active pocket of TGF-ß receptor, resulting in some of the crucial structural fragments which are responsible for inhibitory activity. Therefore, we summarized the following features required for TGF-ß receptor inhibition: electronegative in region1, bulky groups in region2 and smaller groups in region3. Based on the model and related information, we hope the above information provides an important insight for understanding the interactions of the inhibitors and TGF-ß receptor, which may be useful in discovering novel potent inhibitors.


Asunto(s)
Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Sitios de Unión , Humanos , Análisis de los Mínimos Cuadrados , Ligandos , Modelos Químicos , Simulación de Dinámica Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/química , Relación Estructura-Actividad Cuantitativa , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/química
12.
J Am Chem Soc ; 140(12): 4186-4190, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29522674

RESUMEN

Herein, we report a nongenetic and real-time approach for imaging protein dimerization on living cell surfaces by aptamer recognition and proximity-induced DNA assembly. We use the aptamer specific for the receptor monomer as a recognition probe. When receptor dimerization occurs, the dimeric receptors bring two aptamer probes into close proximity, thereby triggering dynamic DNA assembly. The proposed approach was successfully applied to visualize dimerization of Met receptor and transforming growth factor-ß type II receptor. This approach allows us to image the two states (monomer/dimer) of a receptor protein on living cell surfaces in real time, opening a universal method for further investigation of protein dimerization and the corresponding activation processes in signal transduction.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , ADN/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Aptámeros de Nucleótidos/química , Línea Celular Tumoral , ADN/química , Dimerización , Humanos , Microscopía Confocal , Proteínas Proto-Oncogénicas c-met/química , Receptores de Factores de Crecimiento Transformadores beta/química , Transducción de Señal
13.
Acta Biochim Biophys Sin (Shanghai) ; 50(1): 12-36, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29293886

RESUMEN

Transforming growth factor-ß (TGF-ß) family members, which include TGF-ßs, activins and bone morphogenetic proteins, are pleiotropic cytokines that elicit cell type-specific effects in a highly context-dependent manner in many different tissues. These secreted protein ligands signal via single-transmembrane Type I and Type II serine/threonine kinase receptors and intracellular SMAD transcription factors. Deregulation in signaling has been implicated in a broad array of diseases, and implicate the need for intricate fine tuning in cellular signaling responses. One important emerging mechanism by which TGF-ß family receptor signaling intensity, duration, specificity and diversity are regulated and/or mediated is through cell surface co-receptors. Here, we provide an overview of the co-receptors that have been identified for TGF-ß family members. While some appear to be specific to TGF-ß family members, others are shared with other pathways and provide possible ways for signal integration. This review focuses on novel functions of TGF-ß family co-receptors, which continue to be discovered.


Asunto(s)
Activinas/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Activinas/química , Animales , Proteínas Morfogenéticas Óseas/química , Humanos , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Receptores de Factores de Crecimiento Transformadores beta/química , Factor de Crecimiento Transformador beta/química
14.
Reproduction ; 154(4): 521-534, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28733348

RESUMEN

Bone morphogenetic factor 15 (BMP15) and growth differentiation factor 9 (GDF9) are oocyte-secreted factors with demonstrable effects on ovarian follicular development and ovulation rate. However, the molecular forms of BMP15 and GDF9 produced by oocytes remain unclear. The aims herein, using Western blotting (WB) procedures with specific monoclonal antibodies (mabs), were to identify the molecular forms of BMP15 and GDF9 synthesised and secreted by isolated ovine (o) and bovine (b) oocytes in vitro The mabs were known to recognise the biological forms of BMP15 or GDF9 since they had previously been shown to inhibit their bioactivities in vitro and in vivo Using recombinant variants of oBMP15 and oGDF9, including a cysteine mutant form of oBMP15 (S356C) and a human (h) BMP15:GDF9 heterodimer (cumulin), it was established that the mabs were able to identify monomeric, dimeric, promature and higher-molecular-weight forms of BMP15 and GDF9 and cumulin (GDF9 mab only). After using non-reducing, reducing and reducing + cross-linking conditions, the major oocyte-secreted forms of o and b BMP15 and GDF9 were the cleaved and uncleaved monomeric forms of the promature proteins. There was no evidence for dimeric or heterodimeric forms of either mature BMP15 or GDF9. From in silico modelling studies using transforming growth factor beta (TGFB), activin or BMP crystal templates, and both present and previously published data, a model is proposed to illustrate how the monomeric forms of BMP15 and GDF9 may interact with their type II and type I cell-surface receptors to initiate the synergistic actions of these growth factors.


Asunto(s)
Proteína Morfogenética Ósea 15/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Factor 9 de Diferenciación de Crecimiento/metabolismo , Oocitos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Animales , Sitios de Unión , Proteína Morfogenética Ósea 15/química , Proteína Morfogenética Ósea 15/genética , Receptores de Proteínas Morfogenéticas Óseas/química , Bovinos , Células Cultivadas , Femenino , Factor 9 de Diferenciación de Crecimiento/química , Factor 9 de Diferenciación de Crecimiento/genética , Ligandos , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Transformadores beta/química , Oveja Doméstica , Transducción de Señal , Relación Estructura-Actividad , Transfección
15.
Cell Physiol Biochem ; 42(1): 357-372, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28535533

RESUMEN

BACKGROUND/AIMS: Endothelial-to-mesenchymal transition (EndMT) plays significant roles under various pathological conditions including cardiovascular diseases, fibrosis, and cancer. EndMT of endothelial progenitor cells (EPCs) contributes to neointimal hyperplasia following cell therapy Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long non-coding RNA (lncRNA) that promotes metastasis and cancer. MicroRNA-145 (miR-145) is a tumor suppressor that has been reported to inhibit SMAD3-mediated epithelial-to-mesenchymal transition (EMT) of cancer cells. In the present study, we investigated the role of MALAT1 and miR-145 in EndMT of human circulating EPCs induced by transforming growth factor beta1 (TGF-ß1). METHODS: Human circulating EPCs were isolated and characterized by fluorescence-activated cell sorting (FACS). Expression levels of EndMT markers were assessed by qRT-PCR and western blotting. Alpha-smooth muscle actin (α-SMA) expression was measured by cell immunofluorescence staining. The regulatory relationship between MALAT1 and miR-145 and its target genes, TGFBR2 (TGFß receptortype II) and SMAD3 (mothers against decapentaplegic homolog 3) was analyzed using the luciferase reporter assay. RESULTS: We found that EndMT of EPCs induced by TGF-ß1 is accompanied by increased MALAT1 expression and decreased miR-145 expression, and MALAT1 and miR-145 directly bind and reciprocally repress each other in these cells. Dual-Luciferase Reporter assay indicated that miR-145 inhibits TGF-ß1-induced EndMT by directly targeting TGFBR2 and SMAD3. CONCLUSIONS: MALAT1 modulates TGF-ß1-induced EndMT of EPCs through regulation of TGFBR2 and SMAD3 via miR-145. Thus, the MALAT1-miR-145-TGFBR2/SMAD3 signaling pathway plays a key role in TGF-ß1-induced EndMT.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Regiones no Traducidas 3' , Antagomirs/metabolismo , Secuencia de Bases , Células Cultivadas , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Células Progenitoras Endoteliales/citología , Humanos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Alineación de Secuencia , Transducción de Señal , Proteína smad3/química , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
16.
Bioengineered ; 8(5): 600-612, 2017 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-28281868

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) signaling is involved in cell metabolism, growth, differentiation, carcinoma invasion and fibrosis development, which suggests TGF-ß1 can be treated as a therapeutic target extensively. Because TGF-ß1 receptor type α(TGFBR2) is the directed and essential mediator for TGF-ß1 signals, the extracellular domain of TGFBR2 (eTGFBR2), binding partner for TGF-ß1, has been produced in a series of expression systems to inhibit TGF-ß1 signaling. However, eTGFBR2 is unstable with a short half-life predominantly because of enzymatic degradation and kidney clearance. In this study, a fusion protein consisting of human eTGFBR2 fused at the C-terminal of human serum albumin (HSA) was stably and highly expressed in Chinese Hamster Ovary (CHO) cells. The high and stable expression sub-clones with Ig kappa signal peptide were selected by Western blot analysis and used for suspension culture. After fed-batch culture over 8 d, the expression level of HSA-eTGFBR2 reached 180 mg/L. The fusion protein was then purified from culture medium using a 2-step chromatographic procedure that resulted in 39% recovery rate. The TGF-ß1 binding assay revealed that HSA-eTGFBR2 could bind to TGF-ß1 with the affinity constant (KD of 1.42 × 10-8 M) as determined by the ForteBio Octet System. In addition, our data suggested that HSA-eTGFBR2 exhibited a TGF-ß1 neutralizing activity and maintained a long-term activity more than eTGFBR2. It concluded that the overexpressing CHO cell line supplied sufficient recombinant human HSA-eTGFBR2 for further research and other applications.


Asunto(s)
Ingeniería de Proteínas/métodos , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/química , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/química , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Albúmina Sérica/biosíntesis , Albúmina Sérica/química , Animales , Sitios de Unión , Células CHO , Cricetulus , Mejoramiento Genético/métodos , Humanos , Unión Proteica , Receptor Tipo II de Factor de Crecimiento Transformador beta , Proteínas Recombinantes de Fusión/genética , Albúmina Sérica/genética
17.
PLoS One ; 12(2): e0170822, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28182693

RESUMEN

Variants in the TGFBR2 kinase domain cause several human diseases and can increase propensity for cancer. The widespread application of next generation sequencing within the setting of Individualized Medicine (IM) is increasing the rate at which TGFBR2 kinase domain variants are being identified. However, their clinical relevance is often uncertain. Consequently, we sought to evaluate the use of molecular modeling and molecular dynamics (MD) simulations for assessing the potential impact of variants within this domain. We documented the structural differences revealed by these models across 57 variants using independent MD simulations for each. Our simulations revealed various mechanisms by which variants may lead to functional alteration; some are revealed energetically, while others structurally or dynamically. We found that the ATP binding site and activation loop dynamics may be affected by variants at positions throughout the structure. This prediction cannot be made from the linear sequence alone. We present our structure-based analyses alongside those obtained using several commonly used genomics-based predictive algorithms. We believe the further mechanistic information revealed by molecular modeling will be useful in guiding the examination of clinically observed variants throughout the exome, as well as those likely to be discovered in the near future by clinical tests leveraging next-generation sequencing through IM efforts.


Asunto(s)
Simulación de Dinámica Molecular , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Dominios Proteicos , Estructura Secundaria de Proteína , Receptor Tipo II de Factor de Crecimiento Transformador beta
18.
Amino Acids ; 49(2): 283-290, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27826791

RESUMEN

The human transforming growth factor ß (TGF-ß)/bone morphogenic protein (BMP) signaling has been recognized as an attractive target to suppress fibroblast activation in osteonecrosis of the femoral head (ONFH). Here, we reported successful derivation of a self-inhibitory peptide from the crystal complex interface of TGF-ß with its cognate receptor TßRI using rational molecular design and in vitro binding assay. Computational modeling suggested that the peptide possesses a large flexibility and would incur considerable entropy penalty. To minimize the entropy effect, the peptide was extended and cyclized to obtain a modified version of cyclic peptide. Molecular dynamics (MD) simulations revealed that the cyclic peptide exhibits larger rigidity and lower thermal motion in unbound state as compared to its linear counterpart, thus causing less entropy penalty upon binding to TGF-ß. The computational findings were then substantiated by fluorescence polarization (FP) assays, that is, no binding affinity was detected for linear peptide (K d = n.d.), while cyclic version was determined to have a moderate affinity (K d = 76 ± 18 µM). Structural and energetic analysis identified two anchor residues Phe60 and Ser65 in cyclic peptide that can form a π-π stacking and a hydrogen bonding with the residues Trp30 and His68 of TGF-ß, respectively, conferring high stability and specificity to the complex system.


Asunto(s)
Necrosis de la Cabeza Femoral/tratamiento farmacológico , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Ciclización , Diseño de Fármacos , Entropía , Necrosis de la Cabeza Femoral/metabolismo , Polarización de Fluorescencia , Humanos , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Terapia Molecular Dirigida , Conformación Proteica , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/química
19.
Anal Biochem ; 508: 97-103, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27365221

RESUMEN

Half-maximal inhibitory concentration (IC50) is the most widely used and informative measure of a drug's efficacy. It indicates how much drug is needed to inhibit a biological process by half, thus providing a measure of potency of an antagonist drug in pharmacological research. Most approaches to determine IC50 of a pharmacological compound are based on assays that utilize whole cell systems. While they generally provide outstanding potency information, results can depend on the experimental cell line used and may not differentiate a compound's ability to inhibit specific interactions. Here we show using the secreted Transforming Growth Factor-ß (TGF-ß) family ligand BMP-4 and its receptors as example that surface plasmon resonance can be used to accurately determine IC50 values of individual ligand-receptor pairings. The molecular resolution achievable wih this approach can help distinguish inhibitors that specifically target individual complexes, or that can inhibit multiple functional interactions at the same time.


Asunto(s)
Técnicas de Química Analítica/métodos , Receptores de Factores de Crecimiento Transformadores beta/química , Factor de Crecimiento Transformador beta1/química , Bioensayo , Proteína Morfogenética Ósea 4/química , Proteína Morfogenética Ósea 4/metabolismo , Humanos , Concentración 50 Inhibidora , Unión Proteica , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
20.
J Pharmacol Exp Ther ; 358(3): 423-30, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27340210

RESUMEN

Growth differentiation factor-11 (GDF11) and myostatin (MSTN) are highly related transforming growth factor-ß (TGF-ß) ligands with 89% amino acid sequence homology. They have different biologic activities and diverse tissue distribution patterns. However, the activities of these ligands are indistinguishable in in vitro assays. SMAD2/3 signaling has been identified as the canonical pathway for GDF11 and MSTN, However, it remains unclear which receptor heterodimer and which antagonists preferentially mediate and regulate signaling. In this study, we investigated the initiation and regulation of GDF11 and MSTN signaling at the receptor level using a novel receptor dimerization detection technology. We used the dimerization platform to link early receptor binding events to intracellular downstream signaling. This approach was instrumental in revealing differential receptor binding activity within the TGF-ß family. We verified the ActR2b/ALK5 heterodimer as the predominant receptor for GDF11- and MSTN-induced SMAD2/3 signaling. We also showed ALK7 specifically mediates activin-B signaling. We verified follistatin as a potent antagonist to neutralize both SMAD2/3 signaling and receptor dimerization. More remarkably, we showed that the two related antagonists, growth and differentiation factor-associated serum protein (GASP)-1 and GASP2, differentially regulate GDF11 (and MSTN) signaling. GASP1 blocks both receptor dimerization and downstream signaling. However, GASP2 blocks only downstream signaling without interference from receptor dimerization. Our data strongly suggest that physical binding of GDF11 (and MSTN) to both ActR2b and ALK5 receptors is required for initiation of signaling.


Asunto(s)
Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína 2 Relacionada con la Actina/química , Proteína 2 Relacionada con la Actina/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Células Hep G2 , Humanos , Miostatina/metabolismo , Unión Proteica , Multimerización de Proteína , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Cuaternaria de Proteína , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/química , Transducción de Señal , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...