Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 519, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36720854

RESUMEN

Follicle stimulating hormone (FSH) is an essential glycoprotein hormone for human reproduction, which functions are mediated by a G protein-coupled receptor, FSHR. Aberrant FSH-FSHR signaling causes infertility and ovarian hyperstimulation syndrome. Here we report cryo-EM structures of FSHR in both inactive and active states, with the active structure bound to FSH and an allosteric agonist compound 21 f. The structures of FSHR are similar to other glycoprotein hormone receptors, highlighting a conserved activation mechanism of hormone-induced receptor activation. Compound 21 f formed extensive interactions with the TMD to directly activate FSHR. Importantly, the unique residue H6157.42 in FSHR plays an essential role in determining FSHR selectivity for various allosteric agonists. Together, our structures provide a molecular basis of FSH and small allosteric agonist-mediated FSHR activation, which could inspire the design of FSHR-targeted drugs for the treatment of infertility and controlled ovarian stimulation for in vitro fertilization.


Asunto(s)
Infertilidad , Receptores de HFE , Femenino , Humanos , Hormona Folículo Estimulante , Hidrocortisona , Receptores de HFE/agonistas
2.
Biochim Biophys Acta Biomembr ; 1864(4): 183842, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34954201

RESUMEN

Owing to the critical role of follicle stimulating hormone receptor (FSHR) signaling in human reproduction, FSHR has been widely explored for development of fertility regulators. Using high-throughput screening approaches, several low molecular weight (LMW) compounds that can modulate FSHR activity have been identified. However, the information about the binding sites of these molecules on FSHR is not known. In the present study, we extracted the structural and functional information of 161 experimentally validated LMW FSHR modulators available in PubMed records. The potential FSHR binding sites for these modulators were identified through molecular docking experiments. The binding sites were further mapped to the agonist or antagonist activity reported for these molecules in literature. MD simulations were performed to evaluate the effect of ligand binding on conformational changes in the receptor, specifically the transmembrane domain. A peptidomimetic library was screened using these binding sites. Six peptidomimetics that interacted with the residues of transmembrane domain and extracellular loops were evaluated for binding activity using in vitro cAMP assay. Two of the six peptidomimetics exhibited positive allosteric modulatory activity and four peptidomimetics exhibited negative allosteric modulatory activity. All six peptidomimetics interacted with Asp521 of hFSHR(TMD). Several of the experimentally known LMW FSHR modulators also participated in H-bond interactions with Asp521, suggesting its important role in FSHR modulatory activity.


Asunto(s)
Peptidomiméticos/química , Receptores de HFE/agonistas , Receptores de HFE/antagonistas & inhibidores , Regulación Alostérica , Sitios de Unión , Bases de Datos Factuales , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Biblioteca de Péptidos , Peptidomiméticos/metabolismo , Dominios Proteicos , Receptores de HFE/metabolismo
3.
Int J Mol Sci ; 22(18)2021 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-34576014

RESUMEN

Follicle-stimulating hormone receptor (FSHR) plays a key role in reproduction through the activation of multiple signaling pathways. Low molecular weight (LMW) ligands composed of biased agonist properties are highly valuable tools to decipher complex signaling mechanisms as they allow selective activation of discrete signaling cascades. However, available LMW FSHR ligands have not been fully characterized yet. In this context, we explored the pharmacological diversity of three benzamide and two thiazolidinone derivatives compared to FSH. Concentration/activity curves were generated for Gαs, Gαq, Gαi, ß-arrestin 2 recruitment, and cAMP production, using BRET assays in living cells. ERK phosphorylation was analyzed by Western blotting, and CRE-dependent transcription was assessed using a luciferase reporter assay. All assays were done in either wild-type, Gαs or ß-arrestin 1/2 CRISPR knockout HEK293 cells. Bias factors were calculated for each pair of read-outs by using the operational model. Our results show that each ligand presented a discrete pharmacological efficacy compared to FSH, ranging from super-agonist for ß-arrestin 2 recruitment to pure Gαs bias. Interestingly, LMW ligands generated kinetic profiles distinct from FSH (i.e., faster, slower or transient, depending on the ligand) and correlated with CRE-dependent transcription. In addition, clear system biases were observed in cells depleted of either Gαs or ß-arrestin genes. Such LMW properties are useful pharmacological tools to better dissect the multiple signaling pathways activated by FSHR and assess their relative contributions at the cellular and physio-pathological levels.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/farmacología , Receptores de HFE/agonistas , Arrestina beta 2/farmacología , AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Cinética
4.
Endocrinology ; 162(12)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34192304

RESUMEN

Mutations in G protein-coupled receptors (GPCRs) underlie numerous diseases. Many cause receptor misfolding and failure to reach the cell surface. Pharmacological chaperones are cell-permeant small molecules that engage nascent mutant GPCRs in the endoplasmic reticulum, stabilizing folding and "rescuing" cell surface expression. We previously demonstrated rescue of cell surface expression of luteinizing hormone receptor mutants by an allosteric agonist. Here we demonstrate that a similar approach can be employed to rescue mutant follicle-stimulating hormone receptors (FSHRs) with poor cell surface expression using a small-molecule FSHR agonist, CAN1404. Seventeen FSHR mutations described in patients with reproductive dysfunction were expressed in HEK 293T cells, and cell surface expression was determined by enzyme-linked immunosorbent assay of epitope-tagged FSHRs before/after treatment with CAN1404. Cell surface expression was severely reduced to ≤18% of wild-type (WT) for 11, modestly reduced to 66% to 84% of WT for 4, and not reduced for 2. Of the 11 with severely reduced cell surface expression, restoration to ≥57% of WT levels was achieved for 6 by treatment with 1 µM CAN1404 for 24 h, and a corresponding increase in FSH-induced signaling was observed for 4 of these, indicating restored functionality. Therefore, CAN1404 acts as a pharmacological chaperone and can rescue cell surface expression and function of certain mutant FSHRs with severely reduced cell surface expression. These findings aid in advancing the understanding of the effects of genetic mutations on GPCR function and provide a proof of therapeutic principle for FSHR pharmacological chaperones.


Asunto(s)
Membrana Celular/metabolismo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Animales , Células CHO , Membrana Celular/efectos de los fármacos , Membrana Celular/genética , Cricetinae , Cricetulus , Hormona Folículo Estimulante/farmacología , Células HEK293 , Humanos , Mutación con Pérdida de Función/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , Receptores de HFE/agonistas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
5.
J Clin Endocrinol Metab ; 106(2): e534-e550, 2021 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-33119067

RESUMEN

CONTEXT: Follicle-stimulating hormone (FSH) plays an essential role in gonadal function. Loss-of-function mutations in the follicle-stimulating hormone receptor (FSHR) are an infrequent cause of primary ovarian failure. OBJECTIVE: To analyze the molecular physiopathogenesis of a novel mutation in the FSHR identified in a woman with primary ovarian failure, employing in vitro and in silico approaches, and to compare the features of this dysfunctional receptor with those shown by the trafficking-defective D408Y FSHR mutant. METHODS: Sanger sequencing of the FSHR cDNA was applied to identify the novel mutation. FSH-stimulated cyclic adenosine monophosphate (cAMP) production, ERK1/2 phosphorylation, and desensitization were tested in HEK293 cells. Receptor expression was analyzed by immunoblotting, receptor-binding assays, and flow cytometry. Molecular dynamics simulations were performed to determine the in silico behavior of the mutant FSHRs. RESULTS: A novel missense mutation (I423T) in the second transmembrane domain of the FSHR was identified in a woman with normal pubertal development but primary amenorrhea. The I423T mutation slightly impaired plasma membrane expression of the mature form of the receptor and severely impacted on cAMP/protein kinase A signaling but much less on ß-arrestin-dependent ERK1/2 phosphorylation. Meanwhile, the D408Y mutation severely affected membrane expression, with most of the FSH receptor located intracellularly, and both signal readouts tested. Molecular dynamics simulations revealed important functional disruptions in both mutant FSHRs, mainly the loss of interhelical connectivity in the D408Y FSHR. CONCLUSIONS: Concurrently, these data indicate that conformational differences during the inactive and active states account for the distinct expression levels, differential signaling, and phenotypic expression of the I423T and D408Y mutant FSHRs.


Asunto(s)
Insuficiencia Ovárica Primaria/genética , Receptores de HFE/genética , Adulto , Amenorrea/genética , Amenorrea/metabolismo , Sustitución de Aminoácidos , Familia , Femenino , Hormona Folículo Estimulante/farmacología , Células HEK293 , Humanos , Isoleucina/genética , Mutación con Pérdida de Función/genética , Modelos Moleculares , Mutación Missense , Linaje , Insuficiencia Ovárica Primaria/metabolismo , Receptores de HFE/agonistas , Receptores de HFE/química , Receptores de HFE/metabolismo , Treonina/genética
6.
Anal Biochem ; 586: 113433, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31521670

RESUMEN

This study investigated the association of A419T (rs121909661) and T449I (rs28928870) with infertility among Iranian women and possible treatments by agonizing the mutated receptor. 151 women were genotyped at A419T and T449I sites. Homology modeling, pharmacophore modeling, virtual screening, docking and molecular dynamics (MD) were performed. A419T and T449I indicated a significant and a weak association with infertility among Iranian women (P = 0.005 and P = 0.03, respectively). Significant differences found among three genotypes of A419T with FSH (P = 0.01) and LH (P < 0.0001). G-allele carriers of A419T had susceptibility to display higher FSH and LH serum levels. In silico results revealed the most potent agonists among 3041 similar compounds and MD supported this finding. Altogether, genotyping of A419T and T449I as potential markers might be helpful in prognosis and treatment of infertility. Also, a new series of potent FSHR agonists were identified for future drug development and treatment of infertility related to FSHR dysfunction.


Asunto(s)
Infertilidad Femenina/tratamiento farmacológico , Simulación de Dinámica Molecular , Mutación Missense/efectos de los fármacos , Receptores de HFE/agonistas , Receptores de HFE/antagonistas & inhibidores , Adulto , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Infertilidad Femenina/genética , Irán , Ligandos , Masculino , Estructura Molecular , Embarazo , Receptores de HFE/genética , Adulto Joven
7.
Best Pract Res Clin Endocrinol Metab ; 32(2): 189-200, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29678285

RESUMEN

Infertility treatment may represent a paradigmatic example of precision medicine. Follicle-stimulating hormone (FSH) has been proposed as a valuable therapeutic option both in males and in females, even if a standardized approach is far to be established. To date, several genetic mutations as well as polymorphisms have been demonstrated to significantly affect the pathophysiology of FSH-FSH receptor (FSHR) interaction, although the underlying molecular mechanisms remain unclear. This review aims to highlight possible aspects of FSH therapy that could benefit from a pharmacogenetic approach, providing an up-to-date overview of the variability of the response to FSH treatment in both sexes. Specific sections are dedicated to the clinical use of FSH in infertility and how FSHR polymorphisms may affect the therapeutic endpoints.


Asunto(s)
Infertilidad/genética , Infertilidad/terapia , Mutación , Farmacogenética , Receptores de HFE/genética , Receptores Acoplados a Proteínas G/genética , Femenino , Hormona Folículo Estimulante/agonistas , Hormona Folículo Estimulante/análogos & derivados , Hormona Folículo Estimulante/antagonistas & inhibidores , Hormona Folículo Estimulante/uso terapéutico , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Receptores de HFE/agonistas , Receptores de HFE/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores
8.
J Reprod Dev ; 64(2): 129-134, 2018 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-29249775

RESUMEN

Although the expression of gonadotropin-releasing hormone (GnRH) in the ovaries is well established, its physiological role remains unknown. The aim of this study was to determine whether ovarian GnRH mediates the actions of human chorionic gonadotropin (hCG) in the granulosa cells of immature female rats. Follicular growth was induced by administration of pregnant mare serum gonadotropin (PMSG, 15 IU/0.15 ml) on day 25 after birth, and hCG (20 IU/0.2 ml) was administered on day 27 revealing the increase of plasma progesterone level. Primary cultures of granulosa cells were established from large follicles 2 days after PMSG treatment. Progesterone synthesis was augmented by hCG in a dose-dependent manner. Annexin A5 (ANXA5), a biomarker of GnRH, was expressed in the granulosa-luteal cells after hCG treatment, as shown by immunohistochemistry, suggesting that hCG treatment induced GnRH action. The GnRH mRNA level was increased by hCG, and treatment with GnRH agonist (GnRHa) increased ANXA5 mRNA levels in the primary cultures of granulosa cells. Concomitant incubation of GnRH (10-7 M) or GnRHa (fertirelin acetate, 10-8 M) with hCG suppressed progesterone synthesis during a 3 h incubation period. The mRNA expression of luteinizing hormone receptor (LHR) and follicle-stimulating hormone receptor (FSHR) was synergistically stimulated and suppressed by hCG and GnRHa, respectively. GnRHa stimulated p21 expression, and GnRHa and hCG synergistically reduced the mRNA expression levels of p27 and FOXO1. These data suggest that GnRH induced by LH may have a role for the LH-mediated luteinization of granulosa cells. In addition, ANXA5 may be involved in GnRH action. GnRH-ANXA5 would be an important mechanism in cell differentiation.


Asunto(s)
Gonadotropina Coriónica/farmacología , Fármacos para la Fertilidad Femenina/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/agonistas , Células de la Granulosa/efectos de los fármacos , Luteinización/efectos de los fármacos , Ovario/efectos de los fármacos , Animales , Anexina A5/agonistas , Anexina A5/genética , Anexina A5/metabolismo , Biomarcadores/sangre , Biomarcadores/metabolismo , Células Cultivadas , Femenino , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Gonadotropinas Equinas/farmacología , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Humanos , Inmunohistoquímica , Ovario/citología , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Progesterona/agonistas , Progesterona/antagonistas & inhibidores , Progesterona/biosíntesis , Progesterona/sangre , Ratas Wistar , Receptores de HFE/agonistas , Receptores de HFE/antagonistas & inhibidores , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de HL/agonistas , Receptores de HL/genética , Receptores de HL/metabolismo
9.
Endocrinology ; 159(1): 2-8, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29236987

RESUMEN

Follicle-stimulating hormone (FSH) is critical for ovarian folliculogenesis and essential for female fertility. FSH binds to FSH receptors (FSHRs) and regulates estrogen production in ovarian granulosa cells to orchestrate female reproductive physiology. Ovarian senescence that occurs as a function of aging results in loss of estrogen production, and this is believed to be the major reason for bone loss in postmenopausal women. Although conflicting, studies in rodents and humans during the last decade have provided genetic, pharmacological, and physiological evidence that elevated FSH levels that occur in the face of normal or declining estrogen levels directly regulate bone mass and adiposity. Recently, an efficacious blocking polyclonal FSHß antibody was developed that inhibited ovariectomy-induced bone loss and triggered white-to-brown fat conversion accompanied by mitochondrial biogenesis in mice. Moreover, additional nongonadal targets of FSH action have been identified, and these include the female reproductive tract (endometrium and myometrium), the placenta, hepatocytes, and blood vessels. In this mini-review, I summarize these studies in mice and humans and discuss critical gaps in our knowledge, yet unanswered questions, and the rationale for developing novel genetic models to unambiguously address the extragonadal actions of FSH.


Asunto(s)
Envejecimiento , Hormona Folículo Estimulante/fisiología , Modelos Genéticos , Receptores de HFE/agonistas , Transducción de Señal , Adiposidad , Animales , Desarrollo Óseo , Femenino , Hormona Folículo Estimulante/genética , Humanos , Hígado/fisiología , Masculino , Ratones Noqueados , Ratones Transgénicos , Placentación , Embarazo , Receptores de HFE/genética , Receptores de HFE/fisiología
10.
Reprod Fertil Dev ; 29(6): 1144-1154, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27166082

RESUMEN

The aim of the present study was to evaluate the effect of anti-Müllerian hormone (AMH), with and without FSH, on the in vitro development of isolated caprine preantral follicles, as well as follicular steroid production and mRNA levels of AMH, hormone receptors (AMH and FSH), CYP19A1 (cytochrome P450, family 19, subfamily A, polypeptide 1), CYP17 (cytochrome P450, family 17, subfamily A, polypeptide 1), HSD3B (3-beta-hydroxysteroid dehydrogenase) and Myc (myelocytomatosis oncogene). Isolated secondary follicles were cultured in minimum essential medium alpha (α-MEM+) alone or supplemented with 50ng mL-1 AMH and/or 100ng mL-1 FSH added sequentially on different days of culture. Follicles were cultured for a total of 18 days, with different media during the first (Days 0-9) and second (Days 10-18) halves of the culture period, resulting in six treatment groups, as follows: α-MEM+/α-MEM+, FSH/FSH, AMH/AMH, AMH+FSH/AMH+FSH, AMH/FSH, and FSH/AMH. Follicle development was evaluated on the basis of follicular growth, oocyte maturation and steroid secretion. There was a decrease in follicular growth rate in the AMH, AMH+FSH and AMH/FSH treatment groups compared with α-MEM+ and FSH treatment groups (P<0.05). However, the different culture conditions had no effect on rates of meiotic resumption and steroid secretion (P>0.05). Moreover, follicles cultured in the presence of FSH had lower levels of AMH receptor type II (AMHRII) mRNA compared with non-cultured control (freshly isolated follicles), and the AMH and AMH/FSH treatment groups. In conclusion, AMH reduces the follicular growth rate of isolated goat preantral follicles in vitro without affecting follicular survival.


Asunto(s)
Hormona Antimülleriana/metabolismo , Hormona Folículo Estimulante/metabolismo , Regulación del Desarrollo de la Expresión Génica , Oogénesis , Folículo Ovárico/metabolismo , Receptores de HFE/agonistas , Receptores de Péptidos/agonistas , Receptores de Factores de Crecimiento Transformadores beta/agonistas , Mataderos , Animales , Hormona Antimülleriana/genética , Hormona Antimülleriana/farmacología , Brasil , Bovinos , Proliferación Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cruzamientos Genéticos , Estradiol/metabolismo , Femenino , Hormona Folículo Estimulante/genética , Hormona Folículo Estimulante/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Cabras , Humanos , Oogénesis/efectos de los fármacos , Folículo Ovárico/citología , Folículo Ovárico/efectos de los fármacos , Progesterona/metabolismo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Testosterona/metabolismo , Técnicas de Cultivo de Tejidos
11.
Ann N Y Acad Sci ; 1335: 100-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25118101

RESUMEN

Previously we reported that follicle stimulating hormone (FSH) affects bone degradation in human cells and in follicle stimulating hormone receptor (FSH-R) null mice. Here we describe a FSH-R knockout bone-formation phenotype. We used mesenchymal stem cells (MSCs), osteoblast precursors that express FSH-R, to determine whether FSH regulates bone formation. FSH stimulates MSC cell adhesion 1-3 h and proliferation at 24 h after addition. On the basis of phylogenetic and clinical precedents, we also examined effects of pregnant levels of human chorionic gonadotropin (hCG) on MSCs. We found effects similar to those of FSH, and RNAi knockdown of FSH-R abrogated both FSH and hCG effects on MSCs. In contrast to effects on MSCs, neither FSH nor hCG had significant effects on osteoblast maturation. Also in MSCs, short-term treatment by FSH and hCG altered signaling pathways for proliferation, including Erk1/2 phosphorylation. Our results show augmentation of MSC proliferation by either FSH at menopausal levels or hCG at normal pregnant levels. We conclude that FSH-R participates in regulation of MSC precursor pools in response to either FSH or hCG, integrating the effects of these two glycoprotein hormones.


Asunto(s)
Gonadotropina Coriónica/farmacología , Hormona Folículo Estimulante/farmacología , Glicoproteínas/farmacología , Células Madre Mesenquimatosas/fisiología , Receptores de HFE/fisiología , Animales , Células Cultivadas , Femenino , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Embarazo , Receptores de HFE/agonistas
12.
J Biol Chem ; 289(20): 14273-82, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24692546

RESUMEN

Follicle-stimulating hormone receptor (FSHR), a G-protein coupled receptor, is an important drug target in the development of novel therapeutics for reproductive indications. The FSHR extracellular domains were observed in the crystal structure as a trimer, which enabled us to propose a novel model for the receptor activation mechanism. The model predicts that FSHR binds Asnα(52)-deglycosylated FSH at a 3-fold higher capacity than fully glycosylated FSH. It also predicts that, upon dissociation of the FSHR trimer into monomers, the binding of glycosylated FSH, but not deglycosylated FSH, would increase 3-fold, and that the dissociated monomers would in turn enhance FSHR binding and signaling activities by 3-fold. This study presents evidence confirming these predictions and provides crystallographic and mutagenesis data supporting the proposed model. The model also provides a mechanistic explanation to the agonist and antagonist activities of thyroid-stimulating hormone receptor autoantibodies. We conclude that FSHR exists as a functional trimer.


Asunto(s)
Multimerización de Proteína , Receptores de HFE/química , Receptores de HFE/metabolismo , Regulación Alostérica , Animales , Células CHO , Cricetinae , Cricetulus , Hormona Folículo Estimulante/metabolismo , Humanos , Espacio Intracelular/metabolismo , Modelos Moleculares , Mutagénesis , Mutación , Estructura Cuaternaria de Proteína , Receptores de HFE/agonistas , Receptores de HFE/antagonistas & inhibidores , Transducción de Señal
13.
Reprod Fertil Dev ; 26(8): 1142-53, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24044514

RESUMEN

Although several long-acting follicle-stimulating hormone (FSH) therapies have been developed to enhance the ovarian response, a disadvantage of FSH therapy is its relatively short half-life, which requires women to receive one to two injections per day for almost 2 weeks. In the present study, we developed a novel FSH analogue by conjugating recombinant human FSH (rhFSH) and the constant region of the human immunoglobulin G4 fragment via non-peptidyl linkers. The efficacy of the FSH analogue was evaluated in vitro by cAMP level assessments, pharmacokinetic studies and a determination of ovarian weight and by comparing these findings with the results from other FSH analogues. In addition, the total number of antral and Graafian follicles was determined after 7 days of treatment with control, 6µgkg(-1) follitropin ß, 6, 12 or 42µgkg(-1) corifollitropin α or 3, 6 or 12µgkg(-1) long acting protein/peptide discovery-follicle-stimulating hormone (LAPS-FSH). As a result, the animals treated with 12µgkg(-1) LAPS-FSH produced additional and larger healthy follicles. These data demonstrate that LAPS-FSH promotes growth and inhibits atresia of the ovarian follicle compared with other available drugs, suggesting that our new drug enhances the efficacy and duration of treatment. It is expected that our new FSH analogue will result in a higher chance of pregnancy in patients who are unresponsive to other drugs.


Asunto(s)
Fármacos para la Fertilidad/farmacología , Fertilidad/efectos de los fármacos , Hormona Folículo Estimulante Humana/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Infertilidad/tratamiento farmacológico , Ovario/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Testículo/efectos de los fármacos , Animales , Células CHO , Cricetulus , AMP Cíclico/metabolismo , Femenino , Fármacos para la Fertilidad/administración & dosificación , Fármacos para la Fertilidad/farmacocinética , Hormona Folículo Estimulante Humana/administración & dosificación , Hormona Folículo Estimulante Humana/análogos & derivados , Hormona Folículo Estimulante Humana/farmacocinética , Historia del Siglo XV , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Infertilidad/fisiopatología , Inyecciones Subcutáneas , Masculino , Tamaño de los Órganos , Folículo Ovárico/efectos de los fármacos , Ovario/crecimiento & desarrollo , Ovulación/efectos de los fármacos , Ratas Sprague-Dawley , Receptores de HFE/agonistas , Receptores de HFE/genética , Receptores de HFE/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacocinética , Testículo/crecimiento & desarrollo , Transfección
14.
J Immunol Methods ; 400-401: 87-96, 2013 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-24184184

RESUMEN

Characterizing anti-drug antibodies for neutralizing activity is commonly part of the immunogenicity testing package for most therapeutic proteins. Cell-based neutralization assays can generally be categorized as direct- or indirect assays depending on whether they are associated with therapeutics with agonistic- or antagonistic properties. This paper's aim is a comparison of the two direct neutralization assay formats; the variable- and fixed concentration assay format, using recombinant follicle-stimulating hormone as drug agonist. Essential validation- and performance parameters, such as sample through-put, cut-point, precision, sensitivity and drug tolerance, were compared. The fixed concentration assay format offers superior sample through-put (40 versus 6 samples), precision (coefficient of variation of ≤14% versus 34%) and almost 6 times better sensitivity and is generally recommended as the better option particularly for quasi-quantitative assessments of neutralizing antibodies.


Asunto(s)
Hormona Folículo Estimulante/farmacología , Pruebas de Neutralización/métodos , Receptores de HFE/agonistas , Animales , Anticuerpos Bloqueadores/metabolismo , Unión Competitiva , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Ratones , Variaciones Dependientes del Observador , Receptores de HFE/genética , Receptores de HFE/inmunología , Sensibilidad y Especificidad , Transgenes/genética
15.
Biochim Biophys Acta ; 1830(6): 3466-75, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23500014

RESUMEN

BACKGROUND: During the pre-pubertal life, the cessation of Sertoli cell proliferation and the onset of differentiation are associated with a shift in the FSH-mediated signaling leading to inhibition of the ERK-mitogenic pathway and to a concomitant sensitization of cAMP/PKA pathway. METHODS: To highlight the role of cell proteoglycans (PGs) in the shift of FSH signaling, both FSH-induced cAMP production and ERK1/2 inactivation were studied in untreated and sodium chlorate PG-depleted cultured Sertoli cells from 20day-old rats. RESULTS: Depletion of cell membrane PGs by sodium chlorate reduced FSH-, but not cholera toxin-stimulated cAMP production as well as basal ERK phosphorylation through an okadaic acid (OA)-sensitive mechanism. Involvement of PP2A was further substantiated by a marked decrease in membrane- associated PP2A activity under SC conditions and by the OA-induced restoration of PKA-dependent ERK inactivation in SC-treated cells. CONCLUSIONS: In 20-day-old rat Sertoli cells, transmembrane cell PGs, through tethering/activation of PP2A activity exerts regulatory control on both FSH receptor/Gs coupling and ERK phosphorylation. GENERAL SIGNIFICANCE: Besides their antiproliferative roles, cell PGs such as syndecan-1, could be involved in the increase in cAMP response to FSH occurring in Sertoli cells at the time of transition between proliferative and differentiated states.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Fosfatasa 2/metabolismo , Receptores de HFE/metabolismo , Células de Sertoli/metabolismo , Sindecano-1/metabolismo , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de HFE/agonistas , Sistemas de Mensajero Secundario/fisiología , Ovinos
16.
Biochem Pharmacol ; 85(8): 1162-70, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23415902

RESUMEN

Follicle-stimulating hormone (FSH) activates FSH receptors (FSHR) in granulosa cells to induce follicle differentiation, growth and estradiol production. FSH is used clinically to treat female infertility and is administered by injection. To increase patient convenience and compliance, compound homogeneity and composition, low molecular weight (LMW), orally bioavailable, FSHR agonists are now being developed to replace FSH. In this study, we present the signaling mechanisms of a newly developed LMW dihydropyridine agonist of the FSHR, Org 214444-0. Org 214444-0 is shown to be a stereoselective, nanomolar potent FSHR agonist and selective over the structurally related LHR and TSHR. Org 214444-0 is an allosteric agonist interacting with the transmembrane region of the FSHR. When co-incubated with FSH, Org 214444-0 augments FSH's potency in binding (6.5-fold) and adenylyl cyclase/cAMP activation (3.5-fold) in a concentration-dependent manner. Like FSH, Org 214444-0 induces FSHR internalization and is only marginally effective in stimulating phospholipase C. Moreover, Org 214444-0 stimulates cAMP and estradiol production in human granulosa cells in culture and supports the follicular phase in mature female rats. We conclude that Org 214444-0 is a bonafide FSHR agonist.


Asunto(s)
Dihidropiridinas/farmacología , Receptores de HFE/agonistas , Sulfonamidas/farmacología , Regulación Alostérica , Secuencia de Aminoácidos , Animales , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/fisiología , Femenino , Hormona Folículo Estimulante/metabolismo , Datos de Secuencia Molecular , Peso Molecular , Ratas , Receptores de HFE/química , Transducción de Señal , Fosfolipasas de Tipo C/metabolismo
17.
Reproduction ; 142(6): 893-905, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21930683

RESUMEN

During recent decades minor innovative drugs have been developed for the female contraceptive market and they all contain steroidal progestagens (and estrogens) that act centrally and have side effects that can be attributed to this central action. In this study, we present an innovative tissue-specific approach for female contraception by low molecular weight (LMW) FSH receptor (FSHR) agonists, which interact with the FSHR that is dominantly expressed in the granulosa cells. The oral administration of LMW FSHR agonists with a short circulation time, induced formation of luteinized unruptured follicles (LUFs) from the Graafian follicles, thereby preventing the release of the oocyte. The short-acting LMW FSHR compounds were fully agonistic to FSHR (EC(50)=4-5 nM). In an isolated mouse follicle culture, a short incubation period (2 h) resulted in inhibition of follicular rupture, where continuous incubation induced follicle growth. Pharmacokinetics after oral administration showed a surge-like exposure in rats and monkeys. Oral administration of short-acting LMW FSHR agonists inhibited ovulation at 10 mg/kg in rats and guinea pigs by generating LUFs without affecting cyclicity. Also, inhibition of follicular rupture was shown to be reversible within one cycle. Finally, LUFs were induced without affecting the hormonal cyclicity in cynomolgus monkeys, a mono-ovulatory species. In healthy women LUF formation occurs naturally, with a LUF acting as corpus luteum that produces enough progesterone to ensure normal menstrual cyclicity. Together with the presented data this indicates that the innovative approach with short-acting LMW FSHR agonists could lead to oral contraception for females at the ovarian level.


Asunto(s)
Anticonceptivos/farmacología , Folículo Ovárico/efectos de los fármacos , Inhibición de la Ovulación , Receptores de HFE/agonistas , Animales , Relación Dosis-Respuesta a Droga , Femenino , Cobayas , Macaca fascicularis , Ratones , Modelos Animales , Ratas
18.
Biomed Khim ; 56(3): 303-18, 2010.
Artículo en Ruso | MEDLINE | ID: mdl-20695210

RESUMEN

During the last years the low-molecular non-peptidic regulators of the polypeptide hormones receptors containing LGR-repeats were identified. In the review the data on the structure and the molecular mechanisms of action of these regulators as agonists and antagonists of the luteinizing, follicle-stimulating and thyrotropin hormones are analyzed and systematized. The regulators interact with the serpentine domain of LGR-receptor and trigger the signaling cascades coupled with the receptor. Low-molecular agonists and antagonists of the LGR-receptors are considered as a new generation of the drugs that regulates the functional activity of sensitive to pituitary glycoprotein hormones signaling systems with high efficiency and selectivity. These regulators are more accessible compared to the hormones and can be use orally.


Asunto(s)
Receptores de Péptidos , Humanos , Ligandos , Estructura Molecular , Conformación Proteica , Receptores de HFE/agonistas , Receptores de HFE/antagonistas & inhibidores , Receptores de HL/agonistas , Receptores de Péptidos/agonistas , Receptores de Péptidos/antagonistas & inhibidores , Receptores de Péptidos/química , Receptores de Tirotropina/antagonistas & inhibidores , Secuencias Repetitivas de Aminoácido , Transducción de Señal
19.
Biochem Biophys Res Commun ; 394(1): 12-7, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20171950

RESUMEN

Cells of the monocyte series respond to follicle stimulating hormone (FSH) by poorly characterized mechanisms. We studied FSH-receptors (FSH-R) and FSH response in nontransformed human monocytes and in osteoclasts differentiated from these cells. Western blot and PCR confirmed FSH-R expression on monocytes or osteoclasts, although at low levels relative to ovarian controls. Monocyte and osteoclast FSH-Rs differed from FSH-R from ovarian cells, reflecting variable splicing in exons 8-10. Monocytes produced no cAMP, the major signal in ovarian cells, in response to FSH. However, monocytes and osteoclasts transcribed TNFalpha in response to the FSH. No relation of expression of osteoclast FSH-R to the sex of cell donors or to exposure to sex hormones was apparent. Controls for FSH purity and endotoxin contamination were negative. Unamplified cRNA screening in adherent CD14 cells after 2h in 25ng/ml FSH showed increased transcription of RANKL signalling proteins. Transcription of key proteins that stimulate bone turnover, TNFalpha and TSG-6, increased 2- to 3-fold after FSH treatment. Smaller but significant changes occurred in transcripts of selected signalling, adhesion, and cytoskeletal proteins. We conclude that monocyte and osteoclast FSH response diverges from that of ovarian cells, reflecting, at least in part, varying FSH-R isoforms.


Asunto(s)
Remodelación Ósea/genética , Hormona Folículo Estimulante/metabolismo , Monocitos/metabolismo , Osteoclastos/metabolismo , Receptores de HFE/metabolismo , Transcripción Genética , Remodelación Ósea/efectos de los fármacos , AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Estrógenos/farmacología , Femenino , Hormona Folículo Estimulante/farmacología , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Monocitos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Receptores de HFE/agonistas , Testosterona/farmacología , Factor de Necrosis Tumoral alfa/genética
20.
Mol Endocrinol ; 24(3): 561-73, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20107152

RESUMEN

Deglycosylated FSH is known to trigger poor Galphas coupling while efficiently binding its receptor. In the present study, we tested the possibility that a deglycosylated equine LH (eLHdg) might be able to selectively activate beta-arrestin-dependent signaling. We compared native eLH to an eLH derivative [i.e. truncated eLHbeta (Delta121-149) combined with asparagine56-deglycosylated eLHalpha (eLHdg)] previously reported as an antagonist of cAMP accumulation at the FSH receptor (FSH-R). We confirmed that, when used in conjunction with FSH, eLHdg acted as an antagonist for cAMP accumulation in HEK-293 cells stably expressing the FSH-R. Furthermore, when used alone at concentrations up to 1 nM, eLHdg had no detectable agonistic activity on cAMP accumulation, protein kinase A activity or cAMP-responsive element-dependent transcriptional activity. At higher concentrations, however, a weak agonistic action was observed with eLHdg, whereas eLH led to robust responses whatever the concentration. Both eLH and eLHdg triggered receptor internalization and led to beta-arrestin recruitment. Both eLH and eLHdg triggered ERK and ribosomal protein (rp) S6 phosphorylation at 1 nM. The depletion of endogenous beta-arrestins had only a partial effect on eLH-induced ERK and rpS6 phosphorylation. In contrast, ERK and rpS6 phosphorylation was completely abolished at all time points in beta-arrestin-depleted cells. Together, these results show that eLHdg has the ability to preferentially activate beta-arrestin-dependent signaling at the FSH-R. This finding provides a new conceptual and experimental framework to revisit the physiological meaning of gonadotropin structural heterogeneity. Importantly, it also opens a field of possibilities for the development of selective modulators of gonadotropin receptors.


Asunto(s)
Arrestinas/metabolismo , Hormona Luteinizante/análogos & derivados , Receptores de HFE/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Western Blotting , Bovinos , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Caballos , Humanos , Inmunoprecipitación , Hormona Luteinizante/química , Hormona Luteinizante/metabolismo , Hormona Luteinizante/farmacología , Ratones , Fosforilación/efectos de los fármacos , Unión Proteica , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño , Receptores de HFE/agonistas , Receptores de HFE/antagonistas & inhibidores , Proteína S6 Ribosómica/metabolismo , Porcinos , beta-Arrestinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...