Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Int J Mol Sci ; 25(14)2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-39062762

RESUMO

Female fertility depends on the ovarian reserve of follicles, which is determined at birth. Primordial follicle development and oocyte maturation are regulated by multiple factors and pathways and classified into gonadotropin-independent and gonadotropin-dependent phases, according to the response to gonadotropins. Folliculogenesis has always been considered to be gonadotropin-dependent only from the antral stage, but evidence from the literature highlights the role of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) during early folliculogenesis with a potential role in the progression of the pool of primordial follicles. Hormonal and molecular pathway alterations during the very earliest stages of folliculogenesis may be the root cause of anovulation in polycystic ovary syndrome (PCOS) and in PCOS-like phenotypes related to antiepileptic treatment. Excessive induction of primordial follicle activation can also lead to premature ovarian insufficiency (POI), a condition characterized by menopause in women before 40 years of age. Future treatments aiming to suppress initial recruitment or prevent the growth of resting follicles could help in prolonging female fertility, especially in women with PCOS or POI. This review will briefly introduce the impact of gonadotropins on early folliculogenesis. We will discuss the influence of LH on ovarian reserve and its potential role in PCOS and POI infertility.


Assuntos
Gonadotropinas , Folículo Ovariano , Síndrome do Ovário Policístico , Insuficiência Ovariana Primária , Animais , Feminino , Humanos , Hormônio Foliculoestimulante/metabolismo , Gonadotropinas/metabolismo , Hormônio Luteinizante/metabolismo , Folículo Ovariano/metabolismo , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/fisiopatologia , Insuficiência Ovariana Primária/metabolismo , Insuficiência Ovariana Primária/etiologia , Insuficiência Ovariana Primária/patologia
2.
Gynecol Obstet Invest ; 88(4): 214-225, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37369184

RESUMO

OBJECTIVES: The aim of the study was to evaluate dosing of recombinant human luteinizing hormone (r-hLH) or human menopausal gonadotrophin (hMG)-derived medications with LH activity in ovarian stimulation (OS) cycles for in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI). DESIGN: A non-interventional study was performed to analyse data from the German RecDate database (January 2007-December 2011). PARTICIPANTS/MATERIALS, SETTING, METHODS: Starting/total r-hLH/hMG dose, OS duration/cycle number, r-hLH/hMG initiation day (first day of administration), and population/cycle characteristics were assessed in women (≥18 years) undergoing OS for IVF/ICSI using r-hLH or hMG-derived medications (excluding corifollitropin alfa, clomiphene citrate, letrozole, mini/micro-dose human chorionic gonadotrophin, and urofollitropin alone). Data were summarized descriptively. RESULTS: 67,858 identified cycles utilized medications containing r-hLH (10,749), hMG (56,432), or both (677). Mean (standard deviation) OS duration with r-hLH and hMG was 10.1 (4.43) and 9.8 (6.16) days, respectively. Median (25th-75th percentile) r-hLH starting dose (75.0 [75.0-150.0] IU) was consistent across patients regardless of age, infertility diagnosis, or gonadotrophin-releasing hormone (GnRH) protocol. Median (25th-75th percentile) hMG-derived LH activity starting dose was 225.0 (150.0-300.0) IU, regardless of GnRH protocol, but was lower in women aged <35 years and those with ovulation disorders/polycystic ovary syndrome. Median (25th-75th percentile) total dose for r-hLH (750.0 [337.5-1,125.0] IU) and hMG-derived LH activity (1,575.0 [750.0-2,625.0] IU) varied according to patients' age, infertility diagnosis, cycle number, and r-hLH/hMG initiation day. GnRH antagonist use resulted in a numerically higher median total hMG-derived LH activity dose than GnRH agonist use. LIMITATIONS: The data used in this study were taken from electronic medical records relating to a specific timeframe (2007-2011) and therefore may not accurately reflect current clinical practice; however, it is likely that the differences between the two compounds would be maintained. Additionally, secondary data sources may suffer from uniformity and quality issues. CONCLUSIONS: The standard of care for OS cycles is described with respect to IVF/ICSI treatment including an LH component in Germany during the specified timeframe.


Assuntos
Infertilidade , Sêmen , Humanos , Feminino , Masculino , Hormônio Luteinizante , Menotropinas/uso terapêutico , Indução da Ovulação/métodos , Hormônio Liberador de Gonadotropina , Fertilização in vitro/métodos , Menopausa , Fertilidade
3.
Trends Mol Med ; 29(9): 711-725, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37331882

RESUMO

Embryo implantation failure and spontaneous abortions represent the main causes of infertility in developed countries. Unfortunately, incomplete knowledge of the multiple factors involved in implantation and fetal development keeps the success rate of medically assisted procreation techniques relatively low. According to recent literature, cellular and molecular mechanisms of 'immunogenic tolerance' towards the embryo are crucial to establish an 'anti-inflammatory' state permissive of a healthy pregnancy. In this review we dissect the role played by the immune system in the endometrial-embryo crosstalk, with a particular emphasis towards the fork-head-box-p3 (Foxp3+) CD4+CD25+ regulatory T (Treg) cells and discuss the most recent therapeutic advances in the context of early immune-mediated pregnancy loss.


Assuntos
Implantação do Embrião , Ciência Translacional Biomédica , Gravidez , Feminino , Humanos , Linfócitos T Reguladores , Tolerância Imunológica , Endométrio
4.
Front Endocrinol (Lausanne) ; 12: 675670, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34040586

RESUMO

Background: A Delphi consensus was conducted to evaluate global expert opinions on key aspects of assisted reproductive technology (ART) treatment. Methods: Ten experts plus the Scientific Coordinator discussed and amended statements plus supporting references proposed by the Scientific Coordinator. The statements were distributed via an online survey to 35 experts, who voted on their level of agreement or disagreement with each statement. Consensus was reached if the proportion of participants agreeing or disagreeing with a statement was >66%. Results: Eighteen statements were developed. All statements reached consensus and the most relevant are summarised here. (1) Follicular development and stimulation with gonadotropins (n = 9 statements): Recombinant human follicle stimulating hormone (r-hFSH) alone is sufficient for follicular development in normogonadotropic patients aged <35 years. Oocyte number and live birth rate are strongly correlated; there is a positive linear correlation with cumulative live birth rate. Different r-hFSH preparations have identical polypeptide chains but different glycosylation patterns, affecting the biospecific activity of r-hFSH. r-hFSH plus recombinant human LH (r-hFSH:r-hLH) demonstrates improved pregnancy rates and cost efficacy versus human menopausal gonadotropin (hMG) in patients with severe FSH and LH deficiency. (2) Pituitary suppression (n = 2 statements): Gonadotropin releasing hormone (GnRH) antagonists are associated with lower rates of any grade ovarian hyperstimulation syndrome (OHSS) and cycle cancellation versus GnRH agonists. (3) Final oocyte maturation triggering (n=4 statements): Human chorionic gonadotropin (hCG) represents the gold standard in fresh cycles. The efficacy of hCG triggering for frozen transfers in modified natural cycles is controversial compared with LH peak monitoring. Current evidence supports significantly higher pregnancy rates with hCG + GnRH agonist versus hCG alone, but further evidence is needed. GnRH agonist trigger, in GnRH antagonist protocol, is recommended for final oocyte maturation in women at risk of OHSS. (4) Luteal-phase support (n = 3 statements): Vaginal progesterone therapy represents the gold standard for luteal-phase support. Conclusions: This Delphi consensus provides a real-world clinical perspective on the specific approaches during the key steps of ART treatment from a diverse group of international experts. Additional guidance from clinicians on ART strategies could complement guidelines and policies, and may help to further improve treatment outcomes.


Assuntos
Fertilização in vitro/normas , Fase Luteal/fisiologia , Oócitos/crescimento & desenvolvimento , Oogênese , Indução da Ovulação/normas , Hipófise/efeitos dos fármacos , Técnicas de Reprodução Assistida/normas , Gonadotropina Coriônica/administração & dosagem , Consenso , Técnica Delphi , Feminino , Hormônio Foliculoestimulante Humano/metabolismo , Hormônio Liberador de Gonadotropina/agonistas , Humanos , Guias de Prática Clínica como Assunto , Gravidez , Progesterona/metabolismo
5.
Front Endocrinol (Lausanne) ; 12: 797365, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35178027

RESUMO

Background: A Delphi consensus was conducted to evaluate the influence of single nucleotide polymorphisms (SNPs) in genes encoding gonadotropin and gonadotropin receptors on clinical ovarian stimulation outcomes following assisted reproductive technology (ART) treatment. Methods: Nine experts plus two Scientific Coordinators discussed and amended statements plus supporting references proposed by the Scientific Coordinators. The statements were distributed via an online survey to 36 experts, who voted on their level of agreement or disagreement with each statement. Consensus was reached if the proportion of participants agreeing or disagreeing with a statement was >66%. Results: Eleven statements were developed, of which two statements were merged. Overall, eight statements achieved consensus and two statements did not achieve consensus. The statements reaching consensus are summarized here. (1) SNP in the follicle stimulating hormone receptor (FSHR), rs6166 (c.2039A>G, p.Asn680Ser) (N=5 statements): Ser/Ser carriers have higher basal FSH levels than Asn/Asn carriers. Ser/Ser carriers require higher amounts of gonadotropin during ovarian stimulation than Asn/Asn carriers. Ser/Ser carriers produce fewer oocytes during ovarian stimulation than Asn/Asn or Asn/Ser carriers. There is mixed evidence supporting an association between this variant and ovarian hyperstimulation syndrome. (2) SNP of FSHR, rs6165 (c.919G>A, p.Thr307Ala) (N=1 statement): Few studies suggest Thr/Thr carriers require a shorter duration of gonadotropin stimulation than Thr/Ala or Ala/Ala carriers. (3) SNP of FSHR, rs1394205 (-29G>A) (N=1 statement): Limited data in specific ethnic groups suggest that A/A allele carriers may require higher amounts of gonadotropin during ovarian stimulation and produce fewer oocytes than G/G carriers. (4) SNP of FSH ß-chain (FSHB), rs10835638 (-211G>T) (N=1 statement): There is contradictory evidence supporting an association between this variant and basal FSH levels or oocyte number. (5) SNPs of luteinizing hormone ß-chain (LHB) and LH/choriogonadotropin receptor (LHCGR) genes (N=1 statement): these may influence ovarian stimulation outcomes and could represent potential future targets for pharmacogenomic research in ART, although data are still very limited. Conclusions: This Delphi consensus provides clinical perspectives from a diverse international group of experts. The consensus supports a link between some variants in gonadotropin/gonadotropin receptor genes and ovarian stimulation outcomes; however, further research is needed to clarify these findings.


Assuntos
Subunidade beta do Hormônio Folículoestimulante , Indução da Ovulação , Gonadotropina Coriônica , Consenso , Técnica Delphi , Feminino , Subunidade beta do Hormônio Folículoestimulante/genética , Humanos
6.
Int J Mol Sci ; 20(22)2019 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-31703269

RESUMO

Commercial gonadotropin-releasing hormone (GnRH) antagonists differ by 1-2 amino acids and are used to inhibit gonadotropin production during assisted reproduction technologies (ART). In this study, potencies of three GnRH antagonists, Cetrorelix, Ganirelix and Teverelix, in inhibiting GnRH-mediated intracellular signaling, were compared in vitro. GnRH receptor (GnRHR)-transfected HEK293 and neuroblastoma-derived SH-SY5Y cell lines, as well as mouse pituitary LßT2 cells endogenously expressing the murine GnRHR, were treated with GnRH in the presence or absence of the antagonist. We evaluated intracellular calcium (Ca2+) and cAMP increases, cAMP-responsive element binding-protein (CREB) and extracellular-regulated kinase 1 and 2 (ERK1/2) phosphorylation, ß-catenin activation and mouse luteinizing-hormone ß-encoding gene (Lhb) transcription by bioluminescence resonance energy transfer (BRET), Western blotting, immunostaining and real-time PCR as appropriate. The kinetics of GnRH-induced Ca2+ rapid increase revealed dose-response accumulation with potency (EC50) of 23 nM in transfected HEK293 cells, transfected SH-SY5Y and LßT2 cells. Cetrorelix inhibited the 3 × EC50 GnRH-activated calcium signaling at concentrations of 1 nM-1 µM, demonstrating higher potency than Ganirelix and Teverelix, whose inhibitory doses fell within the 100 nM-1 µM range in both transfected HEK293 and SH-SY5Y cells in vitro. In transfected SH-SY5Y, Cetrorelix was also significantly more potent than other antagonists in reducing GnRH-mediated cAMP accumulation. All antagonists inhibited pERK1/2 and pCREB activation at similar doses, in LßT2 and transfected HEK293 cells treated with 100 nM GnRH. Although immunostainings suggested that Teverelix could be less effective than Cetrorelix and Ganirelix in inhibiting 1 µM GnRH-induced ß-catenin activation, Lhb gene expression increase occurring upon LßT2 cell treatment by 1 µM GnRH was similarly inhibited by all antagonists. To conclude, this study has demonstrated Cetrorelix-, Ganirelix- and Teverelix-specific biased effects at the intracellular level, not affecting the efficacy of antagonists in inhibiting Lhb gene transcription.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Antagonistas de Hormônios/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Receptores LHRH/metabolismo , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Hormônio Liberador de Gonadotropina/metabolismo , Células HEK293 , Humanos , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo
7.
Gynecol Endocrinol ; 35(12): 1027-1036, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31392906

RESUMO

It is essential that fertility treatment is individualized based on a thorough diagnostic work-up, with treatment tailored to the patients' requirements. This individualization should be kept in mind during the main decision points that occur before and during treatment. Treatment customization must include consideration of both the woman and her partner involved in the process together, including their collective treatment goals. Once treatment goals have been agreed and diagnostic evaluations performed, personalization based on patient characteristics, together with an understanding of treatment goals and patient preferences, enables the selection of appropriate treatments, protocols, products and their dosing. Following treatment initiation, monitoring and adaptation of product and dose can then ensure optimal outcomes. Currently, it is not possible to base treatment decisions on every characteristic of the patient and personalization is based on biomarkers that have been identified as the most relevant. However, in the future, the use of artificial intelligence coupled with continuous monitoring should enable greater individualization and improve outcomes. This review considers the current state-of-the-art related to decision points during individualized treatment of female infertility, before looking at future developments that might further assist in making individualized treatment decisions, including the use of computer-assisted decision making.


Assuntos
Fármacos para a Fertilidade Feminina/uso terapêutico , Gonadotropinas/uso terapêutico , Infertilidade Feminina/terapia , Técnicas de Reprodução Assistida , Gonadotropina Coriônica/uso terapêutico , Feminino , Fertilização in vitro/métodos , Hormônio Foliculoestimulante/uso terapêutico , Hormônio Liberador de Gonadotropina/agonistas , Humanos , Inseminação Artificial , Hormônio Luteinizante/uso terapêutico , Indução da Ovulação/métodos , Seleção de Pacientes
8.
Cell Death Differ ; 24(1): 72-82, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27689876

RESUMO

Premature ovarian failure and female infertility are frequent side effects of anticancer therapies, owing to the extreme sensitivity of the ovarian reserve oocytes to the damaging effects of irradiation and chemotherapy on DNA. We report here a robust protective effect of luteinizing hormone (LH) on the primordial follicle pool of prepubertal ovaries against the cisplatin (Cs)-induced apoptosis. In vitro LH treatment of prepubertal ovarian fragments generated anti-apoptotic signals by a subset of ovarian somatic cells expressing LH receptor (LHR) through cAMP/PKA and Akt pathways. Such signals, reducing the oocyte level of pro-apoptotic TAp63 protein and favoring the repair of the Cs-damaged DNA in the oocytes, prevented their apoptosis. Noteworthy, in vivo administration to prepubertal female mice of a single dose of LH together with Cs inhibited the depletion of the primordial follicle reserve caused by the drug and preserved their fertility in reproductive age, preventing significant alteration in the number of pregnancy and of delivered pups. In conclusion, these findings establish a novel ovoprotective role for LH and further support the very attracting prospective to use physiological 'fertoprotective' approaches for preventing premature infertility and risks linked to precocious menopause in young patients who survived cancer after chemotherapy.


Assuntos
Apoptose/efeitos dos fármacos , Cisplatino/toxicidade , Hormônio Luteinizante/farmacologia , Androstadienos/farmacologia , Animais , Células Cultivadas , Cromonas/farmacologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dano ao DNA/efeitos dos fármacos , Feminino , Fertilidade/efeitos dos fármacos , Hormônio Foliculoestimulante/farmacologia , Camundongos , Camundongos Transgênicos , Morfolinas/farmacologia , Oócitos/citologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Folículo Ovariano/citologia , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores do LH/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Wortmanina
9.
Int J Mol Sci ; 17(7)2016 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-27428949

RESUMO

A bulk of evidence suggests that d-aspartate (d-Asp) regulates steroidogenesis and spermatogenesis in vertebrate testes. This review article focuses on intracellular signaling mechanisms elicited by d-Asp possibly via binding to the N-methyl-d-aspartate receptor (NMDAR) in both Leydig cells, and spermatogonia. In Leydig cells, the amino acid upregulates androgen production by eliciting the adenylate cyclase-cAMP and/or mitogen-activated protein kinase (MAPK) pathways. d-Asp treatment enhances gene and protein expression of enzymes involved in the steroidogenic cascade. d-Asp also directly affects spermatogonial mitotic activity. In spermatogonial GC-1 cells, d-Asp induces phosphorylation of MAPK and AKT serine-threonine kinase proteins, and stimulates expression of proliferating cell nuclear antigen (PCNA) and aurora kinase B (AURKB). Further stimulation of spermatogonial GC-1 cell proliferation might come from estradiol/estrogen receptor ß (ESR2) interaction. d-Asp modulates androgen and estrogen levels as well as the expression of their receptors in the rat epididymis by acting on mRNA levels of Srd5a1 and Cyp19a1 enzymes, hence suggesting involvement in spermatozoa maturation.


Assuntos
Ácido D-Aspártico/farmacologia , Células Intersticiais do Testículo/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Animais , Humanos , Masculino , Ratos
10.
Theriogenology ; 81(5): 744-51, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24439162

RESUMO

Previous studies have demonstrated that D-aspartic acid (D-Asp) has a role in regulating the release and synthesis of testosterone in rats. In this study, we investigated the molecular pathway by which this amino acid triggers its action in the rat testis. We found expression of N-Methyl-D-Aspartic Acid (NMDA) receptor messenger RNAs for NR1, NR2A, and NR2D receptor subunits. After D-Asp administration, NR1 and NR2A messenger RNA levels were significantly higher than those of controls, whereas NR2D levels remained unchanged. Expression of extracellular signal-regulated kinase (ERK) 1 protein was higher than that of ERK2 protein in the testis of both D-Asp-treated rats and controls. D-Asp administration increased testis levels of both phosphorylated ERK (P-ERK) 1 and 2. Using immunohistochemical technique, NR1 and P-ERK 1 or 2 proteins were preferentially localized within the spermatogonia. Moreover, D-Asp administration increased both serum and testis testosterone levels but not estradiol levels. Finally, in D-Asp-treated rats, testicular androgen receptor protein levels were significantly increased, whereas both estrogen receptor α and P-450 aromatase levels were significantly decreased. Conclusively, our results, besides strengthening the evidence that D-Asp administration in rats induces testosterone synthesis, demonstrate for the first time that D-Asp (1) induces testicular NMDA receptor-ERK pathway, (2) upregulates androgen receptor expression, and (3) downregulates estrogen receptor expression.


Assuntos
Ácido D-Aspártico/farmacologia , Expressão Gênica/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Receptores Androgênicos/genética , Receptores de N-Metil-D-Aspartato/genética , Animais , Aromatase/genética , Regulação para Baixo/efeitos dos fármacos , Estradiol/análise , Estradiol/sangue , Receptor alfa de Estrogênio/análise , Receptor alfa de Estrogênio/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , RNA Mensageiro/análise , Ratos , Ratos Wistar , Receptores Androgênicos/análise , Receptores de N-Metil-D-Aspartato/análise , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/fisiologia , Testículo/química , Testosterona/análise , Testosterona/sangue , Regulação para Cima/efeitos dos fármacos
11.
Reprod Biol Endocrinol ; 11: 51, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23725475

RESUMO

BACKGROUND: V-betaLH is a common genetic variant of LH caused by two polymorphic base changes in the beta subunit gene, altering the amino acid sequence (Trp8Arg and Ile15Thr). In a previous-preliminary trial performed in women undergoing IVF, it was demonstrated that carriers of v-betaLH show sub-optimal ovarian response to a standard long GnRH-agonist down -regulation protocol when stimulated with pure recombinant FSH (r-hFSH). The aim of this study was to confirm the hypothesis that women with v-betaLH display hypo-sensitivity to exogenous FSH in a larger IVF population and to explore the frequency of this variant in a Danish female population. METHODS: In the present study, the effect of v-betaLH was retrospectively investigated in a larger series of women undergoing controlled ovarian stimulation (COS) and, for the first time, in a Danish IVF population. A total of 220 normogonadotrophic women following a long GnRH-agonist down-regulation protocol received an individualized dose of r-hFSH (100 IU and 375 IU s.c. daily) according to antral follicle count, baseline FSH, body mass index and age. The LH genotype was assessed in all patients by immunofluorometric assay. RESULTS: V-betaLH was present in 11% of patients, whereas the allelic frequency was 12%. The study population was divided into two groups according to their LH genotype. Group A consisted of 196 wt/wt women. Group B included 24 individuals with v-betaLH (21 heterozygous and 3 homozygous). No statistically significant differences in the mean number of oocytes retrieved, fertilization rate and pregnancy rate per cycle were observed between groups. However, Group B received a significantly higher cumulative-dose of r-hFSH than Group A (2435.86 +/- 932.8 IU versus 1959.8 +/- 736.45 p = 0.048). When one-way ANOVA in a within design was applied, the LH genotype had a statistically significant effect (p < 0.01) on the cumulative dose of r-hFSH, showing a progressive increase from wt/wt (1959.8 +/- 736.45 IU) to v-betaLH hetero- (2267.5 +/- 824.3) and homozygotic women (3558.3 +/- 970.9). CONCLUSIONS: These results confirm that carriers exhibit hypo-sensitivity to exogenous FSH during COS, documenting that the frequency of v-betaLH in Denmark is similar to a number of European countries.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Hormônio Luteinizante Subunidade beta/genética , Indução da Ovulação/métodos , Técnicas de Reprodução Assistida , Adulto , Alelos , Dinamarca/epidemiologia , Estrogênios/administração & dosagem , Feminino , Hormônio Foliculoestimulante/administração & dosagem , Frequência do Gene , Hormônio Liberador de Gonadotropina/agonistas , Humanos , Infertilidade Feminina/genética , Infertilidade Feminina/terapia , Polimorfismo Genético , Estudos Retrospectivos
12.
PLoS One ; 7(10): e46682, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23071612

RESUMO

Human luteinizing hormone (hLH) and chorionic gonadotropin (hCG) act on the same receptor (LHCGR) but it is not known whether they elicit the same cellular and molecular response. This study compares for the first time the activation of cell-signalling pathways and gene expression in response to hLH and hCG. Using recombinant hLH and recombinant hCG we evaluated the kinetics of cAMP production in COS-7 and hGL5 cells permanently expressing LHCGR (COS-7/LHCGR, hGL5/LHCGR), as well as cAMP, ERK1/2, AKT activation and progesterone production in primary human granulosa cells (hGLC). The expression of selected target genes was measured in the presence or absence of ERK- or AKT-pathways inhibitors. In COS-7/LHCGR cells, hCG is 5-fold more potent than hLH (cAMP ED(50): 107.1±14.3 pM and 530.0±51.2 pM, respectively). hLH maximal effect was significantly faster (10 minutes by hLH; 1 hour by hCG). In hGLC continuous exposure to equipotent doses of gonadotropins up to 36 hours revealed that intracellular cAMP production is oscillating and significantly higher by hCG versus hLH. Conversely, phospho-ERK1/2 and -AKT activation was more potent and sustained by hLH versus hCG. ERK1/2 and AKT inhibition removed the inhibitory effect on NRG1 (neuregulin) expression by hLH but not by hCG; ERK1/2 inhibition significantly increased hLH- but not hCG-stimulated CYP19A1 (aromatase) expression. We conclude that: i) hCG is more potent on cAMP production, while hLH is more potent on ERK and AKT activation; ii) hGLC respond to equipotent, constant hLH or hCG stimulation with a fluctuating cAMP production and progressive progesterone secretion; and iii) the expression of hLH and hCG target genes partly involves the activation of different pathways depending on the ligand. Therefore, the LHCGR is able to differentiate the activity of hLH and hCG.


Assuntos
Gonadotropina Coriônica/fisiologia , Hormônio Luteinizante/fisiologia , Receptores do LH/metabolismo , Transdução de Sinais , 1-Metil-3-Isobutilxantina/farmacologia , Adulto , Animais , Células COS , Forma Celular , Chlorocebus aethiops , Gonadotropina Coriônica/farmacologia , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Células da Granulosa/metabolismo , Humanos , Cinética , Hormônio Luteinizante/farmacologia , Pessoa de Meia-Idade , Cultura Primária de Células , Progesterona/biossíntese , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores do LH/agonistas , Receptores do LH/fisiologia , Transcriptoma
13.
Cardiovasc Res ; 96(3): 381-90, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22875468

RESUMO

AIMS: The suppressors of cytokine signalling (SOCS) are identified inhibitors of cytokine and growth factor signalling that act via the Janus kinase (JAK) signal transducers and activators of transcription (STAT) pathways. Aberrant JAK/STAT signalling promotes progression from hypertrophy to heart failure. Little information is available concerning the role of SOCS in the transition from hypertrophy to heart failure. To this aim, we investigated the effects of SOCS1 overexpression obtained by in vivo adeno-associated gene transfer using an aortopulmonary cross-clamping technique in a chronic pressure-overload cardiac rat model. METHODS AND RESULTS: Rats were randomized into four groups: sham-operated (n = 18), aortic banding (AB) (n = 18), AB + viral vector encoding for haemoagglutinin (AB + HA, n = 16), and AB + viral vector encoding for SOCS1 (AB + SOCS1, n = 18). Echocardiographic and haemodynamic measurements were performed 15 weeks after banding. While SOCS3 was upregulated during the hypertrophic phase, SOCS1 transcript levels increased significantly between 15 and 20 weeks. Remodelling was markedly worse in AB + SOCS1, showed larger left ventricular internal dimensions (+16%), higher end-diastolic pressures (+57%) and wall stress (+45%), and reduced fractional shortening (-32%) compared with AB + HA; apoptotic rate was increased three-fold and the gp130 pathway was inhibited. Ex vivo experiments showed that mechanical stretch upregulated SOCS1 expression, which was in turn attenuated by tumour necrosis factor-α (TNF-α) inhibition. CONCLUSION: Enhanced SOCS1 myocardial signalling is associated with accelerated transition from hypertrophy to failure in an established model of pressure overload. SOCS1 may represent an attractive target for the prevention of heart failure progression.


Assuntos
Receptor gp130 de Citocina/metabolismo , Técnicas de Transferência de Genes , Insuficiência Cardíaca/etiologia , Hipertrofia Ventricular Esquerda/etiologia , Janus Quinase 1/metabolismo , Miocárdio/enzimologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Angiopoietina-2/metabolismo , Animais , Apoptose , Dependovirus/genética , Modelos Animais de Doenças , Progressão da Doença , Vetores Genéticos , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Hemodinâmica , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Hipertrofia Ventricular Esquerda/enzimologia , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/fisiopatologia , Janus Quinase 1/genética , Masculino , Miocárdio/patologia , Fosforilação , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Fator de Transcrição STAT3/genética , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Fatores de Tempo , Ultrassonografia , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo , Função Ventricular Esquerda
14.
Diabetes ; 61(4): 944-53, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22344560

RESUMO

Insulin resistance is a recently identified mechanism involved in the pathophysiology of chronic heart failure (CHF). We investigated the effects of two insulin-sensitizing drugs (metformin and rosiglitazone) in a genetic model of spontaneously hypertensive, insulin-resistant rats (SHHF). Thirty SHHF rats were randomized into three treatment groups as follows: 1) metformin (100 mg/kg per day), 2) rosiglitazone (2 mg/kg per day), and 3) no drug. Ten Sprague-Dawley rats served as normal controls. At the end of the treatment period (12 months), the cardiac phenotype was characterized by histology, echocardiography, and isolated perfused heart studies. Metformin attenuated left ventricular (LV) remodeling, as shown by reduced LV volumes, wall stress, perivascular fibrosis, and cardiac lipid accumulation. Metformin improved both systolic and diastolic indices as well as myocardial mechanical efficiency, as shown by improved ability to convert metabolic energy into mechanical work. Metformin induced a marked activation of AMP-activated protein kinase, endothelial nitric oxide synthase, and vascular endothelial growth factor and reduced tumor necrosis factor-α expression and myocyte apoptosis. Rosiglitazone did not affect LV remodeling, increased perivascular fibrosis, and promoted further cardiac lipid accumulation. In conclusion, long-term treatment with metformin, but not with rosiglitazone, prevents the development of severe CHF in the SHHF model by a wide-spectrum interaction that involves molecular, structural, functional, and metabolic-energetic mechanisms.


Assuntos
Insuficiência Cardíaca/prevenção & controle , Hipoglicemiantes/farmacologia , Resistência à Insulina , Metformina/farmacologia , Animais , Glicemia , Pressão Sanguínea , Doença Crônica , Regulação da Expressão Gênica/efeitos dos fármacos , Hipoglicemiantes/uso terapêutico , Insulina , Metformina/uso terapêutico , Ratos , Ratos Endogâmicos SHR , Ratos Sprague-Dawley , Rosiglitazona , Tiazolidinedionas/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Remodelação Ventricular/efeitos dos fármacos
15.
J Clin Endocrinol Metab ; 94(3): 817-26, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19066295

RESUMO

CONTEXT: The loss of lean body mass (LBM) negatively influences the outcome in bariatric surgery. Impaired GH secretion is frequent in obese patients. OBJECTIVE: Our objective was to investigate if GH treatment prevents LBM loss in the early postoperative period. DESIGN: This was an open, prospective, randomized, and controlled study. PATIENTS: A total of 24 women (body mass index: 44.4 +/- 7.6 kg/m(2), aged 36.8 +/- 11.7 yr) undergoing laparoscopic-adjustable silicone gastric banding (LASGB) and with GH deficiency after LASGB was included in the study. TREATMENT PROTOCOL: Group A (n = 12) included a standardized diet regimen and exercise program plus recombinant human GH (0.5 +/- 0.13 mg every day), and group B (n = 12) included a standardized diet regimen and exercise program. The follow-up duration was 6 months. RESULTS: The excess of body weight loss did not differ between groups A and B after 3 and 6 months. At 3 months, LBM loss was lower (P < 0.0001) and fat mass (FM) loss was higher (P = 0.02) in group A than group B. At 3 and 6 months, appendicular skeletal muscle mass loss was lower (P = 0.000) in group A than group B. At 3 (P = 0.0003 and 0.0005, respectively) and 6 months (P < 0.0001 and 0.0002, respectively), the percent changes of FM and lean body mass were significantly higher in group A than group B. In both groups fasting and postglucose area under the plasma concentration-time curve insulin significantly reduced. The homeostasis model assessment of insulin and insulin sensitivity indexes and total to high-density lipoprotein cholesterol ratio improved only in group A. CONCLUSIONS: GH treatment for 6 months after LASGB reduces loss in LBM and appendicular skeletal muscle mass during a standardized program of low-calorie diet and physical exercise program, with improvement of lipid profile and without a deterioration of glucose tolerance.


Assuntos
Composição Corporal , Gastroplastia/métodos , Hormônio do Crescimento Humano/uso terapêutico , Obesidade Mórbida/cirurgia , Adolescente , Adulto , Índice de Massa Corporal , Metabolismo Energético , Feminino , Humanos , Fator de Crescimento Insulin-Like I/análise , Laparoscopia , Pessoa de Meia-Idade , Obesidade Mórbida/metabolismo , Projetos Piloto , Estudos Prospectivos
16.
Eur Heart J ; 24(7): 664-72, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12657225

RESUMO

AIM: Since growth hormone (GH) has proven beneficial in experimental heart failure, and the natural history of Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) is frequently complicated by the development of dilated cardiomyopathy, we administered GH to six patients with DMD and 10 with BMD, with the evidence of cardiac involvement. METHODS AND RESULTS: Patients were randomized to receive for 3 months either placebo or recombinant human GH, in a double-blind fashion. In GH-treated patients, left ventricular (LV) mass increased by 16% in BMD and by 29% in DMD (both p<0.01), with a significant increase of relative wall thickness (+19%). Systemic blood pressure remained unchanged, while LV end-systolic stress fell significantly by 13% in BMD and by 33% in DMD, with a slight increase of systolic function indexes. No changes were observed related to cardiac arrhythmias and skeletal muscle function in the patient groups during the treatment period, nor any side effects were observed. Brain natriuretic peptide, interleukin-6, and tumor necrosis factor-alpha circulating levels were elevated at baseline. While brain natriuretic peptide decreased by 40%, cytokine levels did not exhibit significant variations during the treatment period. CONCLUSIONS: The 3-month GH therapy in patients with DMD and BMD induces a hypertrophic response associated with a significant reduction of brain natriuretic peptide plasma levels and a slight improvement of systolic function, no changes in skeletal muscle function, and no side effects.


Assuntos
Hormônio do Crescimento Humano/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Adolescente , Adulto , Análise de Variância , Cardiomegalia , Criança , Método Duplo-Cego , Eletrocardiografia , Cardiopatias/complicações , Cardiopatias/tratamento farmacológico , Cardiopatias/fisiopatologia , Humanos , Fator de Crescimento Insulin-Like I/análise , Interleucina-6/sangue , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/fisiopatologia , Peptídeo Natriurético Encefálico/sangue , Análise de Regressão , Fator de Necrose Tumoral alfa/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA