Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Biol Reprod ; 102(1): 156-169, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-31504222

RESUMO

Gonadotropes represent approximately 5-15% of the total endocrine cell population in the mammalian anterior pituitary. Therefore, assessing the effects of experimental manipulation on virtually any parameter of gonadotrope biology is difficult to detect and parse from background noise. In non-rodent species, applying techniques such as high-throughput ribonucleic acid (RNA) sequencing is problematic due to difficulty in isolating and analyzing individual endocrine cell populations. Herein, we exploited cell-specific properties inherent to the proximal promoter of the human glycoprotein hormone alpha subunit gene (CGA) to genetically target the expression of a fluorescent reporter (green fluorescent protein [GFP]) selectively to ovine gonadotropes. Dissociated ovine pituitary cells were cultured and infected with an adenoviral reporter vector (Ad-hαCGA-eGFP). We established efficient gene targeting by successfully enriching dispersed GFP-positive cells with flow cytometry. Confirming enrichment of gonadotropes specifically, we detected elevated levels of luteinizing hormone (LH) but not thyrotropin-stimulating hormone (TSH) in GFP-positive cell populations compared to GFP-negative populations. Subsequently, we used next-generation sequencing to obtain the transcriptional profile of GFP-positive ovine gonadotropes in the presence or absence of estradiol 17-beta (E2), a key modulator of gonadotrope function. Compared to non-sorted cells, enriched GFP-positive cells revealed a distinct transcriptional profile consistent with established patterns of gonadotrope gene expression. Importantly, we also detected nearly 200 E2-responsive genes in enriched gonadotropes, which were not apparent in parallel experiments on non-enriched cell populations. From these data, we conclude that CGA-targeted adenoviral gene transfer is an effective means for selectively labeling and enriching ovine gonadotropes suitable for investigation by numerous experimental approaches.


Assuntos
Estradiol/farmacologia , Gonadotrofos/efeitos dos fármacos , Adeno-Hipófise/efeitos dos fármacos , Adenoviridae , Animais , Gonadotrofos/metabolismo , Hormônio Luteinizante/metabolismo , Adeno-Hipófise/metabolismo , Ovinos , Tireotropina/metabolismo
2.
Biol Reprod ; 93(6): 146, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26559679

RESUMO

The antiviral activity of interferon (IFN) increases in uterine vein serum (UVS) during early pregnancy in sheep. This antiviral activity in UVS collected on Day 15 of pregnancy is blocked by anti-IFN-tau (anti-IFNT) antibodies. Conceptus-derived IFNT was hypothesized to induce IFN-stimulated gene (ISG) expression in endometrium and extrauterine tissues during pregnancy. To test this hypothesis, blood was collected from ewes on Days 12-16 of the estrous cycle or pregnancy. Serum progesterone was >1.7 ng/ml in pregnant (P) and nonpregnant (NP) ewes until Day 13, then declined to <0.6 ng/ml by Day 15 in NP ewes. A validated IFNT radioimmunoassay detected IFNT in uterine flushings (UFs) on Days 13-16 and in UVS on Days 15-16 of pregnancy. IFNT detection in UF correlated with paracrine induction of ISGs in the endometrium and occurred prior to the inhibition of estrogen receptor 1 and oxytocin receptor expression in uterine epithelia on Day 14 of pregnancy. Induction of ISG mRNAs in corpus luteum (CL) and liver tissue occurred by Day 14 and in peripheral blood mononuclear cells by Day 15 in P ewes. Expression of mRNAs for IFN signal transducers and ISGs were greater in the CL of P than that of NP ewes on Day 14. It is concluded that: 1) paracrine actions of IFNT coincide with detection of IFNT in UF; 2) endocrine action of IFNT ensues through induction of ISGs in peripheral tissues; and 3) IFNT can be detected in UVS, but not until Days 15-16 of pregnancy, which may be limited by the sensitivity of the IFNT radioimmunoassay.


Assuntos
Corpo Lúteo/metabolismo , Endométrio/metabolismo , Interferon Tipo I/metabolismo , Proteínas da Gravidez/metabolismo , Animais , Receptor alfa de Estrogênio/metabolismo , Ciclo Estral/metabolismo , Feminino , Leucócitos Mononucleares/metabolismo , Gravidez , Progesterona/metabolismo , Receptores de Ocitocina/metabolismo , Ovinos
3.
Biol Reprod ; 86(6): 171, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22423050

RESUMO

The current dogma is that the differential regulation of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) synthesis and secretion is modulated by gonadotropin-releasing hormone (GnRH) pulse frequency and by changes in inhibins, activins, and follistatins both at the pituitary and at the peripheral level. To date no studies have looked at the overlapping function of these regulators in a combined setting. We tested the hypothesis that changes in GnRH pulse frequency alter the relative abundance of these regulators at the pituitary and peripheral levels in a manner consistent with changes in pituitary and circulating concentrations of FSH; that is, an increase in FSH will be accompanied by increased stimulatory input (activin) and/or reduced follistatin and inhibin. Ovariectomized ewes were subjected to a combination hypothalamic pituitary disconnection (HPD)-hypophyseal portal blood collection procedure. Hypophyseal portal and jugular blood samples were collected for a 6-h period from non-HPD ewes, HPD ewes, or HPD ewes administered GnRH hourly or every 3 h for 4 days. In the absence of endogenous hypothalamic and ovarian hormones that regulate gonadotropin secretion, 3-hourly pulses of GnRH increased pituitary content of FSH more than hourly GnRH, although these differences were not evident in the peripheral circulation. The results failed to support the hypothesis in that the preferential increase of pituitary content of FSH by the lower GnRH pulse frequency could be explained by changes in the pituitary content of inhibin A, follistatin, or activin B. Perhaps the effects of GnRH pulse frequency on FSH is due to changes in the balance of free versus bound amounts of these FSH regulatory proteins or to the involvement of other regulators not monitored in this study.


Assuntos
Ativinas/sangue , Hormônio Foliculoestimulante/metabolismo , Folistatina/sangue , Hormônio Liberador de Gonadotropina/metabolismo , Inibinas/sangue , Animais , Feminino , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/sangue , Hormônio Liberador de Gonadotropina/sangue , Hipotálamo/metabolismo , Hormônio Luteinizante/sangue , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Ovinos
4.
Biol Reprod ; 85(4): 770-8, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21734267

RESUMO

Estradiol-17beta (E2) is the major regulator of GnRH receptor (GnRHR) gene expression and number during the periovulatory period; however, the mechanisms underlying E2 regulation of the GNRHR gene remain undefined. Herein, we find that E2 conjugated to BSA (E2-BSA) mimics the stimulatory effect of E2 on GnRH binding in primary cultures of ovine pituitary cells. The time course for maximal GnRH analog binding was similar for both E2 and E2-BSA. The ability of E2 and E2-BSA to increase GnRH analog binding was blocked by the estrogen receptor (ER) antagonist ICI 182,780. Also, increased GnRH analog binding in response to E2 and the selective ESR1 agonist propylpyrazole triol was blocked by expression of a dominant-negative form of ESR1 (L540Q). Thus, membrane-associated ESR1 is the likely candidate for mediating E2 activation of the GNRHR gene. As cAMP response element binding protein (CREB) is an established target for E2 activation in gonadotrophs, we next explored a potential role for this protein as an intracellular mediator of the E2 signal. Consistent with this possibility, adenoviral-mediated expression of a dominant-negative form of CREB (A-CREB) completely abolished the ability of E2 to increase GnRH analog binding in primary cultures of ovine pituitary cells. Finally, the presence of membrane-associated E2 binding sites on ovine pituitary cells was demonstrated using a fluorescein isothiocyanate conjugate of E2-BSA. We suggest that E2 regulation of GnRHR number during the preovulatory period reflects a membrane site of action and may proceed through a nonclassical signaling mechanism, specifically a CREB-dependent pathway.


Assuntos
Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Adeno-Hipófise/metabolismo , Receptores LHRH/metabolismo , Transdução de Sinais , Regulação para Cima , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Estradiol/análogos & derivados , Antagonistas de Estrogênios/metabolismo , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/genética , Feminino , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Fase Folicular/metabolismo , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/metabolismo , Cinética , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/metabolismo , Adeno-Hipófise/citologia , Adeno-Hipófise/efeitos dos fármacos , Proteínas Recombinantes , Soroalbumina Bovina/metabolismo , Carneiro Doméstico , Transdução de Sinais/efeitos dos fármacos , Propriedades de Superfície/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
5.
Biol Reprod ; 85(6): 1152-60, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21753192

RESUMO

Fertility control is a potential method for managing overabundant wildlife populations; however, current technology is limited by duration of treatment efficacy and unacceptable side effects. The objective of this study was to determine the efficacy of a single immunization with gonadotropin-releasing hormone (GnRH) vaccine to suppress reproductive function in pregnant female elk and to evaluate potential behavioral and pathological side effects of treatment. Eighteen captive adult female elk were randomly allocated to one of two experimental groups. Ten females were administered a conjugated and adjuvanted GnRH vaccine intramuscularly, and eight elk received an adjuvant sham vaccine without conjugated GnRH. We compared success of existing pregnancy, neonatal survival, subsequent fertility, reproductive behavior rates, and side effects of treatment between January 2006 and January 2010. The GnRH vaccination did not affect existing pregnancy or calf survival during the year that it was applied; however, it reduced the proportion of pregnant females for 3 yr. Male precopulatory behavior rates exhibited toward GnRH-vaccinated females tended to be greater than those directed at sham-vaccinated females during the second half of the breeding season, when GnRH vaccinates continued to be proceptive. Strong immune and inflammatory responses, including robust GnRH antibody concentrations in GnRH vaccinates, and sterile pyogranulomatous injection site abscesses in both groups, were consistent with vaccination. In conclusion, this GnRH vaccine resulted in prolonged, albeit reversible, impairment of fertility, and is associated with extended reproductive behaviors and partial suppression of hypothalamic-pituitary-gonadal axis function in captive female elk.


Assuntos
Cervos/fisiologia , Fertilidade/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/administração & dosagem , Gravidez/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Vacinas Anticoncepcionais/farmacologia , Animais , Animais de Zoológico , Anticoncepção Imunológica , Feminino , Hormônio Liberador de Gonadotropina/efeitos adversos , Hormônio Liberador de Gonadotropina/imunologia , Masculino , Distribuição Aleatória
6.
Reprod Biol Endocrinol ; 8: 40, 2010 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-20459750

RESUMO

BACKGROUND: We demonstrated that E2 conjugated to BSA (E2BSA) induces a rapid membrane-initiated inhibition of LH secretion followed hours later by a slight increase in LH secretion. Whether these actions of E2BSA are restricted to the pituitary gland and whether the membrane-initiated pathway of E2BSA contributes to the up-regulation of the number of GnRH receptors during the positive feedback effect of E2 were evaluated here. We have shown that the suppression of LH secretion induced by E2 and E2BSA is the result of a decreased responsiveness of the pituitary gland to GnRH. In this study we further tested the ability of E2BSA to decrease the responsiveness of the pituitary gland to GnRH under the paradigm of the preovulatory surge of LH induced by E2. METHODS: For the first experiment GnRH and LH secretions were determined in samples of pituitary portal and jugular blood, respectively, in ewes treated with 12 mg E2BSA. In the second experiment, the number of GnRH receptors was quantified in ewes 12 h after administration of 25 micrograms E2 (the expected time for the increase in the number of GnRH receptors and the positive feedback effect of E2 in LH secretion) or 12 mg E2BSA. In the third experiment, the preovulatory-like surge of LH was characterized in ewes injected with 25 micrograms E2 alone or followed 8 h later (before the beginning of the LH surge) with 60 mg E2BSA. RESULTS: a) the decrease in LH secretion induced by E2BSA was not accompanied by changes in the pulsatile pattern of GnRH, b) E2BSA increased the number of GnRH receptors, and c) the presence of E2BSA in E2-treated ewes delayed the onset, reduced the length, and decreased the amount of LH released during the preovulatory surge of LH. CONCLUSIONS: a) the rapid suppression of LH secretion induced by E2BSA is mediated only via a direct action on the pituitary gland, b) E2 acting via a membrane-initiated pathway contributes to increase the number of GnRH receptors and, c) administration of E2BSA near the beginning of the pre-ovulatory surge of LH delays and reduces the magnitude of the surge.


Assuntos
Membrana Celular/efeitos dos fármacos , Estradiol/farmacologia , Hormônio Luteinizante/metabolismo , Ovinos/metabolismo , Animais , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Estradiol/sangue , Estradiol/farmacocinética , Feminino , Hormônio Liberador de Gonadotropina/sangue , Hormônio Liberador de Gonadotropina/metabolismo , Ovariectomia , Ovulação/efeitos dos fármacos , Ovulação/metabolismo , Fluxo Pulsátil/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Soroalbumina Bovina/farmacocinética , Soroalbumina Bovina/farmacologia
7.
Theriogenology ; 157: 199-209, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32814247

RESUMO

To more clearly understand the equine gonadotrope response to kisspeptin and gonadotropin releasing hormone (GnRH), peripheral LH and FSH were quantified in diestrous mares after treatment with either equine kisspeptide (eKp-10, 0.5 mg iv), GnRH (25 µg iv), or a combination thereof every 4 h for 3 days. The following observations were made: 1) a diminished LH and FSH response to eKp-10 and GnRH was observed by Day 3, but was not different by treatment, 2) a decrease in basal LH concentration was observed from Day 1 to Day 3 for the eKp-10, but not the GnRH treated mares, 3) there was no change in basal FSH with either treatment. Additionally, pre-treatment with GnRH antagonist (antide 1.0 mg iv) eliminated any measurable change in LH after eKp-10 (1.0 mg iv) treatment. Both GnRH and kisspeptin are Gαq/11 coupled receptors, therefore quantifying the rise in intracellular calcium following treatment with cognate ligand allows simultaneous assessment of receptor activation. Direct stimulation of equine primary pituitary cells with GnRH and/or eKp-10 demonstrates three distinct populations of pituitary cells: one population responded to both eKp-10 and GnRH, a second, independent population, responded to only eKp-10, and a third population responded only to GnRH. These populations were confirmed using co-immunofluorescence of hemipituitaries from mares in diestrus. Although the rise in peripheral LH concentration elicited by eKp-10 is dependent on GnRH, this work suggests that kisspeptin also has a specific and direct effect on the equine gonadotrope, independent of GnRH.


Assuntos
Kisspeptinas , Hormônio Luteinizante , Animais , Feminino , Hormônio Foliculoestimulante , Hormônio Liberador de Gonadotropina/metabolismo , Cavalos , Kisspeptinas/fisiologia , Hipófise/metabolismo
8.
Reprod Biol Endocrinol ; 7: 42, 2009 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-19432978

RESUMO

BACKGROUND: Classically, progesterone has been thought to act only through the well-known genomic pathway involving hormone binding to nuclear receptors and subsequent modulation of gene expression. However, there is increasing evidence for rapid, non-genomic effects of progesterone in a variety of mammalian tissues and it is possible that a membrane PR (mPR) is causing these events. We recently isolated and characterized an ovine mPR referred to as mPR-alpha, distinct from the nuclear PR. Based on predicted structural analysis, the ovine mPR-alpha possesses seven transmembrane domains typical of G protein-coupled receptors. Despite the homology to other reported mPRs, information pertaining to the steroid binding characteristics of the ovine mPR-alpha was lacking. Additionally, the ovine mPR-alpha transcript has been identified in the hypothalamus, pituitary, uterus, ovary and corpus luteum, yet changes in expression of the ovine mPR-alpha in these tissues were not known. Consequently, the purpose of this work was to determine the steroid binding characteristics of the ovine mPR-alpha and to investigate possible changes in expression of the ovine mPR-alpha in reproductive tissues throughout the estrous cycle. METHODS: Binding studies were performed using crude membrane fractions from CHO cells expressing the mPR-alpha. Using quantitative Real-time PCR we determined the expression pattern of mRNA for the ovine mPR-alpha during the ovine estrous cycle in tissues known to express the mPR-alpha. Jugular blood samples were also collected and analyzed for serum concentrations of P4 to ensure ewes were at the appropriate stage of their cycle. RESULTS: Only progesterone, 20alpha-hydroxyprogesterone and 17alpha-hydroxyprogesterone were able to displace binding of 3H-P4 (P < 0.001) to membrane fractions from CHO cells expressing ovine mPR-alpha. The average B-max and Kd values for three separate experiments were 624 +/- 119 fmol/micro gram protein and 122 +/- 50 nM, respectively. Significant changes in expression of mRNA for the mPR-alpha during the estrous cycle were noted in the corpus luteum and uterus. CONCLUSION: The mPR-alpha specifically binds progestins and its expression was correlated to progesterone secretion during the ovine estrous cycle. Results from the present studies suggest that mPR-alpha may have an important physiological role during the ovine estrous cycle.


Assuntos
Ciclo Estral/fisiologia , Progesterona/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Animais , Ligação Competitiva/fisiologia , Células CHO , Cricetinae , Cricetulus , Feminino , Hipotálamo/fisiologia , Proteínas de Membrana/metabolismo , Ovário/fisiologia , Hipófise/fisiologia , RNA Mensageiro/metabolismo , Ovinos , Trítio , Útero/fisiologia
9.
Am J Reprod Immunol ; 82(2): e13146, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31206871

RESUMO

PROBLEM: Current methods to quantify kisspeptin (KP) are limited. To this end, a radioimmunoassay (RIA) specific for KP was developed and validated. We hypothesized that use of a RIA would reveal multiple hypothalamic regions as targets of negative seasonal feedback of estradiol on KP production in sheep. METHOD OF STUDY: Ovariectomized (OVX) ewes bearing a subcutaneous implant of estradiol were euthanized during the breeding season (BS) (n = 4) and non-breeding season (NBS) (n = 3). Coronal sections of preoptic area (POA), anterior hypothalamic area (AHA), and mediobasal hypothalamus (MBH) were collected, as well as the median eminence (ME), cortex, brain stem, and cerebellum. Amounts of KP and gonadotropin-releasing hormone (GnRH) in individual hypothalamic nuclei were quantified by radioimmunoassay. RESULTS: Concentration and content of KP were lower during the NBS than the BS in the MBH (P < 0.01) and POA (P < 0.01). Levels of KP in tissue adjacent to the POA and MBH were much lower, and neither concentration nor content of KP differed between the BS and NBS. Kisspeptin was also detected in the cortex, brain stem, and cerebellum, but concentrations were not affected by season. In addition, concentration and content of GnRH in the POA, AHA, MBH, and ME were similar between seasons. CONCLUSION: Our RIA results indicate that in addition to the MBH, the POA and AHA appear to be involved in the seasonal negative feedback of estradiol on KP expression.


Assuntos
Córtex Cerebelar/metabolismo , Estradiol/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo Médio/metabolismo , Kisspeptinas/metabolismo , Área Pré-Óptica/metabolismo , Ovinos/metabolismo , Animais , Cruzamento , Retroalimentação Fisiológica/fisiologia , Feminino , Radioimunoensaio , Estações do Ano
10.
Endocrinology ; 149(2): 767-73, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17962347

RESUMO

Stress-like elevations in plasma glucocorticoids rapidly inhibit pulsatile LH secretion in ovariectomized sheep by reducing pituitary responsiveness to GnRH. This effect can be blocked by a nonspecific antagonist of the type II glucocorticoid receptor (GR) RU486. A series of experiments was conducted to strengthen the evidence for a mediatory role of the type II GR and to investigate the neuroendocrine site and cellular mechanism underlying this inhibitory effect of cortisol. First, we demonstrated that a specific agonist of the type II GR, dexamethasone, mimics the suppressive action of cortisol on pituitary responsiveness to GnRH pulses in ovariectomized ewes. This effect, which became evident within 30 min, documents mediation via the type II GR. We next determined that exposure of cultured ovine pituitary cells to cortisol reduced the LH response to pulse-like delivery of GnRH by 50% within 30 min, indicating a pituitary site of action. Finally, we tested the hypothesis that suppression of pituitary responsiveness to GnRH in ovariectomized ewes is due to reduced tissue concentrations of GnRH receptor. Although cortisol blunted the amplitude of GnRH-induced LH pulses within 1-2 h, the amount of GnRH receptor mRNA or protein was not affected over this time frame. Collectively, these observations provide evidence that cortisol acts via the type II GR within the pituitary gland to elicit a rapid decrease in responsiveness to GnRH, independent of changes in expression of the GnRH receptor.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hidrocortisona/metabolismo , Hipófise/citologia , Receptores LHRH/genética , Receptores LHRH/metabolismo , Animais , Células Cultivadas , Dexametasona/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Glucocorticoides/farmacologia , Hidrocortisona/farmacologia , Ovariectomia , Hipófise/fisiologia , RNA Mensageiro/metabolismo , Ovinos
11.
PLoS One ; 13(7): e0201570, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30063758

RESUMO

Wildlife and humans are increasingly competing for resources worldwide, and a diverse, innovative, and effective set of management tools is needed. Controlling abundance of wildlife species that are simultaneously protected, abundant, competitive for resources, and in conflict with some stakeholders but beloved by others, is a daunting challenge. Free-ranging horses (Equus caballus) present such a conundrum and managers struggle for effective tools for regulating their abundance. Controlling reproduction of female horses presents a potential alternative. During 2009-2017, we determined the long-term effectiveness of GnRH vaccine (GonaCon-Equine) both as a single immunization and subsequent reimmunization on reproduction and side effects in free-ranging horses. At a scheduled management roundup in 2009, we randomly assigned 57 adult mares to either a GonaCon-Equine treatment group (n = 29) or a saline control group (n = 28). In a second roundup in 2013, we administered a booster vaccination to these same mares. We used annual ground observations to estimate foaling proportions, social behaviors, body condition, and injection site reactions. We found this vaccine to be safe for pregnant females and neonates, with no overt deleterious behavioral side effects during the breeding season. The proportion of treated mares that foaled following a single vaccination was lower than that for control mares for the second (P = 0.03) and third (P = 0.08) post-treatment foaling seasons but was similar (P = 0.67) to untreated mares for the fourth season, demonstrating reversibility of the primary vaccine treatment. After two vaccinations, however, the proportion of females giving birth was lower (P <0.001) than that for control mares for three consecutive years and ranged from 0.0-0.16. The only detectable adverse side effect of vaccination was intramuscular swelling at the vaccination site. Regardless of vaccine treatment (primary/secondary), approximately 62% (34/55) of immunized mares revealed a visible reaction at the vaccine injection site. However, none of these mares displayed any evidence of lameness, altered gait or abnormal range of movement throughout the 8 years they were observed in this study. Our research suggests that practical application of this vaccine in feral horses will require an initial inoculation that may provide only modest suppression of fertility followed by reimmunization that together could result in greater reduction in population growth rates over time.


Assuntos
Anticoncepção Imunológica , Eficácia de Contraceptivos , Hormônio Liberador de Gonadotropina/imunologia , Cavalos , Imunização Secundária , Vacinas Anticoncepcionais/uso terapêutico , Animais , Animais Selvagens , Anticoncepção Imunológica/efeitos adversos , Anticoncepção Imunológica/métodos , Anticoncepção Imunológica/veterinária , Feminino , Cavalos/imunologia , Imunização Secundária/efeitos adversos , Imunização Secundária/métodos , Imunização Secundária/veterinária , Gravidez , Distribuição Aleatória , Vacinação/efeitos adversos , Vacinação/métodos , Vacinação/veterinária
12.
Anim Reprod Sci ; 101(3-4): 265-75, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17055196

RESUMO

The objectives of this study were: to determine if peripheral administration of the ovine C-terminal decapeptide (oKiSS-1) increases secretion of LH and FSH in ovariectomized (OVX) ewes; to evaluate the effect of ovarian steroid hormones on the predicted increase in gonadotropin secretion induced by oKiSS-1; to establish if the stimulatory effect of oKiSS-1 is due to a direct action on the hypothalamus or was mediated directly at the anterior pituitary gland. Intravenous injection of 3mg oKiSS-1 to OVX ewes (n=6) increased (P<0.01) basal and mean serum concentrations of LH. The amplitude and the area under the curve (AUC) of LH induced by oKiSS-1 was greater (P<0.01) than the highest pulse of LH detected before oKiSS-1. Pre-treatment with steroid hormones (1mg progesterone plus 50 microg estradiol-17beta for 3 days) in OVX ewes (n=6) reduced (P<0.1) the magnitude of the stimulatory effect of oKiSS-1 on LH secretion. In these animals, oKiSS-1 increased (P<0.1) basal and mean concentrations of LH, and the amplitude of the pulse of LH induced by oKiSS-1, but not the AUC (P>0.1). In our hands, oKiSS-1 failed to increase serum concentration of FSH at the dose used in this study. Administration of GnRH antiserum completely suppressed the secretory patterns of LH and prevented the increase in secretion of LH induced by oKiSS-1. We conclude that the rapid increase in secretion of LH induced by peripheral administration of oKiSS-1 is the result of a direct action on the hypothalamus.


Assuntos
Hipotálamo/efeitos dos fármacos , Hormônio Luteinizante/metabolismo , Ovinos/fisiologia , Proteínas Supressoras de Tumor/farmacologia , Sequência de Aminoácidos , Animais , Estrogênios/farmacologia , Feminino , Hormônio Liberador de Gonadotropina/farmacologia , Hipotálamo/metabolismo , Dados de Sequência Molecular , Ovariectomia/veterinária , Progesterona/farmacologia , Fatores de Tempo , Proteínas Supressoras de Tumor/química
13.
Theriogenology ; 95: 8-17, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28460684

RESUMO

In previous work, an EP2 prostanoid receptor (EP2R) agonist in vivo increased mRNA expression of luteal LH receptors (LHR), unoccupied and occupied luteal; LHR, and circulating progesterone, while an EP3R or FPR agonist decreased; mRNA expression of luteal LHR, unoccupied and occupied luteal LHR, and; circulating progesterone. An EP4R and lysophosphatidic acid (LPA) LPA2R and LPA3R agonists were reported to inhibit luteal function and sirtuins have been proposed to increase prostaglandin synthesis. The objectives were to determine; whether an EP4R, LPA2R, or LPA3R agonist affect ovine luteal function in vivo or; in vitro. In addition, whether sirtuin (SIRT)-1, 2, or 3; LPA2R or LPA3R; or EP1R, EP2R, EP3R, or EP4R agonists affect caruncular endometrial PGF2α or PGE (PGE1+PGE2) secretion in vitro. Day-10 nonpregnant ewes received a single injection of Vehicle (N = 5); an LPA2R (N = 5); LPA3R (N = 6); or EP4R (N = 5) agonist given into the interstitial tissue of the ovarian vascular pedicle adjacent to the luteal-containing ovary to determine effects on circulating progesterone, mRNA expression of luteal LHR, and luteal unoccupied and occupied LHR. In addition, agonists for LPA2R, LPA3R, EP1R, EP2R, EP3R, or EP4R or SIRT-1, SIRT-2, or SIRT-3 activators were incubated with caruncular endometrial slices in vitro to determine their effect on caruncular endometrial PGF2α, or PGE secretion. LPA2R, LPA3R, or an EP4R agonist in vivo did not affect (P ≥ 0.05) luteal weight, circulating progesterone, or occupied luteal LHR. However, an LPA2R or EP4R agonist, but; not LPA3R agonist, in vivo increased (P ≤ 0.05) mRNA expression of luteal LHR. An; LPA2R, LPA3R, or EP4R agonist increased (P ≤ 0.05) luteal unoccupied LHR, but; not occupied LHR. An LPA2R, LPA3R, or an EP4R agonist did not affect (P ≥ 0.05); luteal progesterone secretion in vitro. An LPA2R or LPA3R agonist did not affect (P ≥ 0.05) luteal PGF2α, or PGE secretion in vitro. However, an EP4R agonist tended to decrease (P < 0.066) luteal PGF2α secretion and increased (P ≤ 0.05) luteal PGE; secretion in vitro. EP1R, EP2R, EP3R, or an EP4R agonist did not affect (P ≥ 0.05); caruncular endometrial PGF2α secretion in vitro. However, EP1R, EP3R, or an EP4R agonist increased caruncular endometrial PGE secretion in vitro, while two different EP2R agonists did not affect (P ≥ 0.05) caruncular endometrial PGE; secretion. A SIRT-1 activator, but not SIRT-2 or SIRT-3 activators, increased (P ≤ 0.05) caruncular endometrial PGE secretion, while sirtuin 1, 2, or 3 activators did not affect (P ≥ 0.05) caruncular endometrial PGF2α secretion. In conclusion, receptors for EP4, LPA2, and LPA3 do not appear to be involved; in luteolysis, but EP4R and LPA2R might participate in preventing luteolysis by maintaining luteal mRNA expression for LHR and preventing loss of unoccupied luteal LHR. In addition, SIRT-1, EP1R, EP3R, and EP4R might be involved in; regulating caruncular endometrial PGE secretion, but not PGF2α secretion.


Assuntos
Corpo Lúteo/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Prostaglandinas E/metabolismo , Receptores de Ácidos Lisofosfatídicos/agonistas , Receptores de Prostaglandina E Subtipo EP4/agonistas , Receptores de Prostaglandina E/agonistas , Sirtuínas/farmacologia , Alprostadil/metabolismo , Animais , Corpo Lúteo/fisiologia , Dinoprosta/metabolismo , Endométrio/fisiologia , Feminino , Lisofosfolipídeos/agonistas , Lisofosfolipídeos/antagonistas & inibidores , Progesterona/sangue , Progesterona/metabolismo , RNA Mensageiro/metabolismo , Ovinos
14.
Mol Endocrinol ; 19(10): 2624-38, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15961508

RESUMO

Recent studies profiling immediate early gene responses to GnRH in the LbetaT2 gonadotrope cell model revealed increased expression of numerous genes including activating transcription factor (ATF) 3. The present studies demonstrate similar results with GnRH administration in vivo in ovariectomized mice. In this model, ATF3 mRNA was markedly up-regulated at 20, 40, and 60 min after in vivo administration of a GnRH analog. In alphaT3-1 gonadotrope cells, ATF3 mRNA and protein were induced by GnRH in a manner consistent with in vivo observations. Pharmacological studies implicated a combined role for the activities of protein kinase C isozymes, ERK and c-Jun N-terminal kinase, in modulating ATF3 expression. The role of ATF3 was further investigated in the activation of the human glycoprotein hormone alpha-subunit gene promoter. GnRH induced the alpha-subunit promoter-luciferase reporter approximately 16-fold, and this induction was completely abolished with mutations in the dual cAMP response elements (CREs) or the combined inhibition of GnRH-induced ERK and c-Jun N-terminal kinase. GnRH induced recruitment of ATF3, c-Jun, and c-Fos to the dual CREs. Overexpression and specific knockdown of ATF3 by small inhibitory RNA implicate a functional role for ATF3 in mediating activation of the alpha-subunit gene promoter. These studies provide clear evidence that ATF3 is a key immediate early gene induced by GnRH administration in vivo and in the alphaT3-1 gonadotrope cell model. These studies support the conclusion that the dual CREs of the human alpha-subunit promoter are the target of GnRH-induced MAPK regulation through ATF3.


Assuntos
Genes Precoces , Subunidade alfa de Hormônios Glicoproteicos/genética , Hormônio Liberador de Gonadotropina/farmacologia , Fatores de Transcrição/genética , Fator 3 Ativador da Transcrição , Animais , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , DNA/genética , DNA/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Hipófise/citologia , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteína Quinase C/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
15.
Cancer Res ; 64(6): 2090-5, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15026348

RESUMO

Pokeweed antiviral protein (PAP) is a plant-derived, highly potent ribosome inactivating protein that causes inhibition of protein translation and rapid cell death. We and others have delivered this protein to various cell types, including cancer cells, using hormones to specifically target cells bearing the hormone receptor. Here, we compare binding and cytotoxicity of GnRH-PAP hormonotoxins prepared either by protein conjugation (GnRH-PAP conjugate) or through recombinant DNA technology (GnRH-PAP fusion). Although GnRH-PAP conjugate protein bound specifically to and caused cell death in cells bearing the gonadotropin-releasing hormone (GnRH) receptor, we could not detect binding or cytotoxicity using two different versions of the fusion protein in receptor-positive cells. We conclude that generation of an active GnRH-PAP fusion protein may not be feasible either because both ends of the GnRH molecule are required for receptor binding, but only the NH(2) terminus is free in the fusion protein and/or that more potent analogues of GnRH (inclusion of which is not feasible in the fusion protein) are needed for efficient targeting. In contrast, the GnRH-PAP conjugate shows promise as a novel anticancer agent, capable of targeting cancer cells expressing the GnRH receptor such as prostate, breast, ovarian, endometrial, and pancreatic cells. It may also be useful as a therapeutic agent to eliminate pituitary gonadotrophs, eliminating the need for chronic GnRH analogue administration to treat hormone-sensitive diseases.


Assuntos
Imunotoxinas/toxicidade , N-Glicosil Hidrolases/metabolismo , Proteínas de Plantas/metabolismo , Receptores LHRH/metabolismo , Proteínas Recombinantes de Fusão/toxicidade , Animais , Apoptose , Western Blotting , Células CHO , Bovinos , Membrana Celular/metabolismo , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Cricetinae , Humanos , Hipófise/metabolismo , Ligação Proteica , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Inativadoras de Ribossomos Tipo 1
16.
J Wildl Dis ; 41(4): 758-67, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16456165

RESUMO

Practical application of fertility control technology in free-ranging wild ungulates often requires remote delivery of the contraceptive agent. The objective of this investigation was to evaluate the potential of remote delivery of leuprolide acetate for suppressing fertility in female elk (Cervus elaphus nelsoni). Fifteen captive adult female elk were randomly allocated to one of three experimental groups. Six elk were injected intramuscularly with a dart containing leuprolide, and the remaining nine elk received the same formulation without leuprolide. We determined pregnancy rates, suppression of luteinizing hormone (LH) and progesterone concentrations, and reversibility of treatments during 1 August 2002 to 3 September 2003. Leuprolide formulation caused a decrease in concentrations of LH and progesterone, temporary suppression of ovulation and steroidogenesis, and effective contraception (100%) for one breeding season. These results extend the practical application of this contraceptive agent to include dart delivery, where in the absence of such technology, wild elk must first be captured and restrained before treatment.


Assuntos
Anticoncepção/veterinária , Anticoncepcionais Femininos/farmacologia , Cervos , Leuprolida/farmacologia , Animais , Animais Selvagens , Anticoncepção/métodos , Anticoncepcionais Femininos/administração & dosagem , Cervos/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Injeções Intramusculares/veterinária , Leuprolida/administração & dosagem , Hormônio Luteinizante/sangue , Masculino , Gravidez , Taxa de Gravidez , Progesterona/sangue , Distribuição Aleatória , Resultado do Tratamento
17.
Endocrinology ; 144(4): 1456-63, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12639929

RESUMO

Pokeweed antiviral protein (PAP), a 29-kDa ribosome-inactivating protein isolated from the leaves of Phytolacca americana, has potent cytotoxic activity once it enters the cytoplasm of a cell. It is incapable of entering cells by itself. Therefore, our objective was to determine whether a GnRH analog could be used to deliver PAP specifically to cells expressing GnRH receptors. D-Lys(6)-GnRH-Pro(9)-ethylamide was conjugated to PAP (GnRH-PAP). Chinese hamster ovary cells stably transfected with cDNA for the murine GnRH receptor and a mouse gonadotroph tumor cell line that expresses endogenous GnRH receptors (alphaT3-1 cells) were used to evaluate the cytotoxic effects of GnRH-PAP. We also examined cytotoxicity of GnRH-PAP using human endometrial, breast, and prostate cancer cell lines. Treatment of GnRH receptor-positive cells with GnRH-PAP resulted in dose-dependent cytotoxicity. Cytotoxicity of GnRH-PAP was dependent on number of GnRH receptors (r(2) = 0.871, P < 0.05) and duration of exposure of GnRH-PAP to the cells. In contrast, GnRH-PAP was not cytotoxic to Chinese hamster ovary cells not harboring GnRH receptors. Moreover, the cytotoxic activity of GnRH-PAP could be inhibited by addition of excess GnRH analog. Neither PAP nor GnRH analog alone was cytotoxic. These results suggest that GnRH analogs can be used to specifically deliver toxin molecules to cells that express GnRH receptors. Thus, a new class of biomedicines that act as hormonotoxins against cells expressing GnRH receptors provides a novel approach for inhibiting reproduction and treating cancers that are dependent on reproductive hormones.


Assuntos
Hormônio Liberador de Gonadotropina/toxicidade , N-Glicosil Hidrolases/toxicidade , Proteínas de Plantas/toxicidade , Receptores LHRH/genética , Animais , Antineoplásicos/farmacologia , Células CHO , Bovinos , Cricetinae , Expressão Gênica , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Técnicas In Vitro , N-Glicosil Hidrolases/metabolismo , Proteínas de Plantas/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Receptores LHRH/metabolismo , Proteínas Inativadoras de Ribossomos Tipo 1 , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos
18.
J Wildl Dis ; 40(4): 713-24, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15650089

RESUMO

Fertility control offers a potential alternative for controlling an abundance of wild ungulate populations where lethal methods are infeasible or unacceptable. A promising nonsteroidal, nonimmunologic approach to reversible contraception consists of agonist of gonadotropin-releasing hormone (GnRH). We evaluated the effects of the GnRH agonist, leuprolide, on reproduction, the suppression of luteinizing hormone (LH) and progesterone, blood parameters, and reproductive behavior in captive female mule deer (Odocoileus hemionus) during December 1999 through June 2001. Leuprolide, administered as a controlled release formulation (ATRIGEL), was 100% effective in preventing pregnancy for one breeding season. Infertility was achieved by suppressing LH levels, which prevented ovulation and the formation of corpus luteum. Treated females regained normal ovarian function and conceived the following breeding season. Leuprolide had no adverse effects on blood chemistry and hematology, body weight dynamics, or the general health of treated females. In contrast to our predictions, leuprolide did not suppress estrous behavior in female deer during the "normal" breeding period, nor did treated females return to normal ovarian function and exhibit reproductive behaviors during the post-breeding period. This prolonged-release leuprolide formulation offers an alternative approach to reversible contraception in female deer that overcomes some of the problems associated with existing technology.


Assuntos
Anticoncepção/veterinária , Cervos/fisiologia , Fármacos para a Fertilidade Feminina/farmacologia , Hormônio Liberador de Gonadotropina/agonistas , Leuprolida/farmacologia , Ovulação/efeitos dos fármacos , Animais , Cruzamento/métodos , Anticoncepção/métodos , Preparações de Ação Retardada , Feminino , Hormônio Luteinizante/sangue , Controle da População/métodos , Gravidez , Progesterona/sangue
19.
Theriogenology ; 82(9): 1224-30, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25219846

RESUMO

Previously, it was reported that intraluteal implants containing prostaglandin E1 or E2 (PGE1 and PGE2) in Angus or Brahman cows prevented luteolysis by preventing loss of mRNA expression for luteal LH receptors and luteal unoccupied and occupied LH receptors. In addition, intraluteal implants containing PGE1 or PGE2 upregulated mRNA expression for FP prostanoid receptors and downregulated mRNA expression for EP2 and EP4 prostanoid receptors. Luteal weight during the estrous cycle of Brahman cows was reported to be lesser than that of Angus cows but not during pregnancy. The objective of this experiment was to determine whether intraluteal implants containing PGE1 or PGE2 alter vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2), angiopoietin-1 (ANG-1), and angiopoietin-2 (ANG-2) protein in Brahman or Angus cows. On Day 13 of the estrous cycle, Angus cows received no intraluteal implant and corpora lutea were retrieved, or Angus and Brahman cows received intraluteal silastic implants containing vehicle, PGE1, or PGE2 on Day 13 and corpora lutea were retrieved on Day 19. Corpora lutea slices were analyzed for VEGF, FGF-2, ANG-1, and ANG-2 angiogenic proteins via Western blot. Day-13 Angus cow luteal tissue served as preluteolytic controls. Data for VEGF were not affected (P > 0.05) by day, breed, or treatment. PGE1 or PGE2 increased (P < 0.05) FGF-2 in luteal tissue of Angus cows compared with Day-13 and Day-19 Angus controls but decreased (P < 0.05) FGF-2 in luteal tissue of Brahman cows when compared w Day-13 or Day-19 Angus controls. There was no effect (P > 0.05) of PGE1 or PGE2 on ANG-1 in Angus luteal tissue when compared with Day-13 or Day-19 controls, but ANG-1 was decreased (P < 0.05) by PGE1 or PGE2 in Brahman cows when compared with Day-19 Brahman controls. ANG-2 was increased (P < 0.05) on Day 19 in Angus Vehicle controls when compared with Day-13 Angus controls, which was prevented (P < 0.05) by PGE1 but not by PGE2 in Angus cows. There was no effect (P > 0.05) of PGE1 or PGE2 on ANG-2 in Brahman cows. PGE1 or PGE2 may alter cow luteal FGF-2, ANG-1, or ANG-2 but not VEGF to prevent luteolysis; however, species or breed differences may exist.


Assuntos
Alprostadil/farmacologia , Indutores da Angiogênese/metabolismo , Corpo Lúteo/efeitos dos fármacos , Dinoprostona/farmacologia , Implantes de Medicamento/farmacologia , Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Animais , Bovinos , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Biol Reprod ; 77(2): 320-8, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17429013

RESUMO

The objectives of this study were to determine whether activation of estrogen receptor 1 (ESR1; also known as ERalpha), or estrogen receptor 2 (ESR2; also known as ERbeta), or both are required to: 1) acutely inhibit secretion of LH, 2) induce the preovulatory-like surge of LH, and 3) inhibit secretion of FSH in ovariectomized (OVX) ewes. OVX ewes (n = 6) were administered intramuscularly 25 micrograms estradiol (E2), 12 mg propylpyrazoletriol (PPT; a subtype-selective ESR1 agonist), 21 mg diaprylpropionitrile (DPN; a subtype-selective ESR2 agonist), or PPT + DPN. Like E2, administration of PPT, DPN, or combination of the two rapidly decreased (P < 0.05) secretion of LH. Each agonist induced a gradual, prolonged rise in secretion of LH after the initial inhibition, but neither agonist alone nor the combined agonists was able to induce a "normal" preovulatory-like surge of LH similar to that induced by E2. Compared with E2-treated ewes, the beginning of the increase in secretion of LH occurred earlier (P < 0.01) in DPN-treated ewes, later (P < 0.05) in PPT-treated ewes, and at a similar interval in ewes receiving the combined agonist treatment. Like E2, PPT decreased (P < 0.05) secretion of FSH, but the duration of suppression was much longer in PPT-treated ewes. DPN did not alter secretion of FSH in this study. Modulation of the number of GnRH receptors by PPT and DPN was examined in primary cultures of ovine pituitary cells. In our hands, both PPT and DPN increased the number of GnRH receptors, but the dose of DPN required to stimulate synthesis of GnRH receptors was 10 times higher than that of PPT. We conclude that in OVX ewes: 1) ESR1 and ESR2 mediate the negative feedback of E2 on secretion of LH at the level of the pituitary gland, 2) ESR1 and ESR2 do not synergize or antagonize the effects of each other; however, they do interact to synchronize the beginning of the stimulatory effect of E2 on secretion of LH, 3) ESR1 and ESR2 may mediate at least partially the positive feedback of E2 on LH secretion by increasing the number of GnRH receptors, and 4) only ESR1 appears to be involved in the negative feedback of E2 on secretion of FSH.


Assuntos
Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Hormônio Foliculoestimulante/metabolismo , Hormônio Luteinizante/metabolismo , Ovinos , Animais , Células Cultivadas , Interações Medicamentosas , Estradiol/administração & dosagem , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Retroalimentação Fisiológica , Feminino , Nitrilas/administração & dosagem , Ovariectomia , Fenóis , Adeno-Hipófise/química , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Pirazóis/administração & dosagem , Receptores LHRH/análise , Receptores LHRH/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA