Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 650
Filter
Add more filters

Publication year range
1.
Reprod Domest Anim ; 59(4): e14555, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38546114

ABSTRACT

Most of the northern hemisphere donkey breeds are faced with the risk of extinction, thus donkey reproduction is considered an emerging branch of theriogenology, and the management of artificial insemination and induction of ovulation is a crucial point in setting up preservation protocols. For four consecutive cycles, six jennies' ovarian activity was routinely monitored; an ultrasound examination was performed daily from the evidence of a follicle diameter ≥27 mm until ovulation. Upon reaching a follicular diameter ≥32 ± 2 mm (Hour 0), oestrous jennies were treated alternatively with 0.1 mg triptorelin acetate, sc, (TRIP), 0.4 mg/sc of buserelin acetate (BUS) or saline, sc, (CTRL) and examined ultrasonographically at Hours 14, 24, 38, 42, 48, 62 and every 24 h until ovulation. Induction of ovulation was considered successful if ovulation occurred from 24 to 48 h after treatment. 11/12 cycles resulted in ovulation for TRIP and 12/12 for BUS and CTRL groups, respectively. Mean ± SD ovulation time after treatment was 37.3 ± 8.3, 47.1 ± 21.0 and 66.8 ± 25.9 h for BUS, TRIP and CTRL, respectively (p = .0032). Ovulation rates between h24 and h48 were 10/12 (83.3%) for both TRIP/BUS and 2/12 (16.7%) for CTRL, respectively (p = .003). Buserelin and triptorelin-treated jennies had a 25 times higher probability to ovulate between Hours 24 and 48 than controls (p = .003), while there were no jenny and cycle effects on the ovulatory rate. The results of this study show how triptorelin successfully induced ovulation in jennies, like other GnRH analogues previously evaluated.


Subject(s)
Equidae , Triptorelin Pamoate , Female , Animals , Triptorelin Pamoate/pharmacology , Ovulation , Buserelin/pharmacology , Ovulation Induction/veterinary , Ovulation Induction/methods , Acetates/pharmacology , Gonadotropin-Releasing Hormone/pharmacology
2.
Reprod Domest Anim ; 59(10): e14734, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39392191

ABSTRACT

This study evaluated the effect of prostaglandin F2α (PGF2α) associated with gonadotropin-releasing hormone (GnRH) for ovulation induction in precocious indicus heifers submitted to a fixed-time superovulation (SOV) programme. Precocious Nellore heifers (n = 35), aged 13 months, were subjected to the SOV protocol. On day 0 (D0), all animals received intravaginal insertion of a progesterone (P4) device along with intramuscular administration of 2 mg of oestradiol benzoate, plus 200 IU of follicle-stimulating hormone in decreasing doses, with 12-h intervals between D4 and D7, in addition to 150 µg of D-cloprostenol on D6 and device removal on D7. On D8, the donors received 10.5 µg of buserelin acetate and the treatment group received 300 µg of D-cloprostenol/PGF2α. Artificial insemination was performed 12 h and 24 h after GnRH administration using frozen semen. On D15 of the protocol (i.e., D7 after insemination), the embryos were collected and evaluated. All animals passed through the control and treatment groups. Results were evaluated by analysis of variance using an adjusted mixed-effects model (p < 0.05). There was no difference in the total number of embryos between the control and treatment groups (10.40 ± 1.52 vs. 9.60 ± 1.36; p = 0.63) or viable embryos (6.30 ± 1.22 vs. 4.30 ± 0.71). For precocious indicus heifers, treatment with PGF2α in association with GnRH did not affect embryo production in the fixed-time SOV protocol.


Subject(s)
Dinoprost , Estradiol , Gonadotropin-Releasing Hormone , Insemination, Artificial , Ovulation Induction , Progesterone , Superovulation , Animals , Cattle , Female , Dinoprost/pharmacology , Dinoprost/administration & dosage , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/administration & dosage , Superovulation/drug effects , Insemination, Artificial/veterinary , Ovulation Induction/veterinary , Ovulation Induction/methods , Estradiol/pharmacology , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Progesterone/pharmacology , Progesterone/administration & dosage , Pregnancy , Cloprostenol/pharmacology , Cloprostenol/administration & dosage , Buserelin/pharmacology , Buserelin/administration & dosage , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/administration & dosage
3.
Anim Biotechnol ; 34(8): 3867-3876, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37477621

ABSTRACT

This study aimed to characterize estrus response and to establish relationships between intensity of estrus, preovulatory follicle (POF) size and estradiol (E2) concentrations on day of AI, luteal profiles and pregnancy outcome in lactating Hariana breed of cows. 200 cyclic cows were subjected to Ovsynch (n = 54) and Pre-OV treatment (n = 146). Ovsynch: Buserelin acetate (BA; 10 µg), Cloprostenol (500 µg) and BA (10 µg) were injected i.m. on day 0, 7 and 9, respectively, irrespective of treatment. Pre-OV: BA (10 µg) and Cloprostenol (500 µg) was also injected i.m. simultaneously 7 days prior to initiate Ovsynch. On the basis of estrus behavior, the cows were classified into three groups: weak, moderate and intense. Artificial insemination performed at 18-24 hours after 2nd BA of Ovsynch in both treatments. The average duration of estrus did not differ (p > 0.05) between Ovsynch and Pre-OV treatment. A positive correlation was observed between estrus response and POF size, concentration of E2 on day of AI and luteal profiles on day 12 post-AI. First service conception rate was higher in cows exhibited intense (45.46%) and moderate (42.56%) estrus response than weak (28.57%) estrus response. In conclusion, intensity of estrus expression could be considered as important determinant for deciding pregnancy outcomes in Bos indicus cows.


Subject(s)
Estrus Synchronization , Lactation , Female , Pregnancy , Cattle , Animals , Lactation/physiology , Estrus/physiology , Fertility , Buserelin/pharmacology , Cloprostenol , Progesterone
4.
Reprod Domest Anim ; 58(2): 314-322, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36349902

ABSTRACT

The aim of the present study was to determine the impact of different strategies for increasing the level of serum progesterone (P4) on luteal morphology and function in bovine females. The effects of increasing P4 on pregnancy rate and gestational loss (GL) in Nelore cows subjected to timed artificial insemination (TAI) were also evaluated. A total of 939 cows were divided into three groups: P4LA (n = 305), 150 mg of long-acting injectable P4 7 days after TAI; GnRH (n = 306), 10 µg of buserelin acetate 7 days after TAI; and control (n = 328), no hormone treatment after TAI. Doppler ultrasound assessments and P4 measurements were performed on days 7 and 16 after TAI. The pregnancy rate and GL as a function of treatment were compared using the SAS GLIMMIX procedure. Corpus luteum (CL) vascular perfusion, volume, and plasma P4 concentration were analysed using the SAS PROC MIXED procedure. No significant difference was found among the treatments in terms of volume, number of pixels, and CL intensity or in the serum P4 concentration at 7 days after ovulation. The CL blood flow at 16 days after ovulation was lower in the P4LA and GnRH groups than that in the control group (p < .01). Serum concentrations of P4 at 16 days after ovulation were higher in the P4LA and GnRH groups than those in the control group (p = .04). A difference in the pregnancy rate (p = .003) and a trend in GL (p = .07) as a function of treatment were found. Overall, long-acting injectable P4 supplementation on day 7 after TAI or GnRH administration affected CL vascularization and increased the serum concentrations of P4 16 days after ovulation, promoting better pregnancy rates than the control.


Subject(s)
Fertility , Progesterone , Pregnancy , Female , Cattle , Animals , Progesterone/pharmacology , Fertility/physiology , Estrus Synchronization/methods , Corpus Luteum , Ovulation/physiology , Gonadotropin-Releasing Hormone/pharmacology , Insemination, Artificial/veterinary , Insemination, Artificial/methods , Buserelin/pharmacology , Lactation/physiology , Dinoprost/pharmacology
5.
Trop Anim Health Prod ; 55(1): 19, 2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36542229

ABSTRACT

This study is aimed at assessing the impact of simultaneous administration of GnRH and prostaglandin F2α (PGF2α) 7 days prior to Ovsynch in Hariana cow. Two hundred cyclic cows (> 4 months postpartum) were assigned to control (n = 54) and pre-OV (n = 146). As per Ovsynch protocol, buserelin acetate (10 µg), cloprostenol (500 µg), and buserelin acetate (10 µg) were injected i.m. on days 0, 7, and 9, respectively, in cows irrespective of treatment. But in pre-OV cows, buserelin acetate (10 µg) and cloprostenol (500 µg) were also injected i.m. simultaneously 7 days prior to initiate the Ovsynch protocol. Artificial insemination was performed between 18 and 24 h after the 2nd GnRH of Ovsynch in both treatments. Ultrasonography and blood sampling for hormonal analysis were done on each day of treatment, on day of AI, and 12 days post-AI. Pre-OV treatment resulted to increased (45.20% vs 29.62%; P < 0.05) pregnancy outcomes and higher (P < 0.01) ovulation rate to first GnRH of Ovsynch than control. Cows showing complete luteolysis in response to PGF2α of Ovsynch were also higher (P < 0.05) in pre-OV than control. Greater (P < 0.05) synchronization rate was recorded in pre-OV than control (86.76% and 68.75%). The circulating concentrations of estradiol on day of AI and progesterone on day 12 post-AI were higher (P < 0.01) in cows diagnosed pregnant than non-pregnant in both control and pre-OV treatment. In conclusion, simultaneous administration of GnRH and PGF2α 7 days before Ovsynch improved the synchronization rate and luteal profile in terms of CL area and hence resulted in higher conception rate in Hariana zebu cow.


Subject(s)
Dinoprost , Gonadotropin-Releasing Hormone , Pregnancy , Female , Cattle , Animals , Buserelin/pharmacology , Estrus Synchronization/methods , Progesterone , Insemination, Artificial/veterinary , Insemination, Artificial/methods , Cloprostenol/pharmacology , Lactation
6.
Reprod Domest Anim ; 55(3): 374-383, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31930759

ABSTRACT

The aim of this study was to assess the efficacy of different doses of buserelin acetate and another GnRH agonist, triptorelin acetate, in saline solution in a single subcutaneous injection, to induce ovulation of growing pre-ovulatory follicle in mare and compare it with the classical treatment of a single injection of hCG. The study is split into 3 experiments over different breeding seasons in the same stud with a random distribution of treatment. The first one was to compare the injection of 6 mg of buserelin with 1,500 IU of hCG; the second one consisted of comparing different doses of buserelin (6 mg and 3 mg); and the third one compared three different doses of buserelin (3, 2 and 1 mg), 0.1 mg of triptorelin with 1,500 IU of hCG as a control group. The results of all experiments showed the same efficacy between all treatments with mares ovulating between 24 and 48 hr after injection: experiment 1: hCG (78% n = 41) and buserelin 6 mg (90% n = 50); experiment 2: buserelin 6 mg (78,1% n = 192) and buserelin 3 mg (78% n = 341); and experiment 3: hCG (87% n = 106), buserelin 3 mg (84,7% n = 137), buserelin 2 mg (82,7% n = 104), buserelin 1 mg (87% n = 54) and triptorelin 0.1 mg (84,7% n = 72). In conclusion, this study contributes to erasing the dogma that has been established since 1975 that a single injection in solution without any long-acting excipient of a GnRH agonist cannot induce ovulation in the mare. This study also shows that a injection of 0.1 mg of triptorelin in solution is a good alternative for ovulation induction and is comparable to small doses of buserelin acetate in solution (1 mg) and 1,500 IU of the gold standard trigger hCG, mainly in countries where human formulation of buserelin is not available.


Subject(s)
Buserelin/pharmacology , Fertility Agents, Female/pharmacology , Horses/physiology , Ovulation Induction/veterinary , Triptorelin Pamoate/pharmacology , Animals , Breeding , Buserelin/administration & dosage , Chorionic Gonadotropin/administration & dosage , Chorionic Gonadotropin/pharmacology , Female , Fertility Agents, Female/administration & dosage , Injections, Subcutaneous/veterinary , Ovulation Induction/methods , Triptorelin Pamoate/administration & dosage
7.
Anim Biotechnol ; 30(2): 99-104, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30595097

ABSTRACT

The present study was aimed to determine the effect of GnRH analog (buserelin acetate) on the quality of bovine spermatozoa stored at 16° C for 24 h. Semen collected in the summer season from June to September from healthy Polish Holstein-Friesian bulls. Ejaculates were centrifuged, divided and diluted to the final concentration of 240 × 106 spermatozoa/mL using animal protein-free commercial BIOXcell® extender (IMV Technologies, L'aigle, France) (Control) or with BIOXcell® extender supplemented with buserelin acetate and stored 0, 8 and 24 h. Sperm motility parameters analysis was performed using a computer-assisted sperm analysis (CASA) system. The viability of spermatozoa was performed using flow cytometer. The addition of buserelin acetate to BIOXcell® extender did positively affect the total motility (was higher in the observed samples with the addition of 2 µg/mL and 4 µg/mL than in the control group), progressive motile (forward progressing sperm was significantly increased (p < 0.05) over the control group at the 0 h and 8 h of incubation following the supplementation of 2, 4 and 8 µg/mL buserelin acetate) and viability of spermatozoa (the number of live spermatozoa was significantly higher (p < 0.05) in 2 µg/mL and 4 µg/mL samples with buserelin acetate at 8th hour of incubation and in sample with 4 µg/mL at 24th hour of incubation compared to the control group). We recommend adding 4 µg/mL to the extender to improve the quality of bovine semen.


Subject(s)
Buserelin/pharmacology , Cattle/physiology , Gonadotropin-Releasing Hormone/analogs & derivatives , Sperm Motility/drug effects , Animals , Cell Survival/drug effects , Male , Semen/drug effects , Semen Analysis/veterinary , Spermatozoa/drug effects , Spermatozoa/physiology
8.
J Obstet Gynaecol ; 38(8): 1183-1185, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29560760

ABSTRACT

We evaluated the efficacy of gonadotropin-releasing hormone agonist (GnRHa) therapy for improving the myometrial thickness in women with thin (less than 1 cm) uterine walls, a contraindication for microwave endometrial ablation (MEA). The normal myometrium thickness was 0.5 cm, 0.7 cm and 0.9 cm. After the third GnRHa dose, the myometrial thickness increased to over 1 cm in all the three patients, and all were able to undergo MEA. The VAS score for menorrhagia improved in all the cases. The patient satisfaction levels were 10 in 2 of the 3 patients, and 5 in the other. There was no symptom recurrence, and no adjuvant therapy was administered. GnRHa therapy in women with submucous leiomyomata and a myometrial thickness of less than 1 cm could effectively thicken the myometrium, allowing for the use of MEA.


Subject(s)
Buserelin/pharmacology , Endometrial Ablation Techniques , Myometrium/drug effects , Female , Gonadotropin-Releasing Hormone/agonists , Humans , Menorrhagia/surgery , Microwaves/therapeutic use , Middle Aged , Preoperative Care , Retrospective Studies
9.
Reprod Domest Anim ; 52(1): 76-82, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27696569

ABSTRACT

Fixed-time post-cervical artificial insemination (FTAI) drastically reduces labour requirements and increases the use of boars with higher genetic merit. This study evaluated the efficiency of eCG administration combined with/without the GnRH agonist buserelin for the induction and synchronization of ovulation in weaned sows submitted to FTAI. The sows were allocated into three groups. In the control group, the first artificial insemination was performed at the onset of oestrus and repeated every 24 hr. In the eCG+GnRH group, sows received 600 IU eCG at weaning and buserelin (10 µg) after 86-89 hr of eCG, and in the GnRH group, sows received only buserelin after 86-89 hr of weaning. The hormone-treated sows received a single FTAI after 30-33 hr of buserelin application. All the sows were inseminated with homospermic doses (1.5 × 109  sperm cells/50 ml). The interval between weaning and ovulation was shorter (p < .05) in the eCG+GnRH (133.3 hr) and GnRH (135.9 hr) groups than the control (141.5 hr) group. In the eCG+GnRH group, the sows ovulated earlier (p < .05) than those in the GnRH group (44.5 vs. 48.2 hr after buserelin administration). The reproductive performance of GnRH sows was not compromised when only sows exhibiting oestrus at the time of insemination were considered, but lower farrowing rate and smaller litter size were observed in eCG+GnRH sows. The reproductive performance of eCG+GnRH sows was primarily compromised because the insemination was performed outside the optimal time relative to ovulation; therefore, it is advisable to inseminate them before 116-122 hr after weaning.


Subject(s)
Buserelin/pharmacology , Estrus/drug effects , Gonadotropins, Equine/pharmacology , Insemination, Artificial/veterinary , Ovulation Induction/veterinary , Ovulation/drug effects , Animals , Drug Therapy, Combination , Estrus Detection , Female , Litter Size/drug effects , Logistic Models , Male , Ovary/diagnostic imaging , Pregnancy , Pregnancy Rate , Swine , Ultrasonography , Weaning
10.
Trop Anim Health Prod ; 49(2): 259-263, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27888394

ABSTRACT

A total of 60 animals (38 cows, 22 heifers) were selected and were divided into three groups of 20 animals each (containing both anoestrus and repeat breeder) in which treatment was performed for 60 days. Group I: control (farmer practice), T1 group: group I + hormone (double synch), and T2 group: group I + hormone (Estra double synch). The growth performances were measured in terms of body weight and average daily gain (ADG). Blood collection was done at the start and end of the experiment for assessment of blood biochemical, hematological, and reproductive status of the animals. Results revealed significant improvement in growth and reproductive performances in treatment group as compared to control group. Higher percentage of conception was achieved in group III (60%) followed by group II (55%). The least percentage was in group I (15%), i.e., in control group. So it was found that the effect of treating the reproductive-disordered animals with Estra double synch gave comparatively better result than double synch hormonal application.


Subject(s)
Buserelin/pharmacology , Cattle/physiology , Dairying/methods , Dinoprost/analogs & derivatives , Estradiol/analogs & derivatives , Estrus Synchronization/drug effects , Reproductive Control Agents/pharmacology , Animals , Cattle/growth & development , Dinoprost/pharmacology , Estradiol/pharmacology , Female , India , Insemination, Artificial/veterinary , Reproduction
11.
Eur Surg Res ; 57(1-2): 22-33, 2016.
Article in English | MEDLINE | ID: mdl-27089503

ABSTRACT

BACKGROUND: Gonadotropin-releasing hormone (GnRH), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) are involved in the reproductive cycle and regulate the secretion of sex steroids from the gonads. In mammals, GnRH1 is secreted as a hormone from the hypothalamus, whereas both GnRH1 and GnRH2 are present as neuropeptides in a variety of tissues. This review describes the role of GnRH in the gastrointestinal tract. SUMMARY: GnRH1, GnRH2, and LH receptors in humans and rats, and GnRH receptors in rats, have been described in the gastrointestinal tract, where they affect motility, gastric and hormone secretion, and cell proliferation. GnRH analogs are clinically used in the treatment of sex hormone-dependent diseases, i.e., endometriosis and malignancies, and as pretreatments for in vitro fertilization. Severe gastrointestinal dysmotility has been shown to develop in some women after such treatment, along with a reduction in the number of enteric neurons and autoantibodies against GnRH. Consequently, a rat model of enteric neurodegeneration has been developed based on the administration of the GnRH analog buserelin. Serum IgM antibodies against GnRH1, the GnRH2 precursor progonadoliberin-2, and the GnRH receptor have also been described in patients with irritable bowel syndrome and dysmotility, as well as in patients with gastrointestinal disorders associated with diabetes mellitus, posterior laryngitis, and primary Sjögren's syndrome, although no treatments using GnRH analogs have been administered. CONCLUSION: GnRH and receptors for GnRH and LH are present in the human and rat gastrointestinal tract. Treatment with GnRH analogs may induce severe dysmotility, and a rat model of enteric neurodegeneration has been developed based on stimulation by the GnRH analog buserelin. Autoantibodies against GnRH and its receptor are found in a subgroup of patients with functional bowel disorders and dysmotility, independent of treatment with GnRH analogs.


Subject(s)
Gastrointestinal Tract/physiology , Gonadotropin-Releasing Hormone/physiology , Animals , Antibody Formation , Buserelin/pharmacology , Gastrointestinal Microbiome , Gastrointestinal Motility , Gonadotropin-Releasing Hormone/immunology , Humans , Rats , Receptors, LHRH/physiology
12.
Reprod Domest Anim ; 51(4): 478-84, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27170495

ABSTRACT

This study was designed to investigate the impact of buserelin acetate (BA) or human chorionic gonadotropin (hCG) administration on the day of first artificial insemination (AI) on subsequent luteal profile (diameter of corpus luteum (CL) and plasma progesterone) and conception rate in Murrah buffalo. The present experiment was carried out at two locations in 117 buffalo that were oestrus-synchronized using cloprostenol (500 µg) administered (i.m.) 11 days apart followed by AI during standing oestrus. Based on treatment (i.m.) at the time of AI, buffalo were randomly categorized (n = 39 in each group) into control (isotonic saline solution, 5 ml), dAI-BA (buserelin acetate, 20 µg) and dAI-hCG (hCG, 3000 IU) group. Out of these, 14 buffalo of each group were subjected to ovarian ultrasonography on the day of oestrus to monitor the preovulatory follicle and on days 5, 12, 16 and 21 post-ovulation to monitor CL diameter. On the day of each sonography, jugular vein blood samples were collected for the estimation of progesterone concentrations. All the buffalo (n = 117) were confirmed for pregnancy on day 40 post-ovulation. The conception rate was better (p < 0.05) in dAI-BA (51.3%) and dAI-hCG (66.7%) groups as compared to their control counterparts (30.8%). Furthermore, the buffalo of dAI-hCG group had improved (p < 0.05) luteal profile, whereas the buffalo of dAI-BA group failed (p > 0.05) to exhibit stimulatory impact of treatment on luteal profile when compared to control group. In brief, buserelin acetate or hCG treatment on the day of first AI leads to an increase in conception rate; however, an appreciable impact on post-ovulation luteal profile was observed only in hCG-treated Murrah buffalo.


Subject(s)
Buffaloes , Buserelin/pharmacology , Chorionic Gonadotropin/pharmacology , Corpus Luteum/drug effects , Insemination, Artificial/veterinary , Animals , Female , Ovulation/drug effects , Ovulation/physiology , Pregnancy , Pregnancy Rate
13.
Reprod Fertil Dev ; 27(8): 1181-6, 2015 Nov.
Article in English | MEDLINE | ID: mdl-24965784

ABSTRACT

Ovulation in camelids is induced by the seminal plasma protein ovulation-inducing factor (OIF), recently identified as ß-nerve growth factor (ß-NGF). The present study measured the total protein concentration in alpaca seminal plasma using a bicinchoninic acid (BCA) protein quantification assay and found it to be 22.2±2.0mgmL(-1). To measure the effects of varying doses of ß-NGF on the incidence and timing of ovulation, corpus luteum (CL) size and plasma progesterone concentration, 24 female alpacas were synchronised and treated with either: (1) 1mL 0.9% saline (n=5); (2) 4µg buserelin (n=5); (3) 1mg ß-NGF protein (n=5); (4) 0.1mg ß-NGF (n=5); or (5) 0.01mg ß-NGF (n=4). Females were examined by transrectal ultrasonography at 1-2-h intervals between 20 and 45h after treatment or until ovulation occurred, as well as on Day 8 to observe the size of the CL, at which time blood was collected to measure plasma progesterone concentrations. Ovulation was detected in 0/5, 5/5, 5/5, 3/5 and 0/4 female alpacas treated with saline, buserelin, 1, 0.1 and 0.01mg ß-NGF, respectively. Mean ovulation interval (P=0.76), CL diameter (P=0.96) and plasma progesterone concentration (P=0.96) did not differ between treatments. Mean ovulation interval overall was 26.2±1.0h. In conclusion, buserelin and 1mg ß-NGF are equally effective at inducing ovulation in female alpacas, but at doses ≤0.1mg, ß-NGF is not a reliable method for the induction of ovulation.


Subject(s)
Corpus Luteum/drug effects , Nerve Growth Factor/administration & dosage , Ovulation/drug effects , Progesterone/blood , Animals , Buserelin/pharmacology , Camelids, New World , Cloprostenol/pharmacology , Female , Male , Ovulation Induction/methods
14.
Reprod Domest Anim ; 49(6): 920-5, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25219366

ABSTRACT

Luteolysis before the time of maternal recognition of pregnancy is one cause of low fertility in high-producing dairy cows. The objective of this study was to assess whether induction of a secondary corpus luteum (CL) late in the luteal phase would delay the time of luteolysis. Twenty high-producing Holstein cows were synchronized to ovulation (Day 0) with the Ovsynch protocol and received hCG (1500 IU im) on Day 12. Corpora lutea formation (as evaluated by ultrasonography) and plasma P4 concentrations were monitored from Days 4 to 36. hCG treatment induced the formation of one secondary CL (CL2) in 11 of 20 cows (55%) from the dominant follicle (mean diameter: 14.2 ± 0.9 mm) of two-wave (3/11) and three-wave (8/11) cycles. The maximal diameter of the CL2 (23.3 ± 1.9 mm) was reached approximately 6 days after hCG treatment and was correlated with its structural lifespan (p < 0.01). Cows that formed a CL2 after hCG had higher mean plasma P4 concentrations on Day 14 (+4.5 ng/ml) and Day 18 (+3.0 ng/ml) compared with cows without CL2 (p < 0.05). The structural regression of CL2 begun approximately 8 days after that of the CL1, and the median time at which the first drop in circulating P4 levels occurred was later in cows that formed a CL2 than in those that did not (Day 26 vs Day 18; p < 0.01). Thus, the induction of a CL2 by hCG on Day 12 might reduce the risk of premature luteolysis in high-producing dairy cows after insemination.


Subject(s)
Cattle/physiology , Chorionic Gonadotropin/pharmacology , Corpus Luteum/physiology , Luteolysis/physiology , Ovulation/physiology , Animals , Buserelin/administration & dosage , Buserelin/pharmacology , Cloprostenol/administration & dosage , Cloprostenol/pharmacology , Estrous Cycle , Female , Lactation/physiology , Pregnancy , Seasons
15.
Theriogenology ; 215: 132-137, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38065020

ABSTRACT

To date, there have been no studies testing the capacity of GnRH analogs and respective doses to induce a LH peak in sheep. In this sense, the present study aimed to evaluate the capacity of different synthetic forms and doses of GnRH in inducing LH release in sheep, and the effect of GnRH administration at timed artificial insemination (TAI) on pregnancy per timed-AI. In experiment 1, ewes (n = 40) received an intravaginal device (IVD) of medroxyprogesterone acetate (MPA; 60 mg) for 7 d and prostaglandin F2α analog on Day 5. On Day 7, the ewes were allocated randomly into one of eight groups (n = 5/group), which received a GnRH analog at a specific dose, as follows: lecirelin (12.5 or 25 µg), gonadorelin (50 or 100 µg), buserelin acetate (4.2 or 8.4 µg), or deslorelin (375 or 750 µg). Blood samples for LH determination were obtained at 0, 2, 4, and 6 h after GnRH and the IVDs were removed after the last blood collection. The maximal LH concentration induced by gonadorelin at doses of 50 µg and 100 µg (12.0 ± 2.4 ng/mL and 28.6 ± 7.1 ng/mL, respectively) was lower (P < 0.05) than serum LH induced by 8.4 µg of buserelin (78.9 ± 12.9 ng/mL), 375 µg and 750 µg of deslorelin (75.6 ± 7.4 ng/mL and 72.1 ± 10.6 ng/mL, respectively) and 12.5 µg and 25 µg of lecirelin (73.3 ± 17.8 ng/mL and 61.6 ± 5.9 ng/mL, respectively). However, the maximal LH concentration induced by 4.2 µg of buserelin (49.4 ± 5.9 ng/mL) was similar (P > 0.05) to the 100 µg of gonadorelin. The total release of LH (area under the curve - AUC) after treatment with 50 µg of gonadorelin (31.7 ± 5.9 ng h/mL) was lower (P < 0.05) than after other agonists. In a second experiment, 330 ewes were treated with IVD containing MPA for 7 d. Simultaneously with IVD removal, 250 µg of cloprostenol and 200 IU of eCG were administered. Then, ewes were assigned randomly to either no further treatment (control); or to receive 4.2 µg of buserelin acetate (GnRH group) at cervical TAI, which was performed with fresh semen 54 h after IVD withdrawal in all the animals. Higher pregnancy per timed-AI was observed for GnRH (50.3 %) compared to control (40.7 %). We conclude that buserelin acetate (8.4 µg), lecirelin (12.5 and 25 µg) and deslorelin (375 and 750 µg) induced a greater stimulatory effect on LH secretion than gonadorelin treatment. Furthermore, buserelin acetate treatment at TAI increased pregnancy per timed-AI in ewes previously treated with MPA and eCG.


Subject(s)
Buserelin , Estrus Synchronization , Pregnancy , Female , Sheep , Animals , Buserelin/pharmacology , Gonadotropin-Releasing Hormone/pharmacology , Medroxyprogesterone Acetate/pharmacology , Insemination, Artificial/veterinary , Prostaglandins F/pharmacology , Progesterone , Dinoprost/pharmacology
16.
Domest Anim Endocrinol ; 88: 106854, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38733943

ABSTRACT

This study evaluated the efficiency of prostaglandin F2α (PGF) to hasten ovulation in weaned sows. In experiment I, weaned sows detected in estrus (0 h) received: no hormone (Control; n = 56); 0.5 mg PGF IM at 0 h and 2 h (PGF0; n = 56); or 0.5 mg PGF IM at 24 h and 26 h (PGF24; n = 55). In experiment II, weaned sows that did not express estrus signs until 72 h after weaning (0 h) were assigned to: no hormone (Control; n = 45); 10 µg buserelin acetate IM at 0 h (Buserelin; n = 43); 0.5 mg PGF IM at 34 h and 36 h (PGF; n = 44); or 10 µg buserelin acetate IM at 0 h plus 0.5 mg PGF IM at 34 h and 36 h (Buserelin + PGF; n = 45). In experiment I, no effect of PGF on the interval treatment onset to ovulation was observed (P > 0.05), and no treatment effect was observed on the relative or cumulative proportion of females that ovulated post-treatment onset (P > 0.05). In experiment II, treatment onset to ovulation interval was shorter for Buserelin group than for PGF group (P < 0.05), and a higher cumulative percentage of Buserelin treated sows ovulated up to 48 h compared to PGF and Control groups (P < 0.01), with no differences from Buserelin + PGF. Treatments did not affect total number of piglets born in both experiments (P > 0.05). In conclusion, PGF did not hasten ovulation timing or affect litter size in weaned sows.


Subject(s)
Buserelin , Dinoprost , Ovulation , Animals , Female , Dinoprost/pharmacology , Dinoprost/administration & dosage , Swine/physiology , Ovulation/drug effects , Ovulation/physiology , Buserelin/pharmacology , Buserelin/administration & dosage , Weaning , Ovulation Induction/veterinary , Ovulation Induction/methods
17.
Theriogenology ; 220: 77-83, 2024 May.
Article in English | MEDLINE | ID: mdl-38490112

ABSTRACT

The present study evaluated follicular and endocrine dynamics during ReBreed21, a reproductive strategy that allows resynchronization of ovulation every 21 days in Bos indicus (Nelore) heifers. A synchronized estrous cycle was induced using a standard timed ovulation protocol (d -10: P4 implant inserted + 2 mg estradiol benzoate; d -2: P4 removed+ 0.5 mg cloprostenol + 0.6 mg estradiol cypionate + 200 IU equine chorionic gonadotropin (eCG); d0: 8.4 µg buserelin) without AI to ensure nonpregnancy in heifers. Day of GnRH was designated d0 of estrous cycle. On d12, heifers (n = 80) were randomized into three experimental groups: (1) ReBreed21 (n = 28) d12 P4 device inserted, d19 P4 device withdrawal plus 200 IU eCG, and d21 8.4 µg buserelin (GnRH); (2) ReBreed21+G (n = 26) same as ReBreed21 plus GnRH (16.8 µg) treatment on d12; and (3) Control (n = 26) no treatment. ReBreed21+G increased two-fold (62.9%; 18/26) percentage of heifers with synchronized follicular wave emergence compared to Control (34.6%; 9/26) whereas ReBreed21 (53.6%; 15/28) was intermediate. The ReBreeed21 groups (eCG on d19) increased (P < 0.01) follicular growth between d19 and d21 in ReBreed21 (2.3 ± 0.2 mm) and ReBreed21+G (3.4 ± 0.2 mm) compared with Control (1.2 ± 0.3 mm), resulting in greater (P < 0.01) follicle diameter on d21 for ReBreed21 (10.7 ± 0.4 mm) and ReBreed21+G (10.8 ± 0.4 mm) compared with Control (9.1 ± 0.5 mm). Structural luteolysis was similar among groups (P = 0.51), although the average day when P4 was <1 ng/mL was later (P < 0.01) for ReBreed21 (20.5 ± 0.2) and ReBreed21+G (20.7 ± 0.2) compared to Control (19.2 ± 0.4). Overall ovulation at the end of the estrous cycle was increased (P = 0.03) for ReBreed21 groups (83.3%; 45/54) compared with Control (57.7%; 15/26). Synchronized ovulation on day 22-23 was greater (P < 0.01) for ReBreed21 (78.6%; 22/28) and ReBreed21+G (76.9%; 20/26) compared with Control (30.8%; 8/26). Thus, the ReBreed21 resynchronization program produced acceptable endocrine and follicular dynamics, including synchronized ovulation at the end of the protocol in nonpregnant heifers providing good rationale for testing the fertility and practical implementation of this protocol under field conditions.


Subject(s)
Buserelin , Estrus Synchronization , Animals , Cattle , Female , Buserelin/pharmacology , Estradiol/pharmacology , Estrus Synchronization/methods , Gonadotropins, Equine/pharmacology , Horses , Insemination, Artificial/veterinary , Insemination, Artificial/methods , Ovarian Follicle , Ovary , Ovulation , Progesterone/pharmacology
18.
Theriogenology ; 226: 343-349, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38964033

ABSTRACT

Two experiments evaluated the effect of different hormonal treatments to synchronize follicle wave emergence on follicle dynamics and pregnancies per AI (P/AI) in estradiol (E2)/progesterone (P4) timed-AI (TAI) protocols in lactating dairy cows. In Experiment 1, lactating, primiparous Holstein cows (n = 36) received a P4 releasing device (Day 0) and were allocated at random to one of the following three treatment groups: Group EB received 2 mg E2 benzoate (EB) intramuscularly (i.m.), Group EB + GnRH received 2 mg EB+20 µg buserelin (GnRH) i.m., or Group EB + P4 received 2 mg EB + 100 mg of injectable P4 (iP4) in oil i.m. All cows received 0.150 mg D-Cloprostenol on Days 7 and 8 followed by P4 device removal, 400 IU eCG and 1 mg ECP on Day 8. Daily ultrasound examinations revealed that although the interval from P4 device removal to ovulation was not affected by treatment, cows that received EB + GnRH had an earlier (P < 0.05) emergence of the new follicular wave (Day 2.6 ± 0.2) than the other two treatment groups (Days 3.5 ± 0.3 and 6.1 ± 0.3, for EB and EB + P4, respectively). In Experiment 2, 808 lactating cows were assigned randomly to the three treatments evaluated in Experiment 1, and all the cows were TAI to determine P/AI. Cows in the EB + GnRH group had greater P/AI (57.4 %, P < 0.01) than those in the EB (44.6 %) or EB + P4 (45.7 %) groups. In conclusion, the administration of GnRH, but not iP4, on the day of insertion of a P4 device improves P/AI in lactating dairy cows synchronized for TAI with an estradiol/P4-based protocol.


Subject(s)
Estradiol , Estrus Synchronization , Gonadotropin-Releasing Hormone , Insemination, Artificial , Lactation , Ovarian Follicle , Progesterone , Animals , Cattle/physiology , Female , Insemination, Artificial/veterinary , Insemination, Artificial/methods , Lactation/drug effects , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Progesterone/administration & dosage , Progesterone/pharmacology , Estradiol/pharmacology , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estrus Synchronization/methods , Pregnancy , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/administration & dosage , Buserelin/pharmacology , Buserelin/administration & dosage
19.
Cell Tissue Res ; 351(3): 521-34, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23254679

ABSTRACT

Gonadotropin-releasing hormone (GnRH) analogs are given to women undergoing in vitro fertilization. Case reports describing the development of chronic intestinal pseudo-obstruction and auto-antibodies against GnRH after such treatment suggest a strong association between intestinal dysfunction and GnRH analogs. No experimental model for studying such a relationship is currently at hand. Our main goal was to investigate possible enteric neurodegeneration and titers of GnRH antibodies in response to repeated administration of the GnRH analog buserelin in rat. Rats were treated for 1-4 sessions with daily subcutaneous injections of buserelin or saline for 5 days, followed by 3 weeks of recovery. Buserelin treatment caused significant loss of submucous and myenteric neurons in the fundus, ileum, and colon. The loss of enteric neurons can, at least partly, be explained by increased apoptosis. No GnRH- or GnRH-receptor-immunoreactive (IR) enteric neurons but numerous luteinizing hormone (LH)-receptor-IR neurons were detected. After buserelin treatment, the relative number of enteric LH-receptor-IR neurons decreased, whereas that of nitric-oxide-synthase-IR neurons increased. No intestinal inflammation or increased levels of circulating interleukins/cytokines were noted in response to buserelin treatment. Serum GnRH antibody titers were undetectable or extremely low in all rats. Thus, repeated administrations of buserelin induce neurodegeneration in rat gastrointestinal tract, possibly by way of LH-receptor hyperactivation. The present findings suggest that enteric neurodegenerative effects of GnRH analog treatment in man can be mimicked in rat. However, in contrast to man, no production of GnRH auto-antibodies has been noted in rat.


Subject(s)
Buserelin/pharmacology , Gastrointestinal Tract/cytology , Gonadotropin-Releasing Hormone/analogs & derivatives , Neurons/metabolism , Animals , Apoptosis/drug effects , Body Weight/drug effects , Cell Count , Cell Survival/drug effects , Colon/cytology , Female , Immunohistochemistry , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Neurons/cytology , Neurons/drug effects , Nitric Oxide Synthase Type I/metabolism , Rats , Rats, Sprague-Dawley , Receptors, LH/metabolism , Receptors, LHRH/metabolism , Vasoactive Intestinal Peptide/metabolism
20.
Hum Reprod ; 28(9): 2522-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23633553

ABSTRACT

STUDY QUESTION: Is severe early ovarian hyperstimulation syndrome (OHSS) completely prevented with the GnRH agonist trigger and 1500 IU hCG luteal rescue protocol? SUMMARY ANSWER: Severe early OHSS can occur even after the GnRH agonist trigger and 1500 IU hCG luteal rescue protocol. WHAT IS KNOWN ALREADY: Prior studies including over 200 women who received the GnRH agonist trigger and 1500 hCG luteal rescue protocol have reported complete prevention of severe early OHSS. Only a few late OHSS cases have been reported and it has been suggested that this protocol can be safely applied to any women under risk. STUDY DESIGN, SIZE, DURATION: This retrospective cohort study included all women who were at high risk of OHSS and were given the GnRH agonist trigger plus hCG luteal rescue protocol between December 2008 and August 2012 in the two participating centers. PARTICIPANTS/MATERIALS, SETTING, METHODS: There were 23 women with a mean estradiol level of 4891 ± 2214 pg/ml and a mean number of >12 mm follicles of 20 ± 6 on the day of ovulation triggering. OHSS was categorized according to the Golan criteria. MAIN RESULTS AND THE ROLE OF CHANCE: Overall 6 of the 23 (26%) women developed severe OHSS. Five women had severe early OHSS requiring ascites drainage and hospitalization and three of these women did not undergo embryo transfer. The number of follicles measuring 10-14 mm on the day of triggering was significantly different between women who developed severe early OHSS and those who did not. LIMITATIONS, REASONS FOR CAUTION: The small number of women with severe early OHSS may have prevented identification of other significant risk factors. WIDER IMPLICATIONS OF THE FINDINGS: Although the GnRH agonist plus 1500 IU hCG luteal rescue protocol significantly decreases the risk of severe OHSS, this life threatening complication can still occur in high-risk patients. It would be prudent to avoid hCG luteal rescue and freeze all embryos for future transfer in such women particularly when there are ≥18 follicles with 10-14 mm diameters even with few larger follicles.


Subject(s)
Chorionic Gonadotropin/adverse effects , Corpus Luteum/drug effects , Fertility Agents, Female/adverse effects , Gonadotropin-Releasing Hormone/agonists , Ovarian Hyperstimulation Syndrome/epidemiology , Ovary/drug effects , Ovulation Induction/adverse effects , Adult , Buserelin/adverse effects , Buserelin/pharmacology , Chorionic Gonadotropin/administration & dosage , Chorionic Gonadotropin/pharmacology , Cohort Studies , Corpus Luteum/diagnostic imaging , Estradiol/blood , Female , Fertility Agents, Female/pharmacology , Fertilization in Vitro/adverse effects , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Humans , Infertility, Female/blood , Infertility, Female/diagnostic imaging , Infertility, Female/therapy , Ovarian Hyperstimulation Syndrome/physiopathology , Ovarian Hyperstimulation Syndrome/prevention & control , Ovary/diagnostic imaging , Quebec/epidemiology , Retrospective Studies , Risk Factors , Severity of Illness Index , Triptorelin Pamoate/adverse effects , Triptorelin Pamoate/pharmacology , Turkey/epidemiology , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL