Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Clin Endocrinol Metab ; 106(12): 3591-3604, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34260712

RESUMEN

CONTEXT: Human embryonic implantation is regulated by neuroendocrine hormones, ovarian steroids, growth factors, and cytokines. Sympathetic innervation of the uterus also may play a role. OBJECTIVE: We tested the hypothesis that cabergoline (Cb), an agonist of type 2 dopamine receptors (DRD2), could influence endometrial decidualization in vitro. METHODS: Immunohistochemistry confirmed the presence of catecholaminergic neurons in human uterine tissue. DRD2 mRNA and protein expression in endometrial tissue and cells were validated by quantitative RT-PCR, cDNA microarrays, RNA sequencing, and Western blotting. Isolated human endometrial stromal cells (ESC) were subjected to dose-response and time-course experiments in the absence or presence of decidualizing hormones (10 nM estradiol, 100 nM progesterone, and 0.5 mM dibutyryl cAMP). In some cases, interleukin (IL)-1ß (0.1 nM) was used as an inflammatory stimulus. Well-characterized in vitro biomarkers were quantified. RESULTS: DRD2 were maximally expressed in vivo in the mid-secretory phase of the cycle and upregulated in ESC in response to decidualizing hormones, as were classical (eg, prolactin) and emerging (eg, VEGF and connexin 43) differentiation biomarkers. Cabergoline treatment more than doubled decidual biomarker expression, whereas risperidone, a dopamine receptor antagonist, inhibited ESC differentiation by >50%. Cabergoline induced characteristic decidual morphology changes and blocked detrimental effects of IL-1ß on decidual cytology. CONCLUSION: Our results support the hypothesis that dopaminergic neurons modulate decidualization in situ. We postulate that dopamine agonists, like Cb, could be developed as therapeutic agents to enhance implantation in couples with inflammation-associated infertility.


Asunto(s)
Cabergolina/farmacología , Diferenciación Celular , Decidua/citología , Agonistas de Dopamina/farmacología , Endometrio/citología , Interleucina-1beta/farmacología , Células del Estroma/citología , Células Cultivadas , Decidua/efectos de los fármacos , Decidua/metabolismo , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Femenino , Humanos , Técnicas In Vitro , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Transcriptoma
2.
Am J Obstet Gynecol ; 225(1): 43-50, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34215353

RESUMEN

Obstetrical complications, often referred to as the "great obstetrical syndromes," are among the most common global causes of mortality and morbidity in young women and their infants. However, treatments for these syndromes are underdeveloped compared with other fields of medicine and are urgently needed. This current paucity of treatments for obstetrical complications is a reflection of the challenges of drug development in pregnancy. The appetite of pharmaceutical companies to invest in research for obstetrical syndromes is generally reduced by concerns for maternal, fetal, and infant safety, poor definition, and high-risk regulatory paths toward product approval. Notably, drug candidates require large investments for development with an unguaranteed return on investment. Furthermore, the discovery of promising drug candidates is hampered by a poor understanding of the pathophysiology of obstetrical syndromes and their uniqueness to human pregnancies. This limits translational extrapolation and de-risking strategies in preclinical studies, as available for other medical areas, compounded with limited fetal safety monitoring to capture early prenatal adverse reactions. In addition, the ethical review committees are reluctant to approve the inclusion of pregnant women in trials, and in the absence of regulatory guidance in obstetrics, clinical development programs are subject to unpredictable regulatory paths. To develop effective and safe drugs for pregnancy complications, substantial commitment, and investment in research for innovative therapies are needed in parallel with the creation of an enabling ethical, legislative, and guidance framework. Solutions are proposed to enable stakeholders to work with a common set of expectations to facilitate progress in this medical discipline. Addressing this significant unmet need to advance maternal and possibly perinatal health requires the involvement of all stakeholders and specifically patients, couples, and clinicians facing pregnancy complications in the dearth of appropriate therapies. This paper focused on the key pharmaceutical research and development challenges to achieve effective and safe treatments for obstetrical syndromes.


Asunto(s)
Desarrollo de Medicamentos , Mortalidad Infantil , Mortalidad Materna , Obstetricia/métodos , Complicaciones del Embarazo/tratamiento farmacológico , Animales , Desarrollo de Medicamentos/ética , Desarrollo de Medicamentos/legislación & jurisprudencia , Desarrollo de Medicamentos/estadística & datos numéricos , Femenino , Feto/efectos de los fármacos , Humanos , Lactante , Recién Nacido , Intercambio Materno-Fetal , Investigación Farmacéutica/ética , Investigación Farmacéutica/legislación & jurisprudencia , Investigación Farmacéutica/estadística & datos numéricos , Embarazo
3.
Mutat Res ; 821: 111722, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32920458

RESUMEN

DNA damage is a common feature of human spermatozoa associated with an impaired capacity to fertilize the oocyte and an increased mutational load in the offspring. However, the etiology of this damage remains poorly defined. In this study we demonstrate that a major pathway for the induction of DNA damage in mammalian spermatozoa is triggered by exposure to exogenous cell free DNA (cfDNA). Exposure of human and mouse spermatozoa to cfDNA (calf thymus, mouse liver and salmon testes) in vitro induced a dose-dependent increase in sperm DNA damage that could be effectively suppressed by the concomitant presence of DNase. The induction of such damage was not accompanied by any concomitant change in sperm motility or vitality and was not directly associated with the induction of oxidative stress. In vivo the injection of exogenous DNA again precipitated an increase in sperm DNA fragmentation that could be reversed by the prior administration of DNase. Similarly, the induction of a transient unilateral testicular ischemia induced an increase in DNA fragmentation that was evident within 24 h and sustained for at least 14 days via mechanisms that could be completely suppressed by the prior administration of DNase. We conclude that exogenous cfDNA activates a defensive response in human spermatozoa associated with the nuclease-mediated induction of DNA fragmentation, possibly involving the participation of TLR9 and CD4. These novel insights have significant implications for our understanding of DNA fragmentation in the male germ line and open up new pathways for the remediation of this condition.


Asunto(s)
Ácidos Nucleicos Libres de Células/farmacología , Daño del ADN , Fertilidad/efectos de los fármacos , Estrés Oxidativo , Motilidad Espermática/efectos de los fármacos , Espermatozoides/patología , Animales , Femenino , Humanos , Masculino , Ratones , Espermatozoides/efectos de los fármacos
6.
Biol Reprod ; 97(3): 466-477, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-29025060

RESUMEN

Implantation is a complex event demanding contributions from both embryo and endometrium. Despite advances in assisted reproduction, endometrial receptivity defects persist as a barrier to successful implantation in women with infertility. We previously demonstrated that maternal haploinsufficiency for the endocrine peptide adrenomedullin (AM) in mice confers a subfertility phenotype characterized by defective uterine receptivity and sparse epithelial pinopode coverage. The strong link between AM and implantation suggested the compelling hypothesis that administration of AM prior to implantation may improve fertility, protect against pregnancy complications, and ultimately lead to better maternal and fetal outcomes. Here, we demonstrate that intrauterine delivery of AM prior to blastocyst transfer improves the embryo implantation rate and spacing within the uterus. We then use genetic decrease-of-function and pharmacologic gain-of-function mouse models to identify potential mechanisms by which AM confers enhanced implantation success. In epithelium, we find that AM accelerates the kinetics of pinopode formation and water transport and that, in stroma, AM promotes connexin 43 expression, gap junction communication, and barrier integrity of the primary decidual zone. Ultimately, our findings advance our understanding of the contributions of AM to uterine receptivity and suggest potential broad use for AM as therapy to encourage healthy embryo implantation, for example, in combination with in vitro fertilization.


Asunto(s)
Adrenomedulina/farmacología , Endometrio/citología , Endometrio/efectos de los fármacos , Fármacos para la Fertilidad Femenina/farmacología , Fertilidad/efectos de los fármacos , Uniones Intercelulares/efectos de los fármacos , Útero/citología , Útero/efectos de los fármacos , Animales , Comunicación Celular/efectos de los fármacos , Conexina 43/biosíntesis , Decidua/citología , Decidua/efectos de los fármacos , Implantación del Embrión/efectos de los fármacos , Transferencia de Embrión , Femenino , Uniones Comunicantes/efectos de los fármacos , Humanos , Ratones , Ratones Noqueados , Agua/metabolismo
7.
Endocrinology ; 155(8): 2953-65, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24823392

RESUMEN

RF-amide-related peptide-3 (RFRP-3), the mammalian ortholog of the avian gonadotropin-inhibiting hormone (GnIH), operates via the NPFF1 receptor (NPFF1R) to repress the reproductive axis, therefore acting as counterpart of the excitatory RF-amide peptide, kisspeptin (ligand of Gpr54). In addition, RFRP-3 modulates feeding and might contribute to the integrative control of energy homeostasis and reproduction. Yet, the experimental evidence supporting these putative functions is mostly indirect, and the physiological roles of RFRP-3 remain debatable and obscured by the lack of proper analytical tools and models. To circumvent these limitations, we characterize herein the first mouse line with constitutive inactivation of NPFF1R. Ablation of NPFF1R did not compromise fertility; rather, litters from NPFF1R null mice were larger than those from wild-type animals. Pubertal timing was not altered in NPFF1R deficient mice; yet, pre-pubertal knockout (KO) males displayed elevated LH levels, which normalized after puberty. Adult NPFF1R null male mice showed increased Kiss1 expression in the hypothalamic arcuate nucleus, higher serum FSH levels, and enhanced LH responses to GnRH. However, genetic elimination of NPFF1R was unable to reverse the state of hypogonadism caused by the lack of kisspeptin signaling, as revealed by double NPFF1R/Gpr54 KO mice. NPFF1R null mice displayed altered feedback responses to gonadal hormone withdrawal. In addition, metabolic challenges causing gonadotropin suppression, such as short-term fasting and high-fat diet, were less effective in dampening LH secretion in NPFF1R-deficient male mice, suggesting that absence of this inhibitory pathway partially prevented gonadotropin suppression by metabolic stress. Our data are the first to document the impact of elimination of GnIH signaling on reproductive parameters and their modulation by metabolic challenges. Whereas, in keeping with its inhibitory role, the NPFF1R pathway seems dispensable for preserved puberty and fertility, our results surface different alterations due to the lack of GnIH signaling that prominently include changes in the sensitivity to fasting- and obesity-associated hypogonadotropism.


Asunto(s)
Gonadotropinas/fisiología , Tamaño de la Camada , Neuropéptidos/fisiología , Receptores de Neuropéptido/fisiología , Maduración Sexual , Animales , Ayuno , Retroalimentación Fisiológica , Femenino , Fertilidad , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/deficiencia , Neuropéptidos/genética , Fenotipo , Receptores de Neuropéptido/deficiencia , Receptores de Neuropéptido/genética , Maduración Sexual/genética , Estrés Fisiológico/genética
8.
PLoS One ; 7(11): e48385, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23152771

RESUMEN

Glucocorticoids (GCs) such as prednisolone are potent immunosuppressive drugs but suffer from severe adverse effects, including the induction of insulin resistance. Therefore, development of so-called Selective Glucocorticoid Receptor Modulators (SGRM) is highly desirable. Here we describe a non-steroidal Glucocorticoid Receptor (GR)-selective compound (Org 214007-0) with a binding affinity to GR similar to that of prednisolone. Structural modelling of the GR-Org 214007-0 binding site shows disturbance of the loop between helix 11 and helix 12 of GR, confirmed by partial recruitment of the TIF2-3 peptide. Using various cell lines and primary human cells, we show here that Org 214007-0 acts as a partial GC agonist, since it repressed inflammatory genes and was less effective in induction of metabolic genes. More importantly, in vivo studies in mice indicated that Org 214007-0 retained full efficacy in acute inflammation models as well as in a chronic collagen-induced arthritis (CIA) model. Gene expression profiling of muscle tissue derived from arthritic mice showed a partial activity of Org 214007-0 at an equi-efficacious dosage of prednisolone, with an increased ratio in repression versus induction of genes. Finally, in mice Org 214007-0 did not induce elevated fasting glucose nor the shift in glucose/glycogen balance in the liver seen with an equi-efficacious dose of prednisolone. All together, our data demonstrate that Org 214007-0 is a novel SGRMs with an improved therapeutic index compared to prednisolone. This class of SGRMs can contribute to effective anti-inflammatory therapy with a lower risk for metabolic side effects.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Dibenzazepinas/farmacología , Receptores de Glucocorticoides/agonistas , Tiadiazoles/farmacología , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/genética , Glucemia , Dibenzazepinas/uso terapéutico , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratones , Simulación del Acoplamiento Molecular , Prednisolona/farmacología , Prednisolona/uso terapéutico , Unión Proteica , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/metabolismo , Tiadiazoles/uso terapéutico
9.
J Biol Chem ; 287(24): 20333-43, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22535964

RESUMEN

We present here the x-ray structures of the progesterone receptor (PR) in complex with two mixed profile PR modulators whose functional activity results from two differing molecular mechanisms. The structure of Asoprisnil bound to the agonist state of PR demonstrates the contribution of the ligand to increasing stability of the agonist conformation of helix-12 via a specific hydrogen-bond network including Glu(723). This interaction is absent when the full antagonist, RU486, binds to PR. Combined with a previously reported structure of Asoprisnil bound to the antagonist state of the receptor, this structure extends our understanding of the complex molecular interactions underlying the mixed agonist/antagonist profile of the compound. In addition, we present the structure of PR in its agonist conformation bound to the mixed profile compound Org3H whose reduced antagonistic activity and increased agonistic activity compared with reference antagonists is due to an induced fit around Trp(755), resulting in a decreased steric clash with Met(909) but inducing a new internal clash with Val(912) in helix-12. This structure also explains the previously published observation that 16α attachments to RU486 analogs induce mixed profiles by altering the binding of 11ß substituents. Together these structures further our understanding of the steric and electrostatic factors that contribute to the function of steroid receptor modulators, providing valuable insight for future compound design.


Asunto(s)
Estrenos/química , Mifepristona/química , Oximas/química , Receptores de Progesterona/agonistas , Receptores de Progesterona/química , Cristalografía por Rayos X , Humanos , Ligandos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
10.
J Med Primatol ; 41(1): 18-23, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22084982

RESUMEN

BACKGROUND: In women, different events of folliculogenesis can be measured and evaluated using ultrasound (US) technology. The availability of a non-invasive translational non-human primate model to study these processes would represent a major contribution to further advance R&D efforts toward novel therapies in assisted reproduction. METHODS: In our study, follicular growth and ovulation was measured in six cyclic Cynomolgus monkeys using abdominal Doppler US. RESULTS: The mean follicular diameter on cycle day -6 (cycle day 0=day of ovulation) was 3.7mm that increased to 6.8mm on cycle day -1. After ovulation, the mean diameter decreased to 4.6mm, confirming ovulation. The mean percentage of follicular size reduction after ovulation was 31%. CONCLUSION: Ultrasonography in combination with color-flow Doppler imaging was shown to be a useful, non-invasive translational method to measure ovarian follicular growth and occurrence and timing of follicular rupture in Cynomolgus monkeys.


Asunto(s)
Macaca fascicularis/fisiología , Folículo Ovárico/fisiología , Ovulación/fisiología , Animales , Estradiol/sangre , Femenino , Macaca fascicularis/sangre , Modelos Animales , Folículo Ovárico/diagnóstico por imagen , Progesterona/sangre , Ultrasonografía Doppler en Color
11.
Dev Biol ; 363(1): 40-51, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22206759

RESUMEN

Molecular mechanisms by which fertilization competent acrosome-reacted sperm bind to the oolemma remain uncharacterized. To identify oolemmal binding partner(s) for sperm acrosomal ligands, affinity panning was performed with mouse oocyte lysates using sperm acrosomal protein, SLLP1 as a target. An oocyte specific membrane metalloproteinase, SAS1B (Sperm Acrosomal SLLP1 Binding), was identified as a SLLP1 binding partner. cDNA cloning revealed six SAS1B splice variants, each containing a zinc binding active site and a putative transmembrane domain, with signal peptides in three variants. SAS1B transcripts were ovary specific. SAS1B protein was first detected in early secondary follicles in day 3 ovaries. Immunofluorescence localized SAS1B to the microvillar oolemma of M2 oocytes. After fertilization, SAS1B decreased on the oolemma and became virtually undetectable in blastocysts. In transfected CHO-K1 cells SAS1B localized to the surface of unpermeabilized cells. Recombinant and native SLLP1 co-localized with SAS1B to the microvillar domain of ovulated M2 oocytes. Molecular interactions between mouse SLLP1 and SAS1B were demonstrated by surface plasmon resonance, far-western, yeast two-hybrid, recombinant- and native-co-IP analyses. SAS1B bound to SLLP1 with high affinity. SAS1B had protease activity, and SAS1B protein or antibody significantly inhibited fertilization. SAS1B knockout female mice showed a 34% reduction in fertility. The study identified SAS1B-SLLP1 as a pair of novel sperm-egg binding partners involving the oolemma and intra-acrosomal compartment during fertilization.


Asunto(s)
Fertilización , Isoantígenos/metabolismo , Metaloproteasas/metabolismo , Oocitos/metabolismo , Proteínas de Plasma Seminal/metabolismo , Acrosoma/metabolismo , Empalme Alternativo , Animales , Unión Competitiva , Far-Western Blotting , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Femenino , Inmunoprecipitación , Isoantígenos/genética , Masculino , Metaloproteasas/genética , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Embarazo , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas de Plasma Seminal/genética , Interacciones Espermatozoide-Óvulo , Resonancia por Plasmón de Superficie , Técnicas del Sistema de Dos Híbridos
12.
Am J Respir Crit Care Med ; 178(6): 565-73, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18583571

RESUMEN

RATIONALE: In a guinea pig model of allergic asthma, using perfused tracheal preparations ex vivo, we demonstrated that L-arginine limitation due to increased arginase activity underlies a deficiency of bronchodilating nitric oxide (NO) and airway hyperresponsiveness (AHR) after the allergen-induced early and late asthmatic reaction. OBJECTIVES: Using the same animal model, we investigated the acute effects of the specific arginase inhibitor 2(S)-amino-6-boronohexanoic acid (ABH) and of L-arginine on AHR after the early and late reaction in vivo. In addition, we investigated the protection of allergen-induced asthmatic reactions, AHR, and airway inflammation by pretreatment with the drug. METHODS: Airway responsiveness to inhaled histamine was measured in permanently instrumented, freely moving guinea pigs sensitized to ovalbumin at 24 hours before allergen challenge and after the allergen-induced early and late asthmatic reactions by assessing histamine PC(100) (provocative concentration causing a 100% increase of pleural pressure) values. MEASUREMENTS AND MAIN RESULTS: Inhaled ABH acutely reversed AHR to histamine after the early reaction from 4.77 +/- 0.56-fold to 2.04 +/- 0.34-fold (P < 0.001), and a tendency to inhibition was observed after the late reaction (from 1.95 +/- 0.56-fold to 1.56 +/- 0.47-fold, P < 0.10). Quantitatively similar results were obtained with inhaled l-arginine. Remarkably, after pretreatment with ABH a 33-fold higher dose of allergen was needed to induce airway obstruction (P < 0.01). Consequently, ABH inhalation 0.5 hour before and 8 hours after allergen challenge protected against the allergen-induced early and late asthmatic reactions, AHR and inflammatory cell infiltration. CONCLUSIONS: Inhalation of ABH or l-arginine acutely reverses allergen-induced AHR after the early and late asthmatic reaction, presumably by attenuating arginase-induced substrate deficiency to NO synthase in the airways. Moreover, ABH considerably reduces the airway sensitivity to inhaled allergen and protects against allergen-induced bronchial obstructive reactions, AHR, and airway inflammation. This is the first in vivo study indicating that arginase inhibitors may have therapeutic potential in allergic asthma.


Asunto(s)
Obstrucción de las Vías Aéreas/prevención & control , Arginasa/antagonistas & inhibidores , Asma/fisiopatología , Hiperreactividad Bronquial/fisiopatología , Obstrucción de las Vías Aéreas/inmunología , Aminocaproatos/farmacología , Animales , Arginasa/metabolismo , Asma/tratamiento farmacológico , Compuestos de Boro/farmacología , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/metabolismo , Pruebas de Provocación Bronquial , Lavado Broncoalveolar , Cobayas , Cloruro de Metacolina/farmacología , Modelos Animales , Óxido Nítrico
13.
Mol Endocrinol ; 21(3): 726-39, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17192404

RESUMEN

The Aurora kinases are cell cycle-regulatory serine-threonine kinases that have been implicated in the function of the centrosomes, kinetechores, chromosome dynamics, and cytokinesis. In comparison with other tissues, there are high levels of expression of Aurora-B and -C in testis. What their respective roles in mammalian spermatogenesis are is an open question. Here we describe the expression and distribution patterns of the three kinases in mouse testis using in situ hybridization and immunohistochemistry. Importantly, the localization of Aurora-B is tightly regulated during spermatogenesis, whereas Aurora-C expression appears to be testis specific. To address the function of Aurora-B in spermatogenesis, we have generated transgenic mice using a pachytene-stage-specific promoter driving the expression of either wild-type Aurora-B or an inactive form of the kinase. Expression of the inactive Aurora-B results in abnormal spermatocytes, increased apoptosis, spermatogenic arrest, and subfertility defects. The function of Aurora-C may also be targeted in the Aurora-B transgenic mutants. To address the function of Aurora-C in testis, we generated Aurora-C knockout mice by homologous recombination. Remarkably, Aurora-C null mice were viable, yet the males had compromised fertility. Aurora-C mutant sperm display abnormalities that included heterogenous chromatin condensation, loose acrosomes, and blunted heads. These findings indicate that Aurora-B and Aurora-C serve specialized functions in mammalian spermatogenesis.


Asunto(s)
Proteínas Serina-Treonina Quinasas/fisiología , Espermatogénesis , Animales , Aurora Quinasa B , Aurora Quinasa C , Aurora Quinasas , Fertilidad/genética , Infertilidad Masculina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/genética , Recombinación Genética , Espermatogénesis/genética , Espermatozoides/citología , Testículo/citología
14.
Dev Biol ; 296(2): 353-62, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16842770

RESUMEN

We previously demonstrated that male mice deficient in the soluble adenylyl cyclase (sAC) are sterile and produce spermatozoa with deficits in progressive motility and are unable to fertilize zona-intact eggs. Here, analyses of sAC(-/-) spermatozoa provide additional insights into the functions linked to cAMP signaling. Adenylyl cyclase activity and cAMP content are greatly diminished in crude preparations of sAC(-/-) spermatozoa and are undetectable after sperm purification. HCO(3)(-) is unable to rapidly accelerate the flagellar beat or facilitate evoked Ca(2+) entry into sAC(-/-) spermatozoa. Moreover, the delayed HCO(3)(-)-dependent increases in protein tyrosine phosphorylation and hyperactivated motility, which occur late in capacitation of wild-type spermatozoa, do not develop in sAC(-/-) spermatozoa. However, sAC(-/-) sperm fertilize zona-free oocytes, indicating that gamete fusion does not require sAC. Although ATP levels are significantly reduced in sAC(-/-) sperm, cAMP-AM ester increases flagellar beat frequency, progressive motility, and alters the pattern of tyrosine phosphorylated proteins. These results indicate that sAC and cAMP coordinate cellular energy balance in wild-type sperm and that the ATP generating machinery is not operating normally in sAC(-/-) spermatozoa. These findings demonstrate that sAC plays a critical role in cAMP signaling in spermatozoa and that defective cAMP production prevents engagement of multiple components of capacitation resulting in male infertility.


Asunto(s)
Adenilil Ciclasas/fisiología , Fertilización/fisiología , Espermatozoides/fisiología , Adenilil Ciclasas/deficiencia , Adenilil Ciclasas/genética , Animales , AMP Cíclico/metabolismo , Femenino , Fertilización/genética , Infertilidad Masculina/enzimología , Infertilidad Masculina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Transducción de Señal/fisiología , Solubilidad , Capacitación Espermática/genética , Espermatozoides/enzimología
15.
Proc Natl Acad Sci U S A ; 101(9): 2993-8, 2004 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-14976244

RESUMEN

To acquire the ability to fertilize, spermatozoa undergo complex, but at present poorly understood, activation processes. The intracellular rise of cAMP produced by the bicarbonate-dependent soluble adenylyl cyclase (sAC) has been suggested to play a central role in initiating the cascade of the events that culminates in spermatozoa maturation. Here, we show that targeted disruption of the sAC gene does not affect spermatogenesis but dramatically impairs sperm motility, leading to male sterility. sAC mutant spermatozoa are characterized by a total loss of forward motility and are unable to fertilize oocytes in vitro. Interestingly, motility in sAC mutant spermatozoa can be restored on cAMP loading, indicating that the motility defect observed is not caused by a structural defect. We, therefore, conclude that sAC plays an essential and nonredundant role in the activation of the signaling cascade controlling motility and, therefore, in fertility. The crucial role of sAC in fertility and the absence of any other obvious pathological abnormalities in sAC-deficient mice may provide a rationale for developing inhibitors that can be applied as a human male contraceptive.


Asunto(s)
Adenilil Ciclasas/deficiencia , Adenilil Ciclasas/genética , Infertilidad Masculina/genética , Motilidad Espermática/genética , Animales , AMP Cíclico/metabolismo , Cartilla de ADN , Femenino , Fertilización/genética , Fertilización In Vitro , Genes Reporteros , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Oocitos/fisiología , Reacción en Cadena de la Polimerasa , Espermatozoides/enzimología , Testículo/enzimología , beta-Galactosidasa/genética
16.
Mol Cell Biol ; 24(2): 687-96, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14701741

RESUMEN

LGR7 is a G-protein coupled receptor with structural homology to the gonadotrophin and thyrotrophin receptors. Recently, LGR7 was deorphanized, and it was shown that relaxin is the ligand for LGR7. To further study the function of this receptor, mice deficient for LGR7 were generated by replacing part of the transmembrane-encoding region with a LacZ reporter cassette. Here we show that LGR7 is expressed in various tissues, including the uterus, heart, brain, and testis. Fertility studies using female LGR7-/- mice showed normal fertility and litter size. However, some females were incapable of delivering their pups, and several pups were found dead. Moreover, all offspring died within 24 to 48 h after delivery because female LGR7-/- mice were unable to feed their offspring due to impaired nipple development. In some male LGR7-/- mice, spermatogenesis was impaired, leading to azoospermia and a reduction in fertility. Interestingly, these phenomena were absent in mutant mice at older ages or in later generations. Taken together, results from LGR7 knockout mice indicate an essential role for the LGR7 receptor in nipple development during pregnancy. Moreover, a defect in parturition was observed, suggesting a role for LGR7 in the process of cervical ripening.


Asunto(s)
Pezones/anomalías , Parto/fisiología , Receptores de Superficie Celular/deficiencia , Animales , Animales Recién Nacidos , Apoptosis/genética , Apoptosis/fisiología , Secuencia de Bases , Maduración Cervical/genética , Maduración Cervical/fisiología , ADN Complementario/genética , Femenino , Fertilidad/genética , Fertilidad/fisiología , Marcación de Gen , Operón Lac , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Parto/genética , Fenotipo , Embarazo , Regiones Promotoras Genéticas , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Receptores Acoplados a Proteínas G , Espermatogénesis/genética , Espermatogénesis/fisiología , Testículo/anomalías
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA