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










Publication year range
1.
Eur J Endocrinol ; 179(3): R143-R150, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29959220

ABSTRACT

The two pituitary gonadotrophins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), and in particular LH-stimulated high intratesticular testosterone (ITT) concentration, are considered crucial for spermatogenesis. We have revisited these concepts in genetically modified mice, one being the LH receptor (R)-knockout mouse (LuRKO), the other a transgenic mouse expressing in Sertoli cells a highly constitutively active mutated Fshr (Fshr-CAM). It was found that full spermatogenesis was induced by exogenous testosterone treatment in LuRKO mice at doses that restored ITT concentration to a level corresponding to the normal circulating testosterone level in WT mice, ≈5 nmol/L, which is 1.4% of the normal high ITT concentration. When hypogonadal LuRKO and Fshr-CAM mice were crossed, the double-mutant mice with strong FSH signaling, but minimal testosterone production, showed near-normal spermatogenesis, even when their residual androgen action was blocked with the strong antiandrogen flutamide. In conclusion, our findings challenge two dogmas of the hormonal regulation of male fertility: (1) high ITT concentration is not necessary for spermatogenesis and (2) strong FSH stimulation can maintain spermatogenesis without testosterone. These findings have clinical relevance for the development of hormonal male contraception and for the treatment of idiopathic oligozoospermia.


Subject(s)
Follicle Stimulating Hormone/physiology , Luteinizing Hormone/physiology , Mutation , Spermatogenesis/physiology , Animals , Crosses, Genetic , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone, beta Subunit/genetics , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Receptors, FSH/genetics , Receptors, LH/deficiency , Receptors, LH/genetics , Sertoli Cells/metabolism , Spermatogenesis/drug effects , Spermatogenesis/genetics , Testis/chemistry , Testosterone/analysis , Testosterone/pharmacology , Testosterone/physiology
2.
Cell Physiol Biochem ; 43(3): 1064-1076, 2017.
Article in English | MEDLINE | ID: mdl-28977799

ABSTRACT

BACKGROUND/AIMS: Physiological role of luteinizing hormone (LH) and its receptor (LHCGR) in adrenal remains unknown. In inhibin-α/Simian Virus 40 T antigen (SV40Tag) (inhα/Tag) mice, gonadectomy-induced (OVX) elevated LH triggers the growth of transcription factor GATA4 (GATA4)-positive adrenocortical tumors in a hyperplasia-adenoma-adenocarcinoma sequence. METHODS: We investigated the role of LHCGR in tumor induction, by crossbreeding inhα/Tag with Lhcgr knockout (LuRKO) mice. By knocking out Lhcgr and Gata4 in Cα1 adrenocortical cells (Lhcgr-ko, Gata4-ko) we tested their role in tumor progression. RESULTS: Adrenal tumors of OVX inhα/Tag mice develop from the hyperplastic cells localized in the topmost layer of zona fasciculata. OVX inhα/Tag/LuRKO only developed SV40Tag positive hyperplastic cells that were GATA4 negative, cleaved caspase-3 positive and did not progress into adenoma. In contrast to Lhcgr-ko, Gata4-ko Cα1 cells presented decreased proliferation, increased apoptosis, decreased expression of Inha, SV40Tag and Lhcgr tumor markers, as well as up-regulated adrenal- and down-regulated sex steroid gene expression. Both Gata4-ko and Lhcgr-ko Cα1 cells had decreased expression of steroidogenic genes resulting in decreased basal progesterone production. CONCLUSION: Our data indicate that LH/LHCGR signaling is critical for the adrenal cell reprogramming by GATA4 induction prompting adenoma formation and gonadal-like phenotype of the adrenocortical tumors in inhα/Tag mice.


Subject(s)
Adrenal Cortex Neoplasms/pathology , GATA4 Transcription Factor/metabolism , Luteinizing Hormone/metabolism , Adrenal Cortex Neoplasms/etiology , Adrenal Cortex Neoplasms/metabolism , Adrenal Glands/metabolism , Adrenal Glands/pathology , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Apoptosis , CRISPR-Cas Systems/genetics , Caspase 3/metabolism , Cell Proliferation , Cell Transformation, Neoplastic , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Down-Regulation , Female , Fluoroimmunoassay , GATA4 Transcription Factor/deficiency , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gonads/surgery , Inhibins/genetics , Inhibins/metabolism , Luteinizing Hormone/blood , Mice , Mice, Knockout , Mice, Transgenic , Phenotype , Phosphoproteins/metabolism , Receptors, LH/deficiency , Receptors, LH/genetics , Steroidogenic Factor 1/metabolism
3.
Reprod Sci ; 22(8): 1000-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25670721

ABSTRACT

We investigated the possible presence of functional human chorionic gonadotropin (hCG)/luteinizing hormone (LH) receptors in ß-cells of pancreas, using a combination of techniques on hCG/LH receptor knockout mice, immortalized rat insulinoma cells, and human pancreatic islets. The results showed the presence of receptors and their activation resulted in a dose-dependent increase in glucose-induced release of insulin. These findings place hCG and LH among the regulators of insulin release with potential implications for insulin-level changes during the periods of altered hCG and LH secretion.


Subject(s)
Insulin-Secreting Cells/metabolism , Receptors, LH/metabolism , Animals , Cadaver , Cell Line, Tumor , Chorionic Gonadotropin/pharmacology , Dose-Response Relationship, Drug , Female , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Mice, Knockout , Rats , Receptors, LH/agonists , Receptors, LH/deficiency , Receptors, LH/genetics , Tissue Culture Techniques
4.
Ann Endocrinol (Paris) ; 71(3): 170-6, 2010 May.
Article in English | MEDLINE | ID: mdl-20362970

ABSTRACT

We have produced over the years several genetically modified mouse models (transgenic [TG], knockout [KO] and knockin [KI]) for the study of normal and aberrant functions of gonadotrophins and their receptors. We summarise in the present review some of our recent findings on these animal models. One is the cascade of extragonadal phenotypes triggered by ovarian hyperstimulation in TG mice overexpressing the human choriongonadotrophin (hCG) beta-subunit and presenting with elevated levels of serum luteinising hormone (LH)/hCG bioactivity. Massively elevated levels of serum progesterone, rather than oestrogens, are responsible for the induction of pituitary prolactinomas and the subsequently elevated prolactin (PRL) levels. Along with normal oestradiol and elevated progesterone levels, the increased concentration of PRL induces lobuloalveolar development of the mammary gland, with ultimate formation of oestrogen and progesterone receptor-negative malignant tumours. Another TG mouse model expressing a constitutively activating mutant form of the follicle-stimulating hormone receptor (FSHR) presents with a strong ovarian phenotype inducing advanced follicular development and depletion, haemorrhagic follicles, teratomas and infertility. A third TG mouse model, coexpressing binding- and signalling-deficient mutants of LHCGR in the KO background for the same receptor (R) gene provided convincing evidence that functional complementation through homo-di/oligomerisation is a physiologically relevant mode of activation of class A G protein-coupled receptors (GPCR). Taken together, genetically modified mouse models provide powerful tools for the elucidation of normal and pathological functions of gonadotrophins and their R.


Subject(s)
Gonadotropins/genetics , Receptors, Gonadotropin/genetics , Animals , Estradiol/physiology , Female , Follicle Stimulating Hormone/genetics , Gonadotropin-Releasing Hormone/genetics , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Ovarian Diseases/genetics , Receptors, FSH/genetics , Receptors, LH/deficiency , Receptors, LH/genetics
6.
Proc Natl Acad Sci U S A ; 107(5): 2319-24, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20080658

ABSTRACT

G protein-coupled receptors (GPCRs) are ubiquitous mediators of signaling of hormones, neurotransmitters, and sensing. The old dogma is that a one ligand/one receptor complex constitutes the functional unit of GPCR signaling. However, there is mounting evidence that some GPCRs form dimers or oligomers during their biosynthesis, activation, inactivation, and/or internalization. This evidence has been obtained exclusively from cell culture experiments, and proof for the physiological significance of GPCR di/oligomerization in vivo is still missing. Using the mouse luteinizing hormone receptor (LHR) as a model GPCR, we demonstrate that transgenic mice coexpressing binding-deficient and signaling-deficient forms of LHR can reestablish normal LH actions through intermolecular functional complementation of the mutant receptors in the absence of functional wild-type receptors. These results provide compelling in vivo evidence for the physiological relevance of intermolecular cooperation in GPCR signaling.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, LH/chemistry , Receptors, LH/metabolism , Animals , Cell Line , Chorionic Gonadotropin/metabolism , Genetic Complementation Test , Humans , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Models, Biological , Models, Molecular , Mutation , Phenotype , Protein Binding , Protein Multimerization , Receptors, LH/deficiency , Receptors, LH/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Testis/metabolism , Testis/pathology
7.
Horm Cancer ; 1(5): 256-64, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21666843

ABSTRACT

Previous studies have revealed a close link between luteinizing hormone (LH)/human chorionic gonadotropin (hCG) signaling and oncogenesis in gonadal and nongonadal tissues. To investigate whether genetic ablation of LH receptor (Lhr) affects the animal's oncogenic susceptibility, adult female wild-type (wt), heterozygous, and homozygous Lhr knockout (LhrKO) mice were intraperitoneally injected with an alkylating agent, N-methyl-N-nitrosourea (MNU, 50 mg/kg of body weight). The mice were sacrificed when they were short of breath or 10 months after the injection. The results showed that MNU induced non-Hodgkin's thymic and lymphonodus lymphomas in 70.6% and 100% of heterozygous and homozygous animals, respectively, compared with 35.7% in wt siblings. The tumor development was rapid; they were more aggressive and metastasized to the spleen, liver, and kidney in Lhr-deficient mice compared to wt siblings. All tumors were immunostained-positive for a T-cell specific marker, CD3, but not for a B-cell marker, CD22, suggesting that all the lymphomas arose from T-cells, which are known to be LH/hCG receptor-positive. There was no rearrangement of the Lhr gene locus or differences in thymic cell proliferation among the genotypes. However, apoptosis was lower in the Lhr-deficient thymuses. The thymic Bcl-2 levels were elevated and caspase-3 activation was reduced in Lhr heterozygous and homozygous animals. In conclusion, MNU induced a higher incidence and an earlier onset of aggressive lymphomas in LhrKO animals, which may be associated with a reduction in apoptosis of thymocytes.


Subject(s)
Lymphoma, T-Cell/metabolism , Receptors, LH/deficiency , Alkylating Agents/toxicity , Animals , Apoptosis/physiology , Blotting, Southern , Blotting, Western , Female , Immunohistochemistry , In Situ Nick-End Labeling , Lymphoma, T-Cell/chemically induced , Lymphoma, T-Cell/genetics , Methylnitrosourea/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LH/genetics , Reverse Transcriptase Polymerase Chain Reaction
8.
Nat Neurosci ; 10(8): 1003-11, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17603480

ABSTRACT

The regulation of female reproductive behaviors may involve memories of male pheromone signatures, formed in part by neural circuitry involving the olfactory bulb and hippocampus. These neural structures are the principal sites of adult neurogenesis; however, previous studies point to their independent regulation by sensory and physiological stimuli. Here we report that the pheromones of dominant (but not subordinate) males stimulate neuronal production in both the olfactory bulb and hippocampus of female mice, which are independently mediated by prolactin and luteinizing hormone, respectively. Neurogenesis induced by dominant-male pheromones correlates with a female preference for dominant males over subordinate males, whereas blocking neurogenesis with the mitotic inhibitor cytosine arabinoside eliminated this preference. These results suggest that male pheromones are involved in regulating neurogenesis in both the olfactory bulb and hippocampus, which may be important for female reproductive success.


Subject(s)
Brain/cytology , Cell Proliferation/drug effects , Neurons/drug effects , Sex Attractants/pharmacology , Sexual Behavior, Animal/drug effects , Animals , Astringents/toxicity , Behavior, Animal , Brain/drug effects , Bromodeoxyuridine/metabolism , Cytarabine/pharmacology , Female , Immunosuppressive Agents/pharmacology , In Situ Nick-End Labeling/methods , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Nerve Tissue Proteins/metabolism , Receptors, LH/deficiency , Receptors, Prolactin/deficiency , Sexual Behavior, Animal/physiology , Social Dominance , Zinc Sulfate/toxicity
9.
Mol Cell Endocrinol ; 252(1-2): 126-35, 2006 Jun 27.
Article in English | MEDLINE | ID: mdl-16675102

ABSTRACT

Numerous genetically modified mouse models have recently been developed for the study of the pituitary-gonadal interactions. They include spontaneous or engineered knockouts (KO) of the gonadotrophin-releasing hormone (GnRH) and its receptor, the gonadotrophin common-alpha(Calpha), luteinising hormone (LH) beta and follicle-stimulating hormone (FSH) beta subunits, and the two gonadotrophin receptors (R), LHR and FSHR. In addition, there are also transgenic (TG) mice overexpressing gonadotrophin subunits and producing supraphysiological levels of these hormones. These models have offered relevant phenocopies for similar mutations in humans and to a great extent expanded our knowledge on normal and pathological functions of the hypothalamic-pituitary-gonadal (HPG) axis. The purpose of this article is to review some of our recent findings on two such mouse models, the LHR KO mouse (LuRKO), and the hCG overexpressing TG mouse (hCG+).


Subject(s)
Animals, Genetically Modified , Luteinizing Hormone/physiology , Mice/genetics , Animals , Female , Male , Mice, Knockout , Mice, Transgenic , Ovary/physiology , Receptors, LH/deficiency , Receptors, LH/genetics , Testis/physiology
10.
Mol Cell Endocrinol ; 254-255: 84-90, 2006 Jul 25.
Article in English | MEDLINE | ID: mdl-16730882

ABSTRACT

During the last 10 years, numerous activating and inactivating mutations have been detected in the genes encoding the two gonadotrophins, luteinising hormone (LH) and follicle-stimulating hormone (FSH), as well as their cognate receptors (R), LHR and FSHR. Because activation of the hypothalamic-pituitary-gonadal axis is a crucial event in the onset and progression of puberty, mutations affecting gonadotrophin action have major influence on this developmental process. Many of the phenotypic effects observed have been expected on the basis of the existing information about gonadotrophin action (e.g. delayed puberty), but also many unexpected findings have been made, including the lack of phenotype in women with activating LHR mutations, and the discrepancy in phenotypes of men with inactivating mutations of FSHbeta (azoospermia and infertility) and FSHR (oligozoospermia and subfertility). Some of the possible mutations, such as inactivating LHbeta and activating FSHR mutations in women, have not yet been detected. Genetically modified mice provide relevant phenocopies for the human mutations and serve as good models for studies on molecular pathogenesis of these conditions. They may also predict phenotypes of the mutations that have not yet been detected in humans. We review here briefly the effects of gonadotrophin subunit and receptor mutations on puberty in humans and contrast the information with findings on genetically modified mice with similar mutations.


Subject(s)
Gonadal Disorders/genetics , Mutation , Pituitary Diseases/genetics , Sexual Maturation/genetics , Animals , Follicle Stimulating Hormone/deficiency , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/physiology , Gonadotropins/deficiency , Gonadotropins/genetics , Gonadotropins/physiology , Humans , Luteinizing Hormone/deficiency , Luteinizing Hormone/genetics , Luteinizing Hormone/physiology , Mice , Mice, Knockout , Mice, Transgenic , Pituitary Gland , Protein Subunits/deficiency , Protein Subunits/genetics , Puberty/genetics , Puberty/physiology , Receptors, FSH/deficiency , Receptors, FSH/genetics , Receptors, FSH/physiology , Receptors, Gonadotropin/deficiency , Receptors, Gonadotropin/genetics , Receptors, Gonadotropin/physiology , Receptors, LH/deficiency , Receptors, LH/genetics , Receptors, LH/physiology , Transfection
11.
J Clin Invest ; 115(7): 1862-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15951841

ABSTRACT

The luteinizing hormone receptor (LHR), mainly expressed in gonads, is essential for normal reproduction. However, numerous recent studies have also demonstrated LHR expression in multiple extragonadal reproductive and nonreproductive tissues. Although some effects of luteinizing hormone (LH) or its agonist, human chorionic gonadotropin, have been shown in extragonadal sites, their physiological significance remains open. In the present study, we have addressed the function of the extragonadal LHR using LHR-KO mice (LuRKO mice), in which the ovaries of prepubertal mice were orthotopically replaced with pieces of WT ovary using similarly transplanted WT mice as controls. Most ovarian transplants attained normal endocrine function in both groups of mice, as demonstrated by normal age at vaginal opening, estrous cycles, and sexual behavior. Both the LuRKO and WT mice repeatedly became pregnant (9/16 vs. 16/20 after first mating; difference not significant) and delivered similarly sized litters, which grew normally after birth, indicating normal lactation. In conclusion, fertility is restored in LuRKO mice by transplantation of WT ovarian tissue. This is achieved in the absence of extragonadal LHR expression, which indicates physiological redundancy for such receptor sites.


Subject(s)
Fertility/physiology , Luteinizing Hormone/physiology , Ovary/transplantation , Receptors, LH/deficiency , Animals , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Ovary/physiology , Pregnancy , Receptors, LH/genetics , Receptors, LH/physiology
12.
Mol Endocrinol ; 19(10): 2591-602, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15941853

ABSTRACT

It is considered a dogma that a secretory peak of LH is indispensable as the trigger of ovulation. However, earlier studies on hypophysectomized rodents have shown that stimulation with recombinant FSH, devoid of any LH activity, is able to boost the final stages of follicular maturation and trigger ovulation. As the expression of ovarian LH receptors (LHRs) still persists after hypophysectomy, such studies cannot totally exclude the possibility that LHR activation is involved in the apparently pure FSH effects. To revisit this question, we analyzed in LHR knockout (LuRKO) mice the progression of folliculogenesis and induction of ovulation by human chorionic gonadotropin and human recombinant FSH treatments. The results provide clear evidence that follicular development and ovulation could not be induced by high doses of FSH in the absence of LHR expression. Ovarian histology and oocyte analyses indicated that follicular maturation did not advance in LuRKO mice beyond the antral follicle stage. Neither were ovulations detected in LuRKO ovaries after any of the gonadotropin treatments. The ovarian resistance to FSH treatment in the absence of LHR was confirmed by real-time RT-PCR and immunohistochemical analyses of a number of gonadotropin-dependent genes, which only responded to the treatments in wild-type control mice. Negative findings were not altered by estradiol priming preceding the gonadotropin stimulations. Hence, the present study shows that, in addition to ovulation, the expression of LHR is essential for follicular maturation in the progression from antral to preovulatory stage.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Ovarian Follicle/drug effects , Ovulation/drug effects , Receptors, LH/deficiency , Animals , Base Sequence , Biomarkers/metabolism , Female , Follicular Phase/drug effects , Follicular Phase/physiology , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Mice , Mice, Knockout , Ovarian Follicle/physiology , Ovulation/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LH/genetics , Receptors, LH/physiology , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
13.
Biol Reprod ; 71(5): 1605-13, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15253923

ABSTRACT

The LH receptor knockout model, developed in our laboratory, was used in determining what FSH alone can do in the absence of LH signaling and whether any of the testicular LH actions are not mediated by androgens. The results revealed that null animals contained smaller seminiferous tubules, which contained the same number of Sertoli cells, spermatogonia, and early spermatocytes as wild-type siblings. The number of late spermatocytes, on the other hand, was moderately decreased, the number of round spermatids was dramatically decreased, and elongated spermatids were completely absent. These changes appear to be due to an increase in apoptosis in spermatocytes. While the number of Leydig cells progressively increased from birth to 60 days of age in wild-type animals, they remained unchanged in null animals. Consequently, 60-day-old null animals contained only a few Leydig cells of fetal type. The age-dependent increase in testicular macrophages lagged behind in null animals compared with wild-type siblings. Orchidopexy indicated that -/- testicular phenotype was not due to abdominal location. Rather, it was mostly due to androgen deficiency, as 21-day testosterone replacement therapy stimulated the growth of seminiferous tubules, decreased apoptosis, and increased the number of late spermatocytes and round spermatids and their subsequent differentiation into mature sperm. The therapy, however, failed to restore adult-type Leydig cells and testicular macrophage numbers to the wild-type levels. In summary, our data support the concept that FSH signaling alone can maintain the proliferation and development of Sertoli cells, spermatogonia, and early spermatocytes. LH actions mediated by testosterone are required for completion of spermatogenesis, and finally, androgen-independent actions of LH are required for the formation of adult-type Leydig cells and recruitment of macrophages into the testes.


Subject(s)
Androgens/pharmacology , Follicle Stimulating Hormone/physiology , Luteinizing Hormone/physiology , Receptors, LH/deficiency , Testis/cytology , Testis/physiology , Testosterone/pharmacology , 11-beta-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Apoptosis , Body Temperature/physiology , Cell Shape , Female , Male , Mice , Mice, Knockout , Phenotype , Seminiferous Tubules/cytology , Seminiferous Tubules/drug effects , Seminiferous Tubules/growth & development , Sperm Count , Spermatids/cytology , Spermatocytes/cytology , Spermatogenesis/drug effects , Spermatogenesis/physiology , Testis/enzymology
14.
Endocrinology ; 144(8): 3555-64, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12865338

ABSTRACT

Considerable attention has been paid to the role of sex steroids during periods of major skeletal turnover, but the interaction of the gonadotropic hormones, which include LH, FSH, and human chorionic gonadotropin (hCG), within bone tissue have been overlooked. The question is pertinent due to the recent detection of extragonadal expression of gonadotropin receptors. Western blotting, immunolocalization, and RT-PCR supported the presence of osteoblast LH receptors. However, osteoblast cells failed to bind [(125)I]hCG and treatment with hCG failed to generate either cAMP or phosphorylated ERK 1/2. Bone mineral density (BMD) and bone histomorphometry were examined in the following models: 1) LH receptor null mutant (LuRKO) mice; 2) transgenic mice overexpressing hCG (hCG alphabeta+); and 3) ovariectomized (OVX) hCG alphabeta+ model. Male LuRKO mice showed a decrease in BMD after 5 months, apparently secondary to suppressed gonadal steroid production. Similarly, 9- to 10-wk-old female LuRKO mice exhibited decreases in histomorphometric parameters tested. The data indicate that loss of LH signaling results in a reduction in bone formation or an increase in bone resorption. By contrast, there were significant increases in BMD and histomorphometric indices for female, but not male, hCG alphabeta+ mice, indicating that chronic exposure to hCG results in bone formation or a decrease in bone resorption. However, OVX of the hCG alphabeta+ mice resulted in a significant reduction in BMD comparable to OVX WT controls. Although gonadotropin levels are tightly linked to sex steroid titers, it appears that their effects on the skeleton are indirect.


Subject(s)
Bone and Bones/physiology , Chorionic Gonadotropin/genetics , Phenotype , Receptors, LH/deficiency , Adult , Animals , Bone Density/physiology , Cell Line , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Chorionic Gonadotropin/physiology , Cyclic AMP/biosynthesis , Female , Humans , Leydig Cell Tumor , Luteinizing Hormone/metabolism , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/chemistry , Osteoblasts/drug effects , Osteoblasts/metabolism , Ovariectomy , Ovary/chemistry , Phosphorylation , RNA, Messenger/analysis , Rats , Receptors, LH/analysis , Receptors, LH/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
15.
Reprod Biol Endocrinol ; 1: 12, 2003 Feb 06.
Article in English | MEDLINE | ID: mdl-12646063

ABSTRACT

Prolactin (PRL) exerts pleiotropic physiological effects in various cells and tissues, and is mainly considered as a regulator of reproduction and cell growth. Null mutation of the PRL receptor (R) gene leads to female sterility due to a complete failure of embryo implantation. Pre-implantatory egg development, implantation and decidualization in the mouse appear to be dependent on ovarian rather than uterine PRLR expression, since progesterone replacement permits the rescue of normal implantation and early pregnancy. To better understand PRL receptor deficiency, we analyzed in detail ovarian and corpora lutea development of PRLR-/- females. The present study demonstrates that the ovulation rate is not different between PRLR+/+ and PRLR-/- mice. The corpus luteum is formed but an elevated level of apoptosis and extensive inhibition of angiogenesis occur during the luteal transition in the absence of prolactin signaling. These modifications lead to the decrease of LH receptor expression and consequently to a loss of the enzymatic cascades necessary to produce adequate levels of progesterone which are required for the maintenance of pregnancy.


Subject(s)
Gene Expression Regulation , Ovary/metabolism , Prolactin/physiology , Receptors, Prolactin/deficiency , 20-alpha-Hydroxysteroid Dehydrogenase/biosynthesis , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 3-Hydroxysteroid Dehydrogenases/biosynthesis , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Apoptosis/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Cholesterol Side-Chain Cleavage Enzyme/genetics , Chorionic Gonadotropin/pharmacology , Corpus Luteum/physiology , Cyclin D2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/biosynthesis , Cyclins/genetics , Embryo Implantation/genetics , Embryo Implantation/physiology , Estrous Cycle/genetics , Female , Gonadotropins, Equine/pharmacology , Infertility, Female/genetics , Infertility, Female/physiopathology , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Insulin-Like Growth Factor Binding Protein 3/genetics , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Ovary/ultrastructure , Ovulation/drug effects , Ovulation/genetics , Ovulation/physiology , Pregnancy , Receptors, LH/biosynthesis , Receptors, LH/deficiency , Receptors, LH/genetics , Receptors, Prolactin/genetics , Receptors, Prolactin/physiology , Superovulation/drug effects , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
16.
Arch Med Res ; 32(6): 601-8, 2001.
Article in English | MEDLINE | ID: mdl-11750736

ABSTRACT

The development of knockout mouse models for the FSH-beta subunit, the FSH receptor, and LH-receptor performed in different laboratories has confirmed and extended our knowledge concerning the critical role of these hormone-signaling systems in spermatogenesis. In this article, we summarize the phenotypic changes observed in male FSH receptor knockout (FORKO) mice. Young FORKO males have underdeveloped testis with 50% reduction in Sertoli cells, suggesting that FSH-R signaling is required very early for gonadal development, maturity, and function. These mice experience delayed puberty with postponement in the formation of round spermatids. Adult males show reduction in serum testosterone levels despite normal circulating LH concentration, indicating disturbances in Sertoli-Leydig cell communication. As a consequence of reduced sperm production and sperm quality, adult FORKO males have reduced fertility. Aberrant sperm from FORKO males have retention of cytoplasmic droplets and inadequate DNA compaction, hallmarks of infertility in many species including man. Interestingly, these changes are also experimentally inducible in FSH- and/or FSH-R-immunized male bonnet monkeys, creating a state of infertility. Reports of human mutations in FSH-beta and the FSH receptor also indicate that spermatogenesis is dependent on this system. Further investigations in FORKO males should be helpful in uncovering the downstream genes involved in sustaining Sertoli cell function and maintenance of the quantitative and qualitative aspects of spermatogenesis. This might pave the way for treatment of male infertility and contraception.


Subject(s)
Follicle Stimulating Hormone/physiology , Signal Transduction/physiology , Spermatogenesis/physiology , Testis/physiopathology , Amino Acid Substitution , Animals , Cell Communication , Cell Nucleus/ultrastructure , Finland/epidemiology , Follicle Stimulating Hormone/deficiency , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone, beta Subunit , Humans , Infertility, Male/epidemiology , Infertility, Male/genetics , Infertility, Male/physiopathology , Macaca radiata , Male , Mice , Mice, Knockout , Mutation, Missense , Phenotype , Puberty, Delayed/genetics , Receptors, FSH/deficiency , Receptors, FSH/genetics , Receptors, LH/deficiency , Receptors, LH/genetics , Sertoli Cells/pathology , Sexual Maturation/genetics , Sperm Motility , Spermatids/pathology , Testis/pathology , Testosterone/blood , Testosterone/deficiency , Time Factors , Vaccines, Contraceptive
17.
Arq. bras. endocrinol. metab ; 45(4): 369-374, ago. 2001.
Article in Portuguese | LILACS | ID: lil-289963

ABSTRACT

As açöes fundamentais das gonadotrofinas hipofisárias na vida sexual reprodutiva de ambos os sexos dependem da integridade estrutural e funcional dos seus respectivos receptores, Os receptores das gonadotrofinas localizados na membrana citoplasmàtica säo membros da grande família dos receptores acoplados à proteína G e apresentam uma estrutura comum caracterizada por uma extensa porçäo extracelular e setes hélices transmembranas. A recente identificaçäo de mutaçöes inativadoras e ativadoras de ocorrência natural nos genes dos receptores do LH e do FSH contribuíram para a maior compreensäo de estados patológicos gonadais. Neste trabalho, revisamos os aspectos moleculares dos defeitos dos genes dos receptores das gonadotrofinas e suas implicaçöes fenotípicas no sexo feminino. Nas mulheres com mutaçöes inativadoras em homozigose nestes genes, sintomas freqüentes como alteraçöes menstruais (amenorréia secundária e oligoamenorréia) e infertilidade podem alertar o endocrinologista para o estabelecimento do diagnõsti-co definitivo da resistência ovariana ao LH ou ao FSH.


Subject(s)
Humans , Female , Infertility, Female/etiology , Receptors, FSH/deficiency , Receptors, LH/deficiency , Reproduction/physiology , Hypogonadism/etiology , Primary Ovarian Insufficiency/complications
18.
Mol Endocrinol ; 15(1): 172-83, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145748

ABSTRACT

To study further the role of gonadotropins in reproductive functions, we generated mice with LH receptor (LHR) knockout (LuRKO) by inactivating, through homologous recombination, exon 11 on the LHR gene. LuRKO males and females were born phenotypically normal, with testes, ovaries, and genital structures indistinguishable from their wild-type (WT) littermates. Postnatally, testicular growth and descent, and external genital and accessory sex organ maturation, were blocked in LuRKO males, and their spermatogenesis was arrested at the round spermatid stage. The number and size of Leydig cells were dramatically reduced. LuRKO females also displayed underdeveloped external genitalia and uteri postnatally, and their age of vaginal opening was delayed by 5-7 days. The (-/-) ovaries were smaller, and histological analysis revealed follicles up to the early antral stage, but no preovulatory follicles or corpora lutea. Reduced gonadal sex hormone production was found in each sex, as was also reflected by the suppressed accessory sex organ weights and elevated gonadotropin levels. Completion of meiosis of testicular germ cells in the LuRKO males differs from other hypogonadotropic/cryptorchid mouse models, suggesting a role for FSH in this process. In females, FSH appears to stimulate developing follicles from the preantral to early antral stage, and LH is the stimulus beyond this stage. Hence, in each sex, the intrauterine sex differentiation is independent of LH action, but it has a crucial role postnatally for attaining sexual maturity. The LuRKO mouse is a close phenocopy of recently characterized human patients with inactivating LHR mutations, although the lack of pseudohermaphroditism in LuRKO males suggests that the intrauterine sex differentiation in this species is not dependent on LH action.


Subject(s)
Embryonic and Fetal Development , Genitalia/embryology , Genitalia/growth & development , Receptors, LH/deficiency , Sexual Maturation , Animals , Cell Line , Cholesterol Side-Chain Cleavage Enzyme/genetics , Chorionic Gonadotropin/metabolism , Embryo, Mammalian , Exons , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/pharmacology , Gene Expression , Luteinizing Hormone/blood , Luteinizing Hormone/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovary/anatomy & histology , Ovary/growth & development , Phenotype , Receptors, LH/genetics , Receptors, LH/physiology , Stem Cells , Steroid 17-alpha-Hydroxylase/genetics , Steroids/blood , Testis/anatomy & histology , Testis/growth & development
19.
Mol Endocrinol ; 15(1): 184-200, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145749

ABSTRACT

LH/hCG receptors were disrupted by gene targeting in embryonic stem cells. The disruption resulted in infertility in both sexes. The gonads contained no receptor mRNA or receptor protein. Serum LH levels were greatly elevated, and FSH levels were moderately elevated in both sexes; estradiol and progesterone levels decreased but were not totally suppressed in females; testosterone levels were dramatically decreased and estradiol levels moderately elevated in males. The external and internal genitalia were grossly underdeveloped in both sexes. Abnormalities included ambiguous vaginal opening, abdominal testes, micropenis, dramatically decreased weights of the gonads and reproductive tract, arrested follicular growth beyond antral stage, disarray of seminiferous tubules, diminished number and hypotrophy of Leydig cells, and spermatogenic arrest beyond the round spermatid stage. LH/hCG receptor gene disruption had no effect on FSH receptor mRNA levels in ovaries and testes, progesterone receptor (PR) levels in ovaries and androgen receptor (AR) levels in testes. However, it caused a dramatic decrease in StAR and estrogen receptor-alpha (ERalpha) mRNA levels and an increase in ERbeta mRNA levels in both ovaries and testes. Estradiol and progesterone replacement therapy in females and testosterone replacement in males, to determine whether phenotype and biochemical changes were a consequence of decreased gonadal steroid levels or due to a loss of LH signaling, revealed complete restoration of some and partial restoration of others. Nevertheless, the animals remained infertile. It is anticipated that the LH receptor knockout animals will increase our current understanding of gonadal and nongonadal actions of LH and hCG.


Subject(s)
Receptors, LH/deficiency , Animals , Estradiol/blood , Estradiol/therapeutic use , Estrogen Receptor alpha , Female , Follicle Stimulating Hormone/blood , Gene Targeting , Genitalia/growth & development , Humans , Infertility/drug therapy , Infertility/etiology , Leydig Cells/pathology , Luteinizing Hormone/blood , Male , Mice , Mice, Knockout , Ovarian Follicle/pathology , Ovary/chemistry , Phosphoproteins/genetics , Progesterone/blood , Progesterone/therapeutic use , RNA, Messenger/analysis , Receptors, Estrogen/genetics , Receptors, LH/genetics , Receptors, LH/physiology , Seminiferous Tubules/pathology , Spermatogenesis , Testis/chemistry , Testosterone/therapeutic use
20.
Minerva Ginecol ; 42(1-2): 25-9, 1990.
Article in Italian | MEDLINE | ID: mdl-2139924

ABSTRACT

The Authors, starting from the description of a clinical case of abdominal wall endometriosis, arisen subsequently to a caesarean section, take into consideration the world literature on the subject, and focus their attention on the problem of the anatomic location and the etiopathogenesis of the pathology.


Subject(s)
Abdominal Muscles , Endometriosis , Adult , Cesarean Section/adverse effects , Cicatrix , Endometriosis/etiology , Endometriosis/pathology , Endometriosis/surgery , Female , Humans , Infertility, Female/etiology , Pregnancy , Receptors, LH/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...