Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Best Pract Res Clin Endocrinol Metab ; 34(6): 101476, 2020 12.
Article in English | MEDLINE | ID: mdl-33353780

ABSTRACT

Congenital absence of the vas deferens (CAVD) is a rare genetic condition first discovered in the mid-18th century related to mutations in the cystic fibrosis transmembrane regulatory genes. The condition is typically found during work-up of male infertility, and the majority of cases can be diagnosed with complete history and physical examination and pertinent investigations. The condition can be separated into three subcategories, and genetic advances have led to a much better understanding behind the disease, its pathogenesis, and options for treatment. In this review, we discuss the genetics, pathogenesis, embryology, and diagnosis of treatment of CAVD. Future work in this area likely will aim to better understand the epigenetic factors that influence the development of the condition in order to identify potential upstream therapeutic targets.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Infertility, Male/genetics , Mutation , Urogenital Abnormalities/genetics , Vas Deferens/abnormalities , Humans , Infertility, Male/diagnosis , Infertility, Male/embryology , Infertility, Male/therapy , Male , Urogenital Abnormalities/diagnosis , Urogenital Abnormalities/embryology , Urogenital Abnormalities/therapy , Vas Deferens/diagnostic imaging , Vas Deferens/embryology , Vas Deferens/pathology
2.
J Neurosci ; 35(37): 12903-16, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26377475

ABSTRACT

The gonadotropin-releasing hormone (GnRH) is the master regulator of fertility and kisspeptin (KP) is a potent trigger of GnRH secretion from GnRH neurons. KP signals via KISS1R, a Gαq/11-coupled receptor, and mice bearing a global deletion of Kiss1r (Kiss1r(-/-)) or a GnRH neuron-specific deletion of Kiss1r (Kiss1r(d/d)) display hypogonadotropic hypogonadism and infertility. KISS1R also signals via ß-arrestin, and in mice lacking ß-arrestin-1 or -2, KP-triggered GnRH secretion is significantly diminished. Based on these findings, we hypothesized that ablation of Gαq/11 in GnRH neurons would diminish but not completely block KP-triggered GnRH secretion and that Gαq/11-independent GnRH secretion would be sufficient to maintain fertility. To test this, Gnaq (encodes Gαq) was selectively inactivated in the GnRH neurons of global Gna11 (encodes Gα11)-null mice by crossing Gnrh-Cre and Gnaq(fl/fl);Gna11(-/-) mice. Experimental Gnaq(fl/fl);Gna11(-/-);Gnrh-Cre (Gnaq(d/d)) and control Gnaq(fl/fl);Gna11(-/-) (Gnaq(fl/fl)) littermate mice were generated and subjected to reproductive profiling. This process revealed that testicular development and spermatogenesis, preputial separation, and anogenital distance in males and day of vaginal opening and of first estrus in females were significantly less affected in Gnaq(d/d) mice than in previously characterized Kiss1r(-/-) or Kiss1r(d/d) mice. Additionally, Gnaq(d/d) males were subfertile, and although Gnaq(d/d) females did not ovulate spontaneously, they responded efficiently to a single dose of gonadotropins. Finally, KP stimulation triggered a significant increase in gonadotropins and testosterone levels in Gnaq(d/d) mice. We therefore conclude that the milder reproductive phenotypes and maintained responsiveness to KP and gonadotropins reflect Gαq/11-independent GnRH secretion and activation of the neuroendocrine-reproductive axis in Gnaq(d/d) mice. SIGNIFICANCE STATEMENT: The gonadotropin-releasing hormone (GnRH) is the master regulator of fertility. Over the last decade, several studies have established that the KISS1 receptor, KISS1R, is a potent trigger of GnRH secretion and inactivation of KISS1R on the GnRH neuron results in infertility. While KISS1R is best understood as a Gαq/11-coupled receptor, we previously demonstrated that it could couple to and signal via non-Gαq/11-coupled pathways. The present study confirms these findings and, more importantly, while it establishes Gαq/11-coupled signaling as a major conduit of GnRH secretion, it also uncovers a significant role for non-Gαq/11-coupled signaling in potentiating reproductive development and function. This study further suggests that by augmenting signaling via these pathways, GnRH secretion can be enhanced to treat some forms of infertility.


Subject(s)
GTP-Binding Protein alpha Subunits/deficiency , Gonadotropin-Releasing Hormone/physiology , Hypogonadism/physiopathology , Infertility, Female/physiopathology , Infertility, Male/physiopathology , Animals , Blastocyst/pathology , Embryonic Development , Female , GTP-Binding Protein alpha Subunits/physiology , Gene Expression Profiling , Genitalia, Female/pathology , Genitalia, Female/physiopathology , Genitalia, Male/pathology , Genitalia, Male/physiopathology , Gonadal Steroid Hormones/metabolism , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropins, Pituitary/metabolism , Gonadotropins, Pituitary/pharmacology , Hypogonadism/genetics , Hypogonadism/pathology , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamus/pathology , Infertility, Female/embryology , Infertility, Female/genetics , Infertility, Male/embryology , Infertility, Male/genetics , Kisspeptins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Oligopeptides/pharmacology , Ovariectomy , Ovulation/drug effects , Peptide Fragments/pharmacology , Peptides/pharmacology , Phenotype , Receptors, G-Protein-Coupled , Receptors, Kisspeptin-1 , Spermatogenesis
3.
Vitam Horm ; 94: 211-27, 2014.
Article in English | MEDLINE | ID: mdl-24388192

ABSTRACT

Imprinting is an epigenetic form of gene regulation that mediates a parent-of-origin-dependent expression of the alleles of a number of genes. Imprinting, which occurs at specific sites within or surrounding the gene, called differentially methylated domains, consists in a methylation of CpGs. The appropriate transmission of genomics imprints is essential for the control of embryonic development and fetal growth. A number of endocrine disruptors (EDs) affect male reproductive tract development and spermatogenesis. It was postulated that the genetic effects of EDs might be induced by alterations in gene imprinting. We tested two EDs: methoxychlor and vinclozolin. Their administration during gestation induced in the offspring a decrease in sperm counts and significant modifications in the methylation pattern of a selection of paternally and maternally expressed canonical imprinted genes. The observation that imprinting was largely untouched in somatic cells suggests that EDs exert their damaging effects via the process of reprogramming that is unique to gamete development. Interestingly, the effects were transgenerational, although disappearing gradually from F1 to F3. A systematic analysis showed a heterogeneity in the CpG sensitivity to EDs. We propose that the deleterious effects of EDs on the male reproductive system are mediated by imprinting defects in the sperm. The reported effects of EDs on human male spermatogenesis might be mediated by analogous imprinting alterations.


Subject(s)
Epigenesis, Genetic/drug effects , Fungicides, Industrial/toxicity , Infertility, Male/chemically induced , Insecticides/toxicity , Methoxychlor/toxicity , Oxazoles/toxicity , Spermatozoa/drug effects , Androgen Antagonists/toxicity , Animals , Endocrine Disruptors/toxicity , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Estrogen Antagonists/toxicity , Estrogens, Non-Steroidal/toxicity , Female , Fetal Development/drug effects , Humans , Infertility, Male/embryology , Infertility, Male/pathology , Male , Pregnancy , Spermatogenesis/drug effects , Spermatozoa/pathology
4.
Mech Dev ; 130(11-12): 567-76, 2013.
Article in English | MEDLINE | ID: mdl-23933588

ABSTRACT

Reproductive tract abnormalities and male infertility have higher incidence in ADPKD patients than in general populations. In this work, we reveal that Pkd1, whose mutations account for 85% of ADPKD cases, is essential for male reproductive tract development. Disruption of Pkd1 caused multiple organ defects in the murine male reproductive tract. The earliest visible defect in the Pkd1(-/-) reproductive tract was cystic dilation of the efferent ducts, which are derivatives of the mesonephric tubules. Epididymis development was delayed or arrested in the Pkd1(-/-) mice. No sign of epithelial coiling was seen in the null mutants. Disruption of Pkd1 in epithelium alone using the Pax2-cre mice was sufficient to cause efferent duct dilation and coiling defect in the epididymis, suggesting that Pkd1 is critical for epithelium development and maintenance in male reproductive tract. In-depth analysis showed that Pkd1 is required to maintain tubulin cytoskeleton and important for Tgf-ß/Bmp signal transduction in epithelium of male reproductive tract. Altogether, our results for the first time provide direct evidence for developmental roles of Pkd1 in the male reproductive tract and provide new insights in reproductive tract abnormalities and infertility in ADPKD patients.


Subject(s)
Body Patterning/genetics , Epididymis/metabolism , Infertility, Male/genetics , TRPP Cation Channels/genetics , Urothelium/metabolism , Wolffian Ducts/metabolism , Animals , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Embryo, Mammalian , Epididymis/abnormalities , Gene Expression Regulation, Developmental , Humans , Infertility, Male/embryology , Infertility, Male/metabolism , Infertility, Male/pathology , Integrases/genetics , Integrases/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Signal Transduction , TRPP Cation Channels/deficiency , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tubulin/metabolism , Tubulin/ultrastructure , Urothelium/abnormalities , Wolffian Ducts/abnormalities
6.
Reproduction ; 142(1): 133-44, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21487003

ABSTRACT

Heat-shock protein 110 (HSP110) family members act as nucleotide exchange factors (NEF) of mammalian and yeast HSP70 chaperones during the ATP hydrolysis cycle. In this study, we describe the expression pattern of murine HSPA4, a member of the HSP110 family, during testis development and the consequence of HSPA4 deficiency on male fertility. HSPA4 is ubiquitously expressed in all the examined tissues. During prenatal and postnatal development of gonad, HSPA4 is expressed in both somatic and germ cells; however, expression was much higher in germ cells of prenatal gonads. Analyses of Hspa4-deficient mice revealed that all homozygous mice on the hybrid C57BL/6J×129/Sv genetic background were apparently healthy. Although HSPA4 is expressed as early as E13.5 in male gonad, a lack of histological differences between Hspa4(-/-) and control littermates suggests that Hspa4 deficiency does not impair the gonocytes or their development to spermatogonia. Remarkably, an increased number of the Hspa4-deficient males displayed impaired fertility, whereas females were fertile. The total number of spermatozoa and their motility were drastically reduced in infertile Hspa4-deficient mice compared with wild-type littermates. The majority of pachytene spermatocytes in the juvenile Hspa4(-/-) mice failed to complete the first meiotic prophase and became apoptotic. Furthermore, down-regulation of transcription levels of genes known to be expressed in spermatocytes at late stages of prophase I and post-meiotic spermatids leads to suggest that the development of most spermatogenic cells is arrested at late stages of meiotic prophase I. These results provide evidence that HSPA4 is required for normal spermatogenesis.


Subject(s)
HSP110 Heat-Shock Proteins/physiology , Infertility, Male/metabolism , Spermatogenesis , Spermatozoa/metabolism , Animals , Apoptosis , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Embryonic Development , Female , Fertility , HSP110 Heat-Shock Proteins/genetics , Homozygote , Infertility, Male/embryology , Infertility, Male/pathology , Male , Meiotic Prophase I , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , RNA, Messenger/metabolism , Semen Analysis , Sperm Motility , Spermatozoa/pathology , Testis/embryology , Testis/growth & development , Testis/metabolism , Testis/pathology
7.
Zygote ; 17(2): 157-62, 2009 May.
Article in English | MEDLINE | ID: mdl-19079830

ABSTRACT

Intracytoplasmic sperm injection (ICSI) is an assisted fertilization technique and has been widely applied in human medicine to overcome some obstacles of infertility. However, this technology has not yet been used as a mainstream technique for animal production, including the rabbit, due to its limited success. The aim of this study was to improve ICSI techniques and establish an efficient ICSI method for rabbits. Spermatozoa used for ICSI were collected from mature New Zealand white male rabbits. They were washed two to three times with HEPES-buffered TCM199 containing 10% FBS and then mixed with 10% polyvinylpyrrolidone (PVP) prior to microinjection. Oocytes were harvested from superovulated donor rabbits after 14-15 h hCG treatment and were fertilized by microinjection of a single living spermatozoon into the ooplasm of each oocyte without additional activation treatment. A total of 317 injected oocytes resulted in the high survival rate of 86.1%. Among the surviving oocytes, 273 were placed into culture dishes for in vitro development. The fertilization, cleavage and blastocyst rates were 59.0%, 88.2% and 45.3% respectively. Furthermore, ICSI embryos were produced with spermatozoa from an infertile male rabbit, and 21 early-stage embryos (2-cell and 4-cell) were surgically transferred into the oviducts of two adult female rabbits. On day 31 after transfer, one out of the two recipients gave birth to two normal and healthy young rabbits. These results demonstrate that rabbit oocytes can be successfully activated and fertilized by the new ICSI protocol. Spermatozoa derived from infertile rabbits can successful fertilize oocytes and produce offspring by the simple ICSI technique.


Subject(s)
Embryonic Development/physiology , Microinjections/methods , Sperm Injections, Intracytoplasmic/methods , Animals , Embryo Transfer , Female , Infertility, Male/embryology , Male , Oocytes/physiology , Rabbits , Spermatozoa/physiology
8.
Biol Reprod ; 78(6): 994-1001, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18322278

ABSTRACT

Amniotes, regardless of genetic sex, develop two sets of genital ducts: the Wolffian and Müllerian ducts. For normal sexual development to occur, one duct must differentiate into its corresponding organs, and the other must regress. In mammals, the Wolffian duct differentiates into the male reproductive tract, mainly the vasa deferentia, epididymides, and seminal vesicles, whereas the Müllerian duct develops into the four components of the female reproductive tract, the oviducts, uterus, cervix, and upper third of the vagina. In males, the fetal Leydig cells produce testosterone, which stimulates the differentiation of the Wolffian duct, whereas the Sertoli cells of the fetal testes express anti-Müllerian hormone, which activates the regression of the Müllerian duct. Anti-Müllerian hormone is a member of the transforming growth factor-beta (TGF-beta) family of secreted signaling molecules and has been shown to signal through the BMP pathway. It binds to its type II receptor, anti-Müllerian hormone receptor 2 (AMHR2), in the Müllerian duct mesenchyme and through an unknown mechanism(s); the mesenchyme induces the regression of the Müllerian duct mesoepithelium. Using tissue-specific gene inactivation with an Amhr2-Cre allele, we have determined that two TGF-beta type I receptors (Acvr1 and Bmpr1a) and all three BMP receptor-Smads (Smad1, Smad5, and Smad8) function redundantly in transducing the anti-Müllerian hormone signal required for Müllerian duct regression. Loss of these genes in the Müllerian duct mesenchyme results in male infertility due to retention of Müllerian duct derivatives in an otherwise virilized male.


Subject(s)
Activin Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Mullerian Ducts/embryology , Mullerian Ducts/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Activin Receptors, Type I/deficiency , Activin Receptors, Type I/genetics , Animals , Anti-Mullerian Hormone/pharmacology , Bone Morphogenetic Protein Receptors, Type I/deficiency , Bone Morphogenetic Protein Receptors, Type I/genetics , Female , Infertility, Male/embryology , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Models, Biological , Mullerian Ducts/drug effects , Pregnancy , Signal Transduction , Smad1 Protein/genetics , Smad5 Protein/genetics , Smad8 Protein/genetics
9.
Reproduction ; 133(5): 1057-67, 2007 May.
Article in English | MEDLINE | ID: mdl-17616734

ABSTRACT

Previously, we reported an association between estrogen receptor-alpha (ERalpha) upregulation and detrimental effects of neonatal diethylstilbestrol (DES) exposure in the rat penis. The objective of this study was to employ the ERalpha knockout (ERalphaKO) mouse model to test the hypothesis that ERalpha mediates DES effects in the developing penis. ERalphaKO and wild-type C57BL/6 mice received oil or DES at a dose of 0.2 microg/pup per day (0.1 mg/kg) on alternate days from postnatal days 2 to 12. Fertility was tested at 80-240 days of age and tissues were examined at 96-255 days of age. DES caused malformation of the os penis, significant reductions in penile length, diameter, and weight, accumulation of fat cells in the corpora cavernosa penis, and significant reductions in weight of the bulbospongiosus and levator ani muscles in wild-type mice. Conversely, ERalphaKO mice treated with DES developed none of the above abnormalities. While nine out of ten male mice sired pups in the wild-type/control group, none did in the wild-type/DES group. ERalphaKO mice, despite normal penile development, are inherently infertile. Both plasma and intratesticular testosterone levels were unaltered in the DES-treated wild-type or DES-treated ERalphaKO mice when compared with controls, although testosterone concentration was much higher in the ERalphaKO mice. Hence, the resistance of ERalphaKO mice to developing penile abnormalities provides unequivocal evidence of an obligatory role for ERalpha in mediating the harmful effects of neonatal DES exposure in the developing penis.


Subject(s)
Diethylstilbestrol/toxicity , Estrogen Receptor alpha/metabolism , Estrogens, Non-Steroidal/toxicity , Infertility, Male/chemically induced , Penis/embryology , Animals , Animals, Newborn , Estrogen Receptor alpha/genetics , Female , Histocytochemistry , Infertility, Male/embryology , Infertility, Male/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Penis/metabolism , Rats , Testis/chemistry , Testis/pathology , Testosterone/analysis , Testosterone/blood
10.
Congenit Anom (Kyoto) ; 47(1): 34-44, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17300688

ABSTRACT

Congenital hypoplasia and dysplasia affect the postnatal development of organs, their physiological functioning in adulthood and the incidence of related diseases at an advanced age. Hypogonadic (hgn/hgn) rats are characterized by male sterility, reduced female fertility, progressive renal insufficiency and growth retardation, all controlled by a single recessive allele (hgn) located on chromosome 10. Since our previous studies indicated that the hypoplasia (dysplasia) of the affected organs was present at birth, we examined the embryonic pathogenesis. We mated hgn/hgn females to Brown Norway males and backcrossed F(1) males to hgn/hgn females. The resulting N(1) fetuses were genotyped using a hgn-linked microsatellite. Both sexes of hgn/hgn fetuses showed low body weight after embryonic day (ED) 15.5 and renal hypoplasia after ED 17.5. Their kidneys contained a reduced number of nephrons in a poorly formed nephrogenic zone and renal cortex. The hgn/hgn ovaries contained a small number of oogonia at ED 15.5 and oocytes after ED 17.5. Testicular growth defects were obvious after ED 17.5, and reduced numbers of Sertoli cells were detected at ED 19.5 and 21.5. The seminiferous cords in hgn/hgn testes contained more apoptotic and mitotic cells than those in +/hgn testes. These findings suggest that the phenotypes described in adult hgn/hgn rats result from embryonic hypogenesis, which continues to early postnatal stage and causes a reduction in functional tissues and cells. Since hgn/hgn rats have an insertion mutation in the microtubule-associated protein Spag5 gene, the embryonic hypogenesis described in hgn/hgn rats might result from defective cell proliferation.


Subject(s)
Hypogonadism/embryology , Infertility, Female/embryology , Infertility, Male/embryology , Kidney/abnormalities , Renal Insufficiency/embryology , Animals , Female , Hypogonadism/pathology , Infertility, Female/pathology , Infertility, Male/pathology , Kidney/anatomy & histology , Kidney/embryology , Kidney/growth & development , Kidney/pathology , Male , Ovum/growth & development , Ovum/pathology , Phenotype , Rats , Rats, Inbred Strains , Renal Insufficiency/pathology , Spermatozoa/growth & development , Spermatozoa/pathology , Testis/embryology , Testis/growth & development , Testis/pathology
11.
Fiziol Zh (1994) ; 52(3): 110-8, 2006.
Article in English | MEDLINE | ID: mdl-16909765

ABSTRACT

The fact that genetic aetiology is responsible for approximately one third of all kinds of male sterility has led to concerns regarding the application of artificial reproductive technologies in the cases of azoospermia and severe oligozoospermia. Congenital sterility could be caused by gene mutation, quantitative or structural abnormality of sexual chromosomes or autosomes. The possibility of inherited transmission of male sterility within the context of applying artificial reproductive technologies is analysed in the article. Klinefelter's syndrome, mutation of cystic fibrosis transmembrane conductance regulator (CFTR) gene, which causes cystic fibrosis, and mutation of azoospermia factor zone (AZF) of Y-chromosome are among the most frequent genetic causes of severe oligozoospermia and azoospermia. The probability of a mutation of the CFTR gene being transmitted to the next generation is 50%. The probability of inherit transmission of Klinefelter's syndrome of mosaic karyotype could reach 70%. The probability of transmission of AZF mutation to children of male sex, generated with ICSI, is 100%. The percent of men with AZF mutation among users of ICSI centers, ranges from 3.2% to 14%. It means that at least 3.2 % of all boys conceived with ICSI will be sterile because of mutation in the AZF zone of Y-chromosome. It should be noted that genetic analysis and counselling do not always occur before the beginning of artificial fertilization cycles. Parents-to-be do not always have an opportunity to learn about the genetic risks of their unborn child and make any subsequent and responsible decisions. Among the decisions is a choice of not resorting to ART, but to live with the infertility and explore other opportunities for parenthood. Responsibility for high risk of conceiving a child with genetic anomalies rests not only with the parents, but also with all of society, including those responsible for research, technology, and legislation, in the health care profession.


Subject(s)
Infertility, Male/congenital , Sex Chromosome Aberrations , Sperm Injections, Intracytoplasmic , Chromosome Deletion , Chromosomes, Human, Y/genetics , Humans , Infertility, Male/embryology , Infertility, Male/genetics , Male , Sex Chromosome Aberrations/embryology
13.
Article in English | MEDLINE | ID: mdl-11097788

ABSTRACT

Our understanding of the importance of environmental and lifestyle factors on sperm count and fertility is constrained by the extreme variation in sperm count between men and between ejaculates. The factors responsible for this variation provide a key to understanding what factors actually affect the sperm count. The relative importance of the various factors and the pathways via which they affect sperm count are discussed. The most important are Sertoli cell number, ejaculatory frequency, season, factors affecting scrotal heating (e.g. the time spent seated), a past history of reproductive tract disorders and ageing. The possible role of other environmental factors commonly supposed to affect sperm count (exposure to pesticides or endocrine disruptors) is discussed, although the evidence for a major influence of such factors is lacking. It is suggested that lifestyle changes, especially in the time spent seated, will exert an adverse effect on sperm production in a progressively larger group of men over the next decade.


Subject(s)
Environment , Infertility, Male/etiology , Life Style , Demography , Environmental Exposure , Estrogens/adverse effects , Hot Temperature/adverse effects , Humans , Infertility, Male/chemically induced , Infertility, Male/embryology , Infertility, Male/pathology , Male , Pesticides/adverse effects , Sperm Count
15.
Br Med Bull ; 56(3): 630-42, 2000.
Article in English | MEDLINE | ID: mdl-11255550

ABSTRACT

This chapter is an overview of recent developments in our understanding and thinking about the importance and nature of environmental effects on sperm counts and fertility in the human male. This area is plagued by imperfect studies, not necessarily because of imperfect design but because of other 'uncontrollable' constraints. The available data, therefore, need to be placed in context and account taken of the limitations of our understanding or, more correctly, our ignorance. As we enter the new millennium, one of the saddest scientific aspects of human reproduction and infertility is our persisting ignorance about the causes and treatment of male infertility. With one notable exception (Y chromosome microdeletions) there has been little advance in our understanding of the causes of male infertility and its direct treatment over the past 20 years. Although most infertile men can now be offered the chance of fertility via ICSI, it is largely ignored that this does not represent treatment of the patient's infertility (which will persist unchanged), but is a means of circumventing the problem and leaving it for the next generation to tackle. There are many reasons for our ignorance about the causes of infertility, and some of these are outlined below in order to emphasise how this limits our ability to establish whether or not specific lifestyle and environmental factors do, or do not, affect human male reproductive function.


Subject(s)
Environment , Infertility, Male/etiology , Life Style , Adult , Anabolic Agents/adverse effects , Drug Residues/adverse effects , Exercise , Humans , Incidence , Infertility, Male/embryology , Male , Occupations , Pesticides/adverse effects , Phenols/adverse effects , Phthalic Acids/adverse effects , Seasons , Sperm Count , Spermatogenesis/physiology , Temperature , Testicular Neoplasms/embryology , Testicular Neoplasms/epidemiology , Testis/physiology , Veterinary Drugs/adverse effects
16.
Mol Endocrinol ; 12(6): 775-84, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9626653

ABSTRACT

Leydig cell hypoplasia (LCH) is characterized by a decreased response of the Leydig cells to LH. As a result, patients with this syndrome display aberrant male development ranging from complete pseudohermaphroditism to males with micropenis but with otherwise normal sex characteristics. We have evaluated three brothers with a mild form of LCH. Analysis of their LH receptor (LHR) gene revealed a homozygous missense mutation resulting in a substitution of a lysine residue for a isoleucine residue at position 625 of the receptor. In vitro analysis of this mutant LHR, LHR(I625K), in HEK293 cells indicated that the signaling efficiency was significantly impaired, which explains the partial phenotype. We have compared this mutant LHR to two other mutant LHRs, LHR(A593P) and LHR(S616Y), identified in a complete and partial LCH patient, respectively. Although the ligand-binding affinity for all three mutant receptors was normal, the hormonal response of LHR(A593P) was completely absent and that of LHR(S616Y) and LHR(I625K) was severely impaired. Low cell surface expression explained the reduced response of LHR(S616Y), while for LHR(I625K) this diminished response was due to a combination of low cell surface expression and decreased coupling efficiency. For LHR(A593P), the absence of a reduced response resulted from both poor cell surface expression and a complete deficiency in coupling. Our experiments further show a clear correlation between the severity of the clinical phenotype of patients and overall receptor signal capacity, which is a combination of cell surface expression and coupling efficiency.


Subject(s)
Gonadotropin-Releasing Hormone/pharmacology , Leydig Cells/metabolism , Receptors, LHRH/genetics , Sex Differentiation/physiology , Testis/pathology , Testosterone/deficiency , Adult , Amino Acid Sequence , Animals , Cell Line , Chorionic Gonadotropin/pharmacology , Codon/genetics , Genitalia, Male/abnormalities , Genitalia, Male/embryology , Humans , Infertility, Male/embryology , Infertility, Male/genetics , Isoleucine/chemistry , Leydig Cells/drug effects , Lysine/chemistry , Male , Middle Aged , Molecular Sequence Data , Phenotype , Pituitary Hormones/blood , Point Mutation , Polymorphism, Single-Stranded Conformational , Puberty, Delayed/genetics , Receptors, LHRH/chemistry , Receptors, LHRH/physiology , Recombinant Fusion Proteins/physiology , Signal Transduction , Structure-Activity Relationship , Testis/metabolism , Testosterone/metabolism , Transfection , Tumor Cells, Cultured
18.
Nature ; 374(6521): 460-3, 1995 Mar 30.
Article in English | MEDLINE | ID: mdl-7700356

ABSTRACT

The Abdominal B (AbdB) genes constitute a distinct subfamily of homeobox genes that exhibit posterior domains of expression, including the genital imaginal disc in Drosophila and the developing urogenital system in vertebrates. We have mutated the AbdB gene Hoxa10 in mice. We report here that homozygotes are fully viable and show an anterior homeotic transformation of lumbar vertebrae. All male homozygotes manifest bilateral cryptorchidism resulting in severe defects in spermatogenesis and increasing sterility with age. Female homozygotes ovulate normally, but about 80% are sterile because of death of embryos between days 2.5 and 3.5 post coitum. This coincides spatially and temporally with expression of maternal Hoxa10 in distal oviductal and uterine epithelium. These results indicate a role for AbdB Hox genes in male and female fertility and suggest that maternal Hoxa10 is required to regulate the expression of a factor that affects the viability of preimplantation embryos.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Homeobox , Homeodomain Proteins , Infertility, Male/genetics , Sex Characteristics , Animals , Cryptorchidism/genetics , DNA-Binding Proteins/physiology , Embryonic and Fetal Development/genetics , Female , Homeobox A10 Proteins , Homozygote , Infertility, Male/embryology , Lumbar Vertebrae/abnormalities , Lumbar Vertebrae/embryology , Male , Mesoderm , Mice , Mutagenesis , Ovulation/genetics , Phenotype , Spermatogenesis/genetics
19.
Dev Neurosci ; 13(4-5): 352-62, 1991.
Article in English | MEDLINE | ID: mdl-1817042

ABSTRACT

Gaucher disease is the most common sphingolipid storage disorder. Due to its high prevalence it may appear with a nonrelated neurological disease and be misinterpreted as Gaucher type 3. A family is described in which 2 Gaucher brothers presented different clinical signs. Molecular analysis has shown that both carried two mutated alleles. One allele had a G to C transversion at nucleotide 3119 of the active gene (Asp140-His) while the other presented two base pair changes, an A to C transversion at nucleotide number 3170 (Lys157-Gly), and a G-A transition at nucleotide number 5309 (Glu324-Lys). Therefore, both presented the same type of Gaucher disease which was accompanied with a nonrelated neurological disease in one of them. Molecular diagnosis of 161 patients has provided a relative abundance of different mutations among Jewish and non-Jewish patients and allowed some genotype-phenotype correlation. Differential expression of the murine glucocerebrosidase activator gene (the prosaposine) has been demonstrated using Northern technique and in situ hybridization. High expression levels were observed in the brain and testes. In the testes the prosaposine expression was confined to the supporting cells. In the female gonad prosaposine expression has also been shown, in the corpus luteum. In a 12 1/2-day-old embryo, prosaposine gene expression was detected mainly in brain stem, in dorsal ganglia and in the genital ridge.


Subject(s)
Gaucher Disease/genetics , Glucosylceramidase/genetics , Glycoproteins/genetics , Protein Precursors/genetics , Adolescent , Adult , Alleles , Animals , Child , DNA/genetics , DNA Mutational Analysis , Diagnosis, Differential , Feedback , Female , Gaucher Disease/diagnosis , Gaucher Disease/ethnology , Gene Expression Regulation , Gene Frequency , Genes , Glucosylceramidase/biosynthesis , Humans , Infant , Infertility, Male/embryology , Jews , Male , Mice/embryology , Mice/genetics , Mutation , Nervous System Diseases/diagnosis , Ovary/metabolism , Pseudogenes , RNA Splicing , Rats , Saposins , Testis/embryology , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...