Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Development ; 127(20): 4383-93, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003838

ABSTRACT

The development of the enteric nervous system is dependent upon the actions of glial cell line-derived neurotrophic factor (GDNF) on neural crest-derived precursor cells in the embryonic gut. GDNF treatment of cultured enteric precursor cells leads to an increase in the number of neurons that develop and/or survive. Here we demonstrate that, although GDNF promoted an increase in neuron number at all embryonic ages examined, there was a developmental shift from a mitogenic to a trophic response by the developing enteric neurons. The timing of this shift corresponded to developmental changes in gut expression of GFR alpha-1, a co-receptor in the GDNF-Ret signaling complex. GFR alpha-1 was broadly expressed in the gut at early developmental stages, at which times soluble GFR alpha-1 was released into the medium by cultured gut cells. At later times, GFR alpha-1 became restricted to neural crest-derived cells. GFR alpha-1 could participate in GDNF signaling when expressed in cis on the surface of enteric precursor cells, or as a soluble protein. The GDNF-mediated response was greater when cell surface, compared with soluble, GFR alpha-1 was present, with the maximal response seen the presence of both cis and trans forms of GFR alpha-1. In addition to contributing to GDNF signaling, cell-surface GFR alpha-1 modulated the specificity of interactions between GDNF and soluble GFR alphas. These experiments demonstrate that complex, developmentally regulated, signaling interactions contribute to the GDNF-dependent development of enteric neurons.


Subject(s)
Drosophila Proteins , Enteric Nervous System/embryology , Nerve Growth Factors , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Cell Division/drug effects , Digestive System/embryology , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , In Vitro Techniques , Ligands , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neural Crest/embryology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/genetics , Stem Cells/drug effects , Tissue Distribution
2.
Am J Pathol ; 156(1): 115-24, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10623659

ABSTRACT

A prominent feature of the hepatic response to injury is production of a fetal isoform of fibronectin, a splice variant containing the EIIIA region, which appears very early after injury and derives from sinusoidal endothelial cells. Previous studies have shown that it is instrumental in initiating the cellular response to injury, specifically the conversion of resting stellate cells to myofibroblast-like cells. The present work describes the regulation of this change in fibronectin expression. Using sinusoidal endothelial cells from normal or injured liver in primary culture, we show that exogenous transforming growth factor beta (TGFbeta) stimulates [EIIIA]Fn expression. To assess the role of TGFbeta in vivo, we used a chimeric IgG containing the extracellular portion of the TGFbeta type II receptor as a competitive inhibitor of the cytokine. Administered to animals at the time of injury, the inhibitor reduced expression of [EIIIA]Fn mRNA by 50% as compared to controls (P < 0.01). There was a corresponding decrease in [EIIIA]Fn protein production as judged by immunohistochemistry. Cell fractionation experiments indicated that the changes observed in whole-liver extracts were localized to sinusoidal endothelial cells. We conclude that TGFbeta initiates wound repair in part by stimulating endothelial expression of [EIIIA]Fn. Results with the soluble inhibitor of the TGFbeta type II receptor suggest a novel strategy for modulating wound repair in vivo.


Subject(s)
Fibronectins/genetics , Genetic Variation , Liver/drug effects , Transforming Growth Factor beta/pharmacology , Wound Healing/physiology , Animals , Autocrine Communication , Bile Ducts , DNA, Recombinant , Endothelium/metabolism , Endothelium/pathology , Ligation , Liver/metabolism , Liver/pathology , Liver/physiology , Male , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Transforming Growth Factor beta/physiology , Transforming Growth Factor beta/physiology , Up-Regulation
3.
J Clin Endocrinol Metab ; 84(10): 3836-44, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10523039

ABSTRACT

The ontogeny of expression of anti-Müllerian hormone (AMH) was examined by immunohistochemistry in 135 human gonadal tissue specimens of various developmental age, ranging from 6 weeks of fetal development to 38 yr of postnatal age. The series included specimens from normal testes and ovaries and from individuals either with pathological conditions affecting gonadal development or with idiopathic infertility manifested as azoospermia or severe oligozoospermia. AMH expression was found only in Sertoli and granulosa cells. A 6-week-old fetal testis at the indifferent gonad stage did not yet express AMH. The protein was first visible at 8.5 weeks of development, when sex cords have not yet been formed. Afterward, a majority of testicular specimens, including those from pathological conditions, strongly expressed AMH through fetal development and childhood until puberty. Markedly prolonged expression of AMH was observed in a 20-yr-old 46,XY female with androgen insensitivity syndrome, who retained prepubertal testicular morphology. In normal testes, the switch-off of AMH expression was usually associated with the appearance of primary spermatocytes, suggesting that their presence had an inhibitory effect on AMH. However, in adolescent boys lacking germ cells because of cancer treatment and in a majority of infertile adult men with idiopathic germ cell aplasia, AMH expression was also down-regulated despite the complete lack of spermatogenesis. The decrease in AMH expression thus reflects the terminal differentiation of Sertoli cells and is probably only partially dependent upon a regulatory factor associated with the onset of meiosis. In fetal ovaries, AMH was first detected at 36 weeks gestation in granulosa cells of preantral follicles. Thus, the onset of ovarian expression is at the end of fetal life and not in infancy as previously reported.


Subject(s)
Aging/metabolism , Genitalia/metabolism , Glycoproteins , Gonads/embryology , Gonads/growth & development , Growth Inhibitors/metabolism , Testicular Hormones/metabolism , Adolescent , Adult , Anti-Mullerian Hormone , Cell Differentiation , Child , Child, Preschool , Disease , Female , Fetus/metabolism , Granulosa Cells/cytology , Humans , Immunohistochemistry , Infant , Male , Reference Values , Sertoli Cells/cytology
4.
Dev Genet ; 24(3-4): 263-72, 1999.
Article in English | MEDLINE | ID: mdl-10322634

ABSTRACT

We have used a RET-Ig fusion protein to disrupt signaling in the rat embryonic kidney development pathway. Treatment of embryonic kidney organ cultures with RET-Ig results in a decrease in branching of the ureteric bud and a down regulation in expression of the Wnt-11, Wnt-4, and ld genes. These data suggest that Wnt-11, Wnt-4, and ld function downstream of RET signaling in normal development. Expression of BMP-7, shh, and ptc were uneffected by RET-Ig treatment, implying that these genes are regulated independently of ret. We have also performed immunohistochemistry with a GFR alpha-1 specific polyclonal antisera to localize GFR alpha-1 protein expression in the developing kidney.


Subject(s)
Drosophila Proteins , Kidney/embryology , Kidney/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Trans-Activators , Transforming Growth Factor beta , Animals , Base Sequence , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , DNA Primers/genetics , Fetal Proteins/genetics , Formins , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor Receptors , Glycoproteins/genetics , Hedgehog Proteins , Humans , Immunohistochemistry , In Situ Hybridization , Membrane Proteins/genetics , Microfilament Proteins , Nuclear Proteins/genetics , Organ Culture Techniques , Patched Receptors , Proteins/genetics , Proto-Oncogene Proteins c-ret , Rats , Receptors, Cell Surface , Recombinant Fusion Proteins/genetics , Signal Transduction , Wnt Proteins , Wnt4 Protein
5.
Oncogene ; 16(5): 597-601, 1998 Feb 05.
Article in English | MEDLINE | ID: mdl-9482105

ABSTRACT

GDNFR-alpha is a glycosyl-phosphotidylinositol-linked receptor for glial cell line-derived neurotrophic factor (GDNF). GDNF binds to GDNFR-alpha and this complex, in turn, is believed to interact with the RET receptor tyrosine kinase to effect downstream signalling. GDNFR-alpha belongs to a novel gene family without strong homology to known genes. Thus, little information has been available to help predict genomic structure or location of this gene. In this study, the genomic organization of human GDNFR-alpha was delineated through a combination of PAC clone characterization, long distance PCR and sequence analyses. Exon-intron boundaries were defined by comparing the size and sequence of the genomic PCR products to those predicted by the cDNA sequence. The human GDNFR-alpha gene comprises 9 exons. GDNFR-alpha PAC clones were used for FISH analysis to map this gene to 10q26.


Subject(s)
Chromosomes, Human, Pair 10 , Drosophila Proteins , Genome, Human , Nerve Growth Factors , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Base Sequence , Carcinoma, Medullary/genetics , Chromosome Mapping , Exons , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , In Situ Hybridization, Fluorescence , Introns , Nerve Tissue Proteins/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/metabolism , Thyroid Neoplasms/genetics , Tumor Cells, Cultured
6.
J Biol Chem ; 273(7): 4135-42, 1998 Feb 13.
Article in English | MEDLINE | ID: mdl-9461608

ABSTRACT

We report the identification of rat and human cDNAs for a type 1 membrane protein that contains a novel six-cysteine immunoglobulin-like domain and a mucin domain; it is named kidney injury molecule-1 (KIM-1). Structurally, KIM-1 is a member of the immunoglobulin gene superfamily most reminiscent of mucosal addressin cell adhesion molecule 1 (MAdCAM-1). Human KIM-1 exhibits homology to a monkey gene, hepatitis A virus cell receptor 1 (HAVcr-1), which was identified recently as a receptor for the hepatitis A virus. KIM-1 mRNA and protein are expressed at a low level in normal kidney but are increased dramatically in postischemic kidney. In situ hybridization and immunohistochemistry revealed that KIM-1 is expressed in proliferating bromodeoxyuridine-positive and dedifferentiated vimentin-positive epithelial cells in regenerating proximal tubules. Structure and expression data suggest that KIM-1 is an epithelial cell adhesion molecule up-regulated in the cells, which are dedifferentiated and undergoing replication. KIM-1 may play an important role in the restoration of the morphological integrity and function to postischemic kidney.


Subject(s)
Cell Adhesion Molecules/chemistry , Immunoglobulins/chemistry , Kidney/injuries , Membrane Proteins , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Gene Expression Regulation/genetics , Immunohistochemistry , In Situ Hybridization , Ischemia/metabolism , Kidney/chemistry , Male , Molecular Sequence Data , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Up-Regulation/physiology
7.
Biochem Biophys Res Commun ; 237(3): 741-6, 1997 Aug 28.
Article in English | MEDLINE | ID: mdl-9299437

ABSTRACT

We have determined the sequence and structure of the mouse Müllerian-inhibiting substance (MIS) type II receptor gene. Sequence comparisons demonstrate that the mouse, rat, rabbit, and human MIS type II receptors are highly conserved. The mouse MIS type II receptor gene is encoded by 11 exons and spans approximately 9-kb. Only half of the intron/exon boundaries of its kinase domain are conserved in comparison to the kinase domain of the related activin type II receptor. Whereas the activin type II receptor gene contains large introns (> 40-kb), the largest intron of the MIS type II receptor gene is only 4.3-kb. The MIS type II receptor gene (Amhr) is closely linked to Hoxc on mouse chromosome 15. Knowledge of the sequence and genomic structure of Amhr provides important information for the genetic manipulation of the Amhr locus.


Subject(s)
Chromosome Mapping , Mice/genetics , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Amino Acid Sequence , Animals , Base Sequence , Consensus Sequence , Conserved Sequence , Exons , Humans , Introns , Molecular Sequence Data , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Rabbits , Rats , Receptors, Peptide/biosynthesis , Receptors, Transforming Growth Factor beta , Restriction Mapping , Sequence Alignment , Sequence Homology, Amino Acid
8.
Proc Natl Acad Sci U S A ; 94(12): 6238-43, 1997 Jun 10.
Article in English | MEDLINE | ID: mdl-9177201

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF)-dependent activation of the tyrosine kinase receptor RET is necessary for kidney and enteric neuron development, and mutations in RET are associated with human diseases. Activation of RET by GDNF has been shown to require an accessory component, GDNFR-alpha (RETL1). We report the isolation and characterization of rat and human cDNAs for a novel cell-surface associated accessory protein, RETL2, that shares 49% identity with RETL1. Both RETL1 and RETL2 can mediate GDNF dependent phosphorylation of RET, but they exhibit different patterns of expression in fetal and adult tissues. The most striking differences in expression observed were in the adult central and peripheral nervous systems. In addition, the mechanisms by which the two accessory proteins facilitate the activation of RET by GDNF are quite distinct. In vitro binding experiments with soluble forms of RET, RETL1 and RETL2 demonstrate that while RETL1 binds GDNF tightly to form a membrane-associated complex which can then interact with RET, RETL2 only forms a high affinity complex with GDNF in the presence of RET. This strong RET dependence of the binding of RETL2 to GDNF was confirmed by FACS analysis on RETL1 and RETL2 expressing cells. Together with the recent discovery of a GDNF related protein, neurturin, these data raise the possibility that RETL1 and RETL2 have distinctive roles during development and in the nervous system of the adult. RETL1 and RETL2 represent new candidate susceptibility genes and/or modifier loci for RET-associated diseases.


Subject(s)
Brain/metabolism , Drosophila Proteins , Nerve Tissue Proteins/pharmacology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Spinal Cord/metabolism , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Embryo, Mammalian , Flow Cytometry , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Kidney/metabolism , Kinetics , Molecular Sequence Data , Nerve Growth Factors/pharmacology , Organ Specificity , Protein Binding , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-ret , Rats , Rats, Wistar , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Sequence Alignment
9.
Genes Dev ; 10(20): 2577-87, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8895659

ABSTRACT

Müllerian-inhibiting substance (MIS) is a member of the transforming growth factor-beta (TGF-beta) gene family. MIS expression in males causes the regression of the Müllerian ducts, an essential process in male sexual differentiation. Recently, an MIS type II receptor gene has been isolated that is expressed during embryogenesis in mesenchymal cells adjacent to the Müllerian duct epithelium and in Sertoli and granulosa cells of the fetal and adult, male and female gonads, respectively. MIS receptor mutant males develop as internal pseudohermaphrodites, possessing a complete male reproductive tract and also a uterus and oviducts, a phenocopy of MIS ligand-deficient male mice. They express both MIS mRNA and protein, showing that ligand was present, but target organs were hormone-insensitive. All produce sperm, but the majority were infertile because the presence of their female reproductive organs blocks sperm transfer into females. Focal seminiferous tubule atrophy accompanied by Leydig cell hyperplasia was observed and began as early as 2 months of age. The phenotype of MIS ligand/MIS receptor double mutant males was indistinguishable from those of each single mutant. MIS receptor/alpha-inhibin double mutant males developed testicular stromal tumors and large fluid-filled uteri that were identical in phenotype to MIS ligand/alpha-inhibin double mutant males. These studies provide in vivo evidence that MIS is the only ligand of the MIS type II receptor, in contrast to the complexity of other TGF-beta gene family signaling pathways.


Subject(s)
Receptors, Peptide/genetics , Sex Differentiation/genetics , Signal Transduction/genetics , Alleles , Animals , Carcinogenicity Tests , Disorders of Sex Development , Female , Fertility , Germ Cells , Hyperplasia , Inhibins/genetics , Leydig Cells , Ligands , Male , Mice , Mice, Inbred C57BL , Rabbits , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta , Seminiferous Tubules , Testicular Neoplasms/physiopathology
10.
Nat Genet ; 11(4): 382-8, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7493017

ABSTRACT

Anti-Müllerian hormone (AMH) and its receptor are involved in the regression of Müllerian ducts in male fetuses. We have now cloned and mapped the human AMH receptor gene and provide genetic proof that it is required for AMH signalling, by identifying a mutation in the AMH receptor in a patient with persistent Müllerian duct syndrome. The mutation destroys the invariant dinucleotide at the 5' end of the second intron, generating two abnormal mRNAs, one missing the second exon, required for ligand binding, and the other incorporating the first 12 bases of the second intron. The similar phenotypes observed in AMH-deficient and AMH receptor-deficient individuals indicate that the AMH signalling machinery is remarkably simple, consisting of one ligand and one type II receptor.


Subject(s)
Disorders of Sex Development/genetics , Glycoproteins , Growth Inhibitors/physiology , Mullerian Ducts/abnormalities , Point Mutation , Receptors, Peptide/genetics , Testicular Hormones/physiology , Alternative Splicing , Amino Acid Sequence , Anti-Mullerian Hormone , Base Sequence , Chromosome Mapping , Cloning, Molecular , Cryptorchidism/genetics , Humans , Infant , Male , Molecular Sequence Data , Organ Specificity , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptors, Transforming Growth Factor beta , Sequence Analysis, DNA , Syndrome , Testis/chemistry , Transcription, Genetic/genetics
11.
AIDS Res Hum Retroviruses ; 11(4): 517-25, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7632466

ABSTRACT

Since monoclonal antibodies (MAb) specific for CD4 are potent inhibitors of HIV and SIV replication in vitro, we explored their potential usefulness in vivo as an AIDS therapy. The anti-CD4 MAb 5A8 binds to domain 2 of the CD4 molecule and inhibits virus replication and virus-induced cell fusion at a postvirus binding step. Administration of this MAb to normal rhesus monkeys coats all circulating and lymph node CD4 cells and induces neither CD4 cell clearance nor measurable immunosuppression. In the present study, monkeys chronically infected with the simian immunodeficiency virus of macaques (SIVmac) had stable levels of SIVmac provirus in PBMC prior to treatment as measured by a quantitative polymerase chain reaction technique. Six infected monkeys treated with anti-CD4 MAb demonstrated a significant decrease in SIVmac provirus level after 9 days. Of these monkeys, 3 had > 800 CD4 cells/microliter and developed strong antimouse Ig responses that prevented further treatment. The remaining 3 monkeys had < 800 CD4 cell/microliter and failed to develop antimouse Ig antibody responses. When treatment was continued for 12-21 days in these monkeys, a sustained or further decrease in SIVmac provirus load occurred over the extended treatment period. Four monkeys that received a control MAb of irrelevant specificity for 9-22 days showed either no significant change or a transient increase in SIVmac provirus. Thus, the passive administration of anti-CD4 MAb may exert a specific antiviral effect in controlling immunodeficiency virus infection in vivo.


Subject(s)
Antibodies, Monoclonal/administration & dosage , CD4 Antigens/immunology , Proviruses/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Anti-Idiotypic/biosynthesis , Base Sequence , CD4 Lymphocyte Count , DNA Primers/genetics , DNA, Viral/analysis , DNA, Viral/genetics , HIV Infections/therapy , HIV Infections/virology , Humans , Immunotherapy, Adoptive , Macaca mulatta , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Proviruses/genetics , Proviruses/physiology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Virus Replication/immunology
12.
Cell ; 79(3): 415-25, 1994 Nov 04.
Article in English | MEDLINE | ID: mdl-7954809

ABSTRACT

To investigate the role of Müllerian-inhibiting substance (MIS) in mammalian sexual development, we generated MIS-deficient mice. Although MIS-deficient males had testes that were fully descended and produced functional sperm, they also developed female reproductive organs, which interfered with sperm transfer into females, rendering most of these males infertile. Their testes had Leydig cell hyperplasia and, in one instance, neoplasia. The actions of the two primary hormones of male sexual differentiation were genetically eliminated using the testicular feminization (Tfm) mutation in combination with the MIS mutant allele. XY Tfm/MIS double mutants developed as females, with a uterus, coiled oviducts, and no male reproductive organs except undescended dysfunctional testes. These results suggest that eliminating the presumptive female reproductive tract in male fetuses facilitates fertility and that in testes MIS is a negative regulator of Leydig cell proliferation. Eliminating the presumptive male reproductive tract is necessary for proper oviductal morphogenesis during female mouse development.


Subject(s)
Genitalia, Male/embryology , Glycoproteins , Growth Inhibitors/physiology , Mullerian Ducts/embryology , Sex Differentiation/physiology , Testicular Hormones/physiology , Androgen-Insensitivity Syndrome/embryology , Animals , Anti-Mullerian Hormone , Cell Division , Disorders of Sex Development/genetics , Disorders of Sex Development/pathology , Female , Fertility/physiology , Genitalia, Male/anatomy & histology , Growth Inhibitors/genetics , Leydig Cells/physiology , Male , Mice , Mice, Knockout , Morphogenesis , Oviducts/growth & development , Testicular Hormones/genetics , Testis/pathology , Wolffian Ducts/embryology
13.
J Clin Endocrinol Metab ; 79(4): 960-4, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7962305

ABSTRACT

Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance or factor, is secreted in high amounts by the immature Sertoli cell; it is negatively regulated by testosterone at puberty. In the present study, we measured serum AMH in 20 patients with defects of androgen synthesis or action: 9 with complete androgen insensitivity syndrome, 9 with a partial form, 1 patient with 3 beta-hydroxysteroid dehydrogenase deficiency, and 1 with Leydig cell agenesis. AMH was also determined in 15 control patients with idiopathic male pseudohermaphroditism. The serum AMH concentration was elevated in all testosterone-insensitive or -deficient patients compared with control levels during the first year of life. From 1 yr of age to the onset of puberty, serum AMH levels in patients with androgen insensitivity returned to normal values, but after pubertal development began, AMH levels again rose to extremely high levels in the complete androgen insensitivity syndrome. These results suggest that AMH is negatively regulated by testosterone not only at puberty, but also during the postnatal period. An elevation of serum AMH appears to be an interesting marker of androgen resistance or defect of androgen production in sexually ambiguous male infants.


Subject(s)
Androgens/physiology , Glycoproteins , Growth Inhibitors/blood , Testicular Hormones/blood , Adolescent , Aging/blood , Anti-Mullerian Hormone , Child , Child, Preschool , Disorders of Sex Development/blood , Drug Resistance , Humans , Infant , Male , Osmolar Concentration , Reference Values , Testosterone/physiology
14.
J Clin Endocrinol Metab ; 77(5): 1220-6, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8077315

ABSTRACT

Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance or factor, is produced by Sertoli cells from fetal life until puberty. In the present study, AMH, testosterone (T), LH, and FSH were measured by immunochemical methods in the serum of 50 boys with normal or delayed pubertal development, 4 patients with suspected androgen insensitivity, and 11 patients with either central (CPP) or gonadotropin-independent (GIPP) precocious puberty to investigate the hormonal regulatory mechanisms of AMH secretion at puberty. An inverse relationship between AMH and T levels was demonstrated. In boys with normal or delayed puberty with T concentrations below 6.7 nmol/L, AMH values were elevated (mean +/- SEM, 22.4 +/- 3.1 micrograms/L) and widely dispersed. In subjects with T levels over 6.7 nmol/L, AMH levels were uniformly low (3.4 +/- 0.5 micrograms/L), except in patients with suspected androgen insensitivity. No significant relationship was found between AMH and gonadotropin levels. Similar results were obtained in patients with either CPP or GIPP. Longitudinal studies were performed on four boys with CPP and two with GIPP before and after treatment. At the time of diagnosis, the T concentration was high, and AMH levels were usually low in CPP and GIPP patients alike. When appropriate treatment was initiated, the T concentration was normalized within 2-4 weeks, but restoration of prepubertal AMH levels required several months. Mature Sertoli cells were observed in testicular biopsies performed in three patients with untreated GIPP. Our results suggest that gonadotropins are not directly implicated in repression of AMH synthesis at puberty, but, rather, that the decrease in AMH production is the consequence of an androgen-mediated, long term, reversible chain of events leading to morphological and functional maturation of the Sertoli cells. Thus, the fall in serum AMH levels appears to be an excellent marker of Sertoli cell pubertal development.


Subject(s)
Glycoproteins , Growth Inhibitors/blood , Puberty, Precocious/blood , Testicular Hormones/blood , Adolescent , Androgens/physiology , Anti-Mullerian Hormone , Child , Drug Resistance , Follicle Stimulating Hormone/blood , Humans , Luteinizing Hormone/blood , Male , Mullerian Ducts/physiology , Puberty, Delayed/blood , Reference Values , Testosterone/blood
15.
Mol Endocrinol ; 7(2): 247-57, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8469238

ABSTRACT

Mullerian inhibiting substance (MIS)/anti-Mullerian hormone is a differentiation factor that causes regression of the Mullerian duct in the developing male fetus and an apparent sex reversal of the fetal ovary when inappropriately exposed to it. The purified product is a 140-kilodalton glycoprotein composed of two identical subunits. We show that a C-terminal fragment of MIS, which shares homology with transforming growth factor-beta, causes regression of the Mullerian duct and inhibits the biosynthesis of aromatase in the fetal ovary. However, both activities are enhanced dramatically by addition of the N-terminal portion of MIS. Under conditions where potentiation occurs, the N- and C-terminal domains of MIS reassociate. These results indicate that the N-terminus of MIS, unlike that of the other members of the transforming growth factor-beta family, plays a role in maintaining the biological activity of the C-terminus.


Subject(s)
Glycoproteins , Growth Inhibitors/chemistry , Mullerian Ducts/drug effects , Multigene Family , Peptide Fragments/pharmacology , Testicular Hormones/chemistry , Transforming Growth Factor beta , Animals , Anti-Mullerian Hormone , Antibodies, Monoclonal/immunology , Aromatase/biosynthesis , CHO Cells , Cattle , Cricetinae , Enzyme Induction/drug effects , Female , Fibrinolysin/metabolism , Growth Inhibitors/immunology , Growth Inhibitors/pharmacology , Humans , Ovary/embryology , Ovary/enzymology , Protein Folding , Rats , Recombinant Proteins/pharmacology , Testicular Hormones/immunology , Testicular Hormones/pharmacology
17.
Methods Enzymol ; 217: 398-414, 1993.
Article in English | MEDLINE | ID: mdl-8474341

ABSTRACT

The chemiluminescent detection methods described in this chapter have been successfully applied to the detection of plasmid DNA and genomic DNA in Southern and sequencing protocols. The high sensitivity and the simplicity of AMPPD are instrumental in making the chemiluminescent detection of DNA successful in hybridization assays. This detection technique has also been used to detect DNA in dot blots and in situ hybridization experiments as well as proteins in enzyme-linked immunosorbent assays (ELISAs) and Western blots.


Subject(s)
DNA, Recombinant/analysis , DNA/analysis , Oligonucleotide Probes , Adamantane/analogs & derivatives , Alkaline Phosphatase , Autoradiography/methods , Base Sequence , Blotting, Southern/methods , DNA/genetics , Electrophoresis, Polyacrylamide Gel/methods , In Situ Hybridization , Indicators and Reagents , Luminescent Measurements , Molecular Sequence Data , Nucleic Acid Hybridization , Phosphorus Radioisotopes , Plasmids , Proteins/analysis
18.
Mol Endocrinol ; 6(9): 1403-11, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1331774

ABSTRACT

Mullerian inhibiting substance (MIS) is a glycoprotein hormone expressed by Sertoli cells that induces the regression of Mullerian ducts during development of the male reproductive tract. Transgenic mice carrying a fusion gene composed of human MIS transcriptional regulatory sequences linked to the SV40 T-antigen gene specifically develop testicular tumors composed of a cell type histologically resembling the Sertoli cell. The lack of pathology at other sites suggests tissue-restricted expression of the transgene. A cell line derived from one of the testicular tumors has been established that continues to express markers associated with Sertoli cells, such as transferrin, sulfated glycoprotein-2, and inhibin-beta B. The cell line does not express detectable levels of inhibin-alpha, MIS, or FSH receptor. However, the cells have retained forskolin responsiveness. As adult Sertoli cells cannot be propagated in vitro, the availability of an immortal cell line displaying features characteristic of normal Sertoli cells should aid in subsequent analyses of the biology of this cell type.


Subject(s)
Antigens, Polyomavirus Transforming/biosynthesis , Genes, Synthetic , Glycoproteins , Growth Inhibitors/genetics , Oncogenes , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Sertoli Cells/metabolism , Testicular Hormones/genetics , Testicular Neoplasms/genetics , Animals , Anti-Mullerian Hormone , Antigens, Polyomavirus Transforming/genetics , Cell Line, Transformed , Colforsin/pharmacology , Gene Expression Regulation/drug effects , Humans , Male , Mice , Mice, Transgenic/metabolism , Organ Specificity , Recombinant Fusion Proteins/genetics , Simian virus 40/genetics , Testicular Neoplasms/pathology
19.
Genomics ; 12(4): 665-9, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1572639

ABSTRACT

Mullerian inhibiting substance (MIS), a testicular glycoprotein also known as anti-Mullerian hormone, plays a key role in male sexual development by causing regression of the Mullerian duct, the anlagen of the uterus, the Fallopian tubes, and part of the vagina. MIS is also expressed in the postnatal ovary, but its precise function is still not known. We report here the complete nucleotide sequence of the rat MIS gene. Rat MIS is encoded in five exons and is synthesized as a precursor of 553 amino acids, containing a 24-amino-acid leader. Based on homology with human MIS, we predict that the rat protein undergoes proteolytic processing at a site 108 amino acids from the C-terminus. Expression of the rat MIS mRNA is high in the 1-day-postnatal testis and decreases to a low level in the adult testis. In contrast, expression is not detected in the 1-day ovary, but increases to an intermediate level in the adult ovary. The rat gene should provide a good model for studying transcriptional regulation of MIS in the testis and ovary.


Subject(s)
Glycoproteins , Growth Inhibitors/genetics , Testicular Hormones/genetics , Animals , Anti-Mullerian Hormone , Base Sequence , Cattle , DNA/genetics , Female , Gene Expression Regulation , Humans , Male , Molecular Sequence Data , Mullerian Ducts/embryology , Ovary/growth & development , Ovary/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Sequence Homology, Nucleic Acid , Sex Differentiation/genetics , Testis/growth & development , Testis/metabolism
20.
Development ; 114(3): 721-7, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1319894

ABSTRACT

Anti-Müllerian hormone (AMH), also known as Müllerian-inhibiting substance or factor, has previously been shown to sex-reverse the steroidogenic pattern of fetal mammalian ovaries through repression of aromatase biosynthesis. Study of the ontogeny of the response of cyclic AMP-stimulated aromatase activity of rat fetal ovaries to AMH has allowed us to develop a quantitative bioassay for the hormone. Linear responses as a function of the logarithm of AMH concentration were observed over ranges of 0.2-7.5 micrograms/ml for the bovine protein and 0.15-2 micrograms/ml for the human protein, with a maximal decrease in aromatase activity of 90% for both proteins. Under the same in vitro conditions, AMH treatment did not affect cyclic AMP-stimulated fetal rat testicular aromatase activity. Partially purified chick AMH also decreased rat ovarian aromatase activity, allowing us to use this test to study AMH ontogeny in chick gonads. Analysis of the species specificity of AMH repression of ovarian aromatase activity indicated that turtle and rat fetal ovaries responded to AMH of other vertebrate classes, whereas aromatase activity of chick embryo ovaries could be repressed only by the homospecific hormone.


Subject(s)
Aromatase/metabolism , Glycoproteins , Growth Inhibitors/analysis , Ovary/enzymology , Testicular Hormones/analysis , Animals , Anti-Mullerian Hormone , Biological Assay/methods , Cattle , Cyclic AMP/metabolism , Depression, Chemical , Electrophoresis, Polyacrylamide Gel , Female , Growth Inhibitors/pharmacology , Humans , Male , Mullerian Ducts/anatomy & histology , Organ Culture Techniques , Ovary/drug effects , Rats , Rats, Inbred Strains , Species Specificity , Testicular Hormones/pharmacology , Testis/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...