Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancer Res ; 58(6): 1159-64, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9515800

ABSTRACT

Insulin-like growth factor I action has been implicated in the pathogenesis of many different malignancies, including breast cancer. Insulin-like growth factor I receptors (IGF-IRs) are overexpressed in virtually all breast cancer cell lines, in which they are believed to enhance growth and inhibit apoptosis. In this study, the functional activity of IGF-IRs from normal and malignant human breast tissue was assessed. IGF-IR expression was 14-fold higher in malignant breast tissue than in normal breast tissue. IGF-IR autophosphorylation and kinase activity were 2-4-fold higher in purified receptor preparations from malignant breast tissue as compared to normal breast tissue when normalized for receptor number. This increase in receptor function, coupled with the enhanced receptor expression, amounts to a 40-fold elevation in IGF-IR tyrosine kinase activity in malignant breast tissue. The enhanced receptor autophosphorylation and kinase activity were observed in the absence of hormonal stimulation and seem to result from an alteration in the intrinsic activity of the receptor itself. Protein tyrosine phosphatase activity is also increased in malignant breast tissue. These data suggest that the IGF-IR is an important target for breast cancer therapy.


Subject(s)
Breast Neoplasms/enzymology , Breast/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Somatomedin/metabolism , Humans , Ligands , Phosphorylation , Phosphotyrosine/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptor, Insulin/metabolism
2.
Calcif Tissue Int ; 61(4): 345-7, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9312206

ABSTRACT

Calcitonin (CT) and other bone-active peptides have been restrained in clinical use by the need for parenteral administration. Although nasal and other transmucosal routes can be used for CT treatment, bioavailability and bioactivity of the peptide thus delivered are limited. We have evaluated the intrapulmonary route (IP) for the delivery of salmon calcitonin (SCT) in normal subjects. SCT was administered with a dry powder delivery inhaler. For comparison, each subject also received intramuscular (IM) SCT. Inhaled SCT produced significant hypocalcemia in all subjects as did injected SCT, and the peptide could be readily measured in serum by immunoassay. Compared by dose, IP SCT had 66% of the bioactivity and 28% of the bioavailability of IM SCT. This intrapulmonary route of administration should enhance the clinical acceptability of SCT and could also be applicable to other bone-active peptides.


Subject(s)
Administration, Inhalation , Analgesics/administration & dosage , Calcitonin/administration & dosage , Calcium/blood , Drug Delivery Systems , Adult , Aged , Aged, 80 and over , Analgesics/blood , Analgesics/pharmacology , Biological Availability , Calcitonin/blood , Calcitonin/pharmacology , Feasibility Studies , Humans , Injections, Intramuscular , Male , Middle Aged , Reference Values
3.
Biochem Mol Med ; 62(2): 139-50, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9441865

ABSTRACT

The Wilms tumor gene WT1 has been implicated in the early development of the kidney. Mutations in WT1 are found in a small fraction of Wilms tumor, a pediatric nephroblastoma, and Denys-Drash syndrome, characterized by genitourinary abnormalities. The WT1 gene product functions as a transcriptional repressor of growth factor-related genes. The kidney is one of the major sites of insulin action in vivo and expresses high levels of insulin receptors (IR). IR expression has been detected during early embryogenesis, suggesting that it may play a role in development. We investigated whether two WT1 splice variants lacking or including a three-amino-acid (KTS) insertion between the third and fourth zinc finger in the DNA-binding domain could repress the IR promoter in vitro. We show that the +KTS variant effectively represses promoter activity under all conditions tested but the -KTS variant was only able to repress in the presence of cotransfected C/EBP beta or a dominant-negative p53 mutation. Deletional mapping indicated that distinct regions of the IR promoter mediated the effects of the two isoforms and DNaseI footprint analysis identified potential WT1 binding sites within these regions.


Subject(s)
Alternative Splicing/genetics , DNA-Binding Proteins/genetics , Genes, Wilms Tumor/physiology , Promoter Regions, Genetic , Receptor, Insulin/genetics , Transcription Factors/genetics , Animals , Base Sequence , Binding Sites/genetics , Carcinoma, Hepatocellular , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/physiology , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Precipitin Tests , Rats , Sequence Deletion , Transcription Factors/biosynthesis , Transcription Factors/physiology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology , WT1 Proteins , Wilms Tumor/genetics , Zinc Fingers/genetics
4.
Diabetes ; 45(10): 1379-85, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8826975

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is a protein tyrosine phosphatase of unknown function, although increasing evidence supports a role for this phosphatase in insulin action. We have investigated the interaction of PTP1B with the insulin receptor using a PTP1B glutathione S-transferase (GST) fusion protein with a point mutation in the enzyme's catalytic domain. This fusion protein is catalytically inactive, but the phosphatase's phosphotyrosine binding site is maintained. The activated insulin receptor was precipitated from purified receptor preparations and whole-cell lysates by the inactive PTP1B-GST, demonstrating a direct association between the insulin receptor and PTP1B. A p120 of unknown identity was also precipitated from whole-cell lysates by the PTP1B fusion protein, but IRS-1 (pp185) was not. A catalytically inactive [35S]PTP1B-fusion protein bound directly to immobilized insulin receptor kinase domains and was displaced in a concentration-dependent manner. Finally, tyrosine-phosphorylated PTP1B was precipitated from whole-cell lysates by an anti-insulin receptor antibody after insulin stimulation. The site of interaction between PTP1B and the insulin receptor was studied using phosphopeptides modeled after the receptor's kinase domain, the NPXY domain, and the COOH-terminal. Each phosphopeptide inhibited the PTP1B-GST:insulin receptor interaction. Study of mutant insulin receptors demonstrated that activation of the kinase domain is necessary for the PTP1B:insulin receptor interaction, but receptors with deletion of the NPXY domain or of the COOH-terminal can still bind to the PTP1B-GST. We conclude that PTP1B can associate directly with the activated insulin receptor at multiple different phosphotyrosine sites and that dephosphorylation by PTP1B may play a significant role in insulin receptor signal transduction.


Subject(s)
Protein Tyrosine Phosphatases/metabolism , Receptor, Insulin/metabolism , Amino Acid Sequence , Animals , Antibodies , Binding Sites , Cell Line , Cloning, Molecular , Glutathione Transferase , Humans , Immunoblotting , Macromolecular Substances , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Phosphopeptides/chemistry , Phosphopeptides/isolation & purification , Point Mutation , Protein Tyrosine Phosphatases/chemistry , Rats , Receptor, Insulin/chemistry , Receptor, Insulin/isolation & purification , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Transfection
5.
Clin Chem ; 42(9): 1483-7, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8787709

ABSTRACT

We describe a patient with signs and symptoms of virilization caused by 21-hydroxylase deficiency. The patient, a Hispanic woman, first sought medical attention at age 24, when she presented to a medical clinic with an uncomplicated urinary tract infection. At that time several signs of virilization were noted and she was referred to the endocrinology clinic. Evaluation revealed temporal balding, hyperpigmentation, acne, absent breast development, a muscular habitus, and clitoromegaly. Radiological studies revealed bilaterally enlarged adrenal glands and ovaries. Laboratory evaluation revealed markedly increased concentrations of 17-hydroxyprogesterone, androstenedione, and testosterone. The patient was diagnosed with congenital adrenal hyperplasia (CAH) and received hormone therapy. In her sister, encouraged to undergo testing for this autosomal recessive disorder, HLA testing demonstrated that certain haplotypes in this family were associated with CAH. The case highlights key steps in the laboratory diagnosis and genetics of CAH.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Gonadal Steroid Hormones/blood , 17-alpha-Hydroxyprogesterone , Acne Vulgaris/complications , Adrenal Hyperplasia, Congenital/blood , Adult , Androstenedione/blood , Clitoris/abnormalities , Cortodoxone/blood , Dehydroepiandrosterone/blood , Female , Hirsutism/complications , Histocompatibility Testing , Humans , Hydroxyprogesterones/blood , Hyperpigmentation/complications , Testosterone/blood , Urinary Tract Infections/complications
6.
Cancer Res ; 56(12): 2781-8, 1996 Jun 15.
Article in English | MEDLINE | ID: mdl-8665514

ABSTRACT

There is strong evidence to suggest that insulin and insulin-like growth factor (IGF)-I may be important for tumor growth. Both the insulin and IGF-I receptors (IGF-IR) are overexpressed in breast cancer, and antibody blockade of the IGF-IR inhibits the growth of some breast cancer cell lines. Furthermore, expression of an insulin receptor (IR) in a normal mammary epithelia] cell line causes insulin-dependent transformation. Functional inactivation of p53 is also very frequent in many tumors. In this paper, we investigated whether inactivation of p53 might be involved in the overexpression of the IR in malignancy, specifically breast cancer. We demonstrate a positive correlation between IR and IGF-IR levels and p53 overexpression in primary human breast malignancies. To examine possible mechanisms by which p53 may regulate IR gene expression, we show that p53 can repress the IR promoter and that a dominant-negative p53 (248Q) can de-repress the promoter in cells containing normal p53. The p53 effect was shown to be mediated by C/EBP and Sp1 transcription factors. We also documented that p53-null mice had elevated levels of Sp1, but not C/EBPalpha, and that insulin binding to liver extracts was increased compared to wild-type controls. These results suggest that p53 inactivation may lead to an up-regulation of genes, such as the IR, that are dependent on these transcription factors.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Sp1 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , Binding, Competitive , CCAAT-Enhancer-Binding Proteins , Female , Genes, Reporter , Humans , Mice , Molecular Sequence Data , Receptor, Insulin/genetics , Transfection , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/physiology
7.
J Biol Chem ; 270(32): 19151-7, 1995 Aug 11.
Article in English | MEDLINE | ID: mdl-7642582

ABSTRACT

Potential signaling substrates for the insulin-like growth factor I (IGF-I) receptor are SH2 domain proteins including the p85 subunit of phosphatidylinositol 3-kinase, the tyrosine phosphatase Syp, GTPase activating protein (GAP), and phospholipase C-gamma (PLC-gamma). In this study, we demonstrate an association between the IGF-I receptor and p85, Syp, and GAP, but not with PLC-gamma in lysates of cells overexpressing the human IGF-I receptor. We further investigated these interactions using glutathione S-transferase (GST) fusion proteins containing the amino-terminal SH2 domains of p85 or GAP, or both SH2 domains of Syp or PLC-gamma to precipitate the IGF-I receptor from purified receptor preparations and from whole cell lysates. p85-, Syp-, and GAP-GSTs precipitated the IGF-I receptor, whereas the PLC-gamma-GST did not. Using phosphopeptides corresponding to IGF-I receptor phosphorylation sites, we determined that the p85- and Syp-GST association with the IGF-I receptor could be inhibited by a carboxyl-terminal peptide containing pY1316 and that the GAP-GST association could be inhibited by a NPXY domain peptide. The GAP-GST binding site was confirmed by showing that a mutant IGF-I receptor with a deletion of the NPXY domain including tyrosine 950 was poorly precipitated by the GAP-GST. We conclude that p85 and Syp may bind directly to the IGF-I receptor at tyrosine 1316, and that GAP may bind to the IGF-I receptor at and PLC-gamma was not evident. p85, Syp, and GAP are potential modulators of IGF-I receptor signal transduction.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Tyrosine Phosphatases/metabolism , Proteins/metabolism , Receptor, IGF Type 1/metabolism , Amino Acid Sequence , Animals , Arsenicals/pharmacology , Binding Sites , CHO Cells , Cricetinae , GTPase-Activating Proteins , Insulin-Like Growth Factor I/pharmacology , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Phosphatidylinositol 3-Kinases , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , SH2 Domain-Containing Protein Tyrosine Phosphatases , Type C Phospholipases/metabolism , Tyrosine/metabolism
8.
Endocrinology ; 136(4): 1635-41, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7895674

ABSTRACT

Insulin/insulin-like growth factor-I (IGF-I) hybrid receptors are composed of an alpha beta-heterodimer from an insulin receptor and an alpha beta-heterodimer from an IGF-I receptor. In this study, we evaluate the effect of insulin receptor overexpression on hybrid formation. The more human insulin receptors expressed in rodent fibroblasts, the greater the percentage of endogenous rat IGF-I receptors that form hybrid receptors. The IGF-I receptor in rodent fibroblasts has two receptor isoforms, one with a 95-kilodalton (kDa) beta-subunit and one with an 105 kDa beta-subunit. A truncated mutant insulin receptor was used to demonstrate that only activated IGF-I receptors with the 105-kDa beta-subunit form hybrid receptors with the insulin receptor. Insulin/IGF-I hybrid receptors with a kinase-defective insulin heterodimer undergo trans and a small amount of cis autophosphorylation, but overall autophosphorylation is markedly decreased from that seen in hybrids with a kinase-competent insulin receptor. The kinase-defective insulin receptor heterodimer functions as a dominant-negative, inhibiting phosphorylation by the kinase-competent IGF-I receptor heterodimer. The kinase-defective hybrid receptors are, however, able to undergo internalization. Despite an increasing percentage of insulin/IGF-I hybrid receptors in the three cell lines studied, the rates of IGF-I internalization and degradation remain similar to those mediated by the IGF-I receptor and distinct from those of insulin receptor heterotetramers. In conclusion, IGF-I-stimulated insulin/IGF-I hybrid receptors function like IGF-I receptors, rather than like insulin receptors.


Subject(s)
Protein Multimerization , Receptor, IGF Type 1/physiology , Receptor, Insulin/physiology , Animals , Binding, Competitive , Fibroblasts/metabolism , Humans , Immunosorbent Techniques , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Kinetics , Macromolecular Substances , Phosphorylation , Rats , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/genetics , Receptor, Insulin/chemistry , Receptor, Insulin/genetics , Transfection
9.
J Biol Chem ; 269(44): 27186-92, 1994 Nov 04.
Article in English | MEDLINE | ID: mdl-7525547

ABSTRACT

The insulin receptor is known to interact with the SH2 domain proteins p85 (the regulatory subunit of phosphatidylinositol 3-kinase), Syp (a tyrosine phosphatase), and GAP (GTPase-activating protein). In this study, we mapped the insulin receptor binding sites for each of these proteins by examining the ability of phosphopeptides, corresponding to insulin receptor phosphorylation sites, and mutant insulin receptors to inhibit an insulin receptor-SH2 domain interaction. Precipitation of partially purified insulin receptors by glutathione S-transferase fusion proteins containing the N-terminal SH2 domains of p85 and GAP and both SH2 domains of Syp was demonstrated. The effect of the addition of each phosphopeptide on insulin receptor precipitation was tested. pY1322, the C-terminal insulin receptor peptide, inhibited insulin receptor precipitation by both p85- and Syp-GST. The NPXY internalization domain peptide inhibited insulin receptor precipitation by GAP-GST. These data were confirmed by mutant insulin receptor experiments. The insulin receptor C-terminal mutants, delta CT and Y/F2, were not precipitated by p85- or Syp-GST and the NPXY mutant insulin receptors, delta Ex16 and HI delta NPEY, were not precipitated by GAP-GST. Therefore, we conclude that p85 and Syp bind to the insulin receptor C terminus at tyrosine 1322 and GAP binds to the insulin receptor NPXY domain at tyrosine 960.


Subject(s)
Phosphopeptides/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Tyrosine Phosphatases/metabolism , Proteins/metabolism , Receptor, Insulin/metabolism , Amino Acid Sequence , Animals , Binding Sites , GTPase-Activating Proteins , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Phosphatidylinositol 3-Kinases , Phosphotyrosine , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Rats , SH2 Domain-Containing Protein Tyrosine Phosphatases , Tyrosine/analogs & derivatives , Tyrosine/metabolism
10.
Mol Cell Biol ; 14(11): 7466-75, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7935461

ABSTRACT

We have investigated the functional role of the SH2 domain of the 85-kDa subunit (p85) of the phosphatidylinositol 3-kinase in the insulin signal transduction pathway. Microinjection of a bacterial fusion protein containing the N-terminal SH2 domain of p85 inhibited insulin- and other growth factor-induced DNA synthesis by 90% and c-fos protein expression by 80% in insulin-responsive rat fibroblasts. The specificity of the fusion protein was examined by in vitro precipitation experiments, which showed that the SH2 domain of p85 can independently associate with both insulin receptor substrate 1 and the insulin receptor itself in the absence of detectable binding to other phosphoproteins. The microinjection results were confirmed through the use of an affinity-purified antibody directed against p85, which gave the same phenotype. Additional studies were carried out in another cell line expressing mutant insulin receptors which lack the cytoplasmic tyrosine residues with which p85 interacts. Microinjection of the SH2 domain fusion protein also inhibited insulin signaling in these cells, suggesting that association of p85 with insulin receptor substrate 1 is a key element in insulin-mediated cell cycle progression. In addition, coinjection of purified p21ras protein with the p85 fusion protein or the antibody restored DNA synthesis, suggesting that ras function is either downstream or independent of p85 SH2 domain interaction.


Subject(s)
DNA/biosynthesis , Genes, fos/drug effects , Insulin/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Animals , Antibodies/administration & dosage , Cell Line , Humans , Insulin Receptor Substrate Proteins , Microinjections , Molecular Weight , Phosphatidylinositol 3-Kinases , Phosphoproteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/administration & dosage , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Protein Conformation , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins p21(ras)/administration & dosage , Rats , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/administration & dosage , Signal Transduction/drug effects
12.
Invest Ophthalmol Vis Sci ; 34(6): 2105-11, 1993 May.
Article in English | MEDLINE | ID: mdl-8491561

ABSTRACT

PURPOSE: To study the growth regulation of bovine corneal endothelium, the effects of insulin and insulin-like growth factor-I on the expression of the c-fos proto-oncogene as well as on DNA synthesis were analyzed. Bovine corneal endothelial cells were also analyzed for the presence of insulin and insulin-like growth factor-I receptors. METHODS: Indirect immunofluorescence was used to assess the effect of stimulation by insulin and insulin-like growth factor-I on c-fos protein expression and DNA synthesis quiescent bovine corneal endothelium. Receptor number was determined by 125I-insulin or 125I-IGF-I binding studies. RESULTS: Fetal bovine serum strongly stimulated c-fos protein expression and DNA synthesis. Insulin-like growth factor-I was less effective while insulin was effective only at high concentrations. Scatchard analysis of 125I-insulin-like growth factor-I and 125I-insulin binding to bovine corneal endothelium revealed 180,000 IGF-I receptors and 7,000 insulin receptors. CONCLUSION: The number of insulin-like growth factor-I receptors far exceeds the number of insulin receptors in bovine corneal endothelium, suggesting that the effects of insulin on c-fos gene expression and mitogenesis were likely to be mediated through the insulin-like growth factor-I receptor.


Subject(s)
DNA/biosynthesis , Endothelium, Corneal/metabolism , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Animals , Cattle , Cells, Cultured , DNA Replication , Fluorescent Antibody Technique , Radioligand Assay , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism
13.
J Biol Chem ; 266(26): 17522-30, 1991 Sep 15.
Article in English | MEDLINE | ID: mdl-1910046

ABSTRACT

Biologically active colloid-gold complexes were used to compare ligand-induced microaggregation, redistribution, and internalization of insulin receptors on Rat 1 fibroblasts expressing wild type (HIRc) or tyrosine kinase-defective (HIR A/K1018) human insulin receptors. Insulin-like growth factor I (IGF I) and alpha 2-macroglobulin receptors also were compared. On both cell types, all four unoccupied receptor types occurred predominantly as single receptors. Ligand binding caused receptor microaggregation. Microaggregation of wild type or kinase-defective insulin receptors or IGF I receptors was not different. alpha 2-Macroglobulin receptors formed larger microaggregates. Compared to wild type insulin or IGF I receptors, accumulation of kinase-defective insulin receptor microaggregates in endocytic structures was decreased, and the size of microaggregates in coated pits was significantly smaller. As a result, receptor-mediated internalization of gold-insulin by HIR A/K1018 cells was less than 6% of the cell-associated particles compared to approximately 60% of the particles in HIRc cells. On HIR A/K1018 cells, alpha 2-macroglobulin and IGF I were internalized via coated pits demonstrating that those structures were functional. These results suggest that: 1) ATP binding, receptor autophosphorylation, and activation of receptor kinase activity are not required for receptor microaggregation; 2) receptor microaggregation per se is not sufficient to cause ligand-induced receptor-mediated internalization or the biological effects of insulin; and 3) autophosphorylation of the beta-subunit or activation of the receptor kinase activity is required for the insulin-induced concentration of occupied receptors in coated pits.


Subject(s)
Coated Pits, Cell-Membrane/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor Aggregation , Receptor, Insulin/metabolism , Animals , Cells, Cultured , Humans , Ligands , Microscopy, Electron , Rats , Receptor, Insulin/ultrastructure , Temperature
14.
Am J Hum Genet ; 36(4): 899-903, 1984 Jul.
Article in English | MEDLINE | ID: mdl-6475960

ABSTRACT

Regional chromosomal mosaicism has been found in tissue from the clitoral mass of an infant presenting with ambiguous genitalia. Chromosome analysis of skin from the clitoral mass was interpreted as 46,XX/52,XX, +2, +7, +8, +12, +13, +20, whereas study of ovarian tissue and peripheral lymphocytes found each to have normal 46,XX karyotypes with no indication of mosaicism. We believe that somatic cell mutation led to a hyperdiploid clonal cell line resulting in maldevelopment of this patient's external genitalia. This observation, which to our knowledge has not been previously reported, indicates that localized chromosomal abnormalities in some cases may be etiologic of isolated congenital malformations.


Subject(s)
Clitoris/abnormalities , Gonadal Dysgenesis/genetics , Mosaicism , Female , Humans , Infant , Karyotyping , Ploidies
SELECTION OF CITATIONS
SEARCH DETAIL
...