Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J UOEH ; 41(2): 249-257, 2019.
Article in Japanese | MEDLINE | ID: mdl-31292372

ABSTRACT

Here we report a case of a 12-year-old girl who was referred to our department because of marked short stature of more than -5 SD below the median. Although her growth failure began suddenly at 6 years of age, she never had an examination because she had no other symptoms. Brain MRI examination suggested a tumor in the suprasellar region, and endocrine examination revealed combined pituitery hormone deficiency due to the tumor. Before surgery, the supplementation with hydrocortisone and levothyroxine was initiated. The pathological diagnosis of the surgically removed tumor was xanthogranuloma. The pattern of her growth curve showed a growth failure with sudden onset, which is a typical pattern of short stature secondary to pituitary disfunction including growth hormone deficiency associated with brain tumors. This case suggests that growth failure could be the only symptom in pediatric cases with brain tumors. Improved awareness regarding the association of growth failure with brain tumors is needed for earlier diagnosis and treatment. Furthermore, the growth curves should be carefully evaluated in regular health examinations at school.


Subject(s)
Body Height , Failure to Thrive , Growth Disorders/etiology , Pituitary Diseases/complications , Xanthogranuloma, Juvenile/complications , Abnormalities, Multiple/etiology , Age Factors , Child , Early Diagnosis , Facies , Female , Growth Disorders/pathology , Growth Disorders/prevention & control , Humans , Hypothyroidism/etiology , Magnetic Resonance Imaging , Physical Examination , Pituitary Diseases/diagnostic imaging , Pituitary Diseases/pathology , Pituitary Diseases/surgery , Pituitary Hormones, Anterior/deficiency , Schools , Severity of Illness Index , Transcription Factor Pit-1/deficiency , Xanthogranuloma, Juvenile/diagnostic imaging , Xanthogranuloma, Juvenile/pathology , Xanthogranuloma, Juvenile/surgery
2.
Gynecol Endocrinol ; 34(1): 36-39, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28649890

ABSTRACT

Ghrelin, an endoggenous for the growth hormone secretagogue receptor, has been shown to participate in the regulation of energy homeostasis and pituitary hormone secretion. Obestatin, encoded by the same gene as ghrelin, is described as a physiological opponent of ghrelin. Ghrelin and obestatin are altered in polycystic ovary syndrome (PCOS), which is characterized by insulin resistance and pituitary hormone secretion disorder. The aim of this study was to evaluate ghrelin/obestatin imbalance in relation to insulin resistance and pituitary hormone in adolescence with PCOS. This restrospective case-control study included 33 adolescence with PCOS and 38 control adolescence. Ghrelin and obestatin concentrations in serum were determined by RIA, and the serum fasting glucose and Insulin were determined by the glucose oxidase color method and INS-EASIA. The serum LH and FSH were measured by highly specific hemiluminescence immunoassays. We found that the serum ghrelin levels and ghrelin/obestatin ratio were significant lower in PCOS group than in control group, and the serum obestatin levels were significant higher in PCOS group than in control group. The ghrelin/obestatin ratios were negatively correlation with LH/FSH ratio and insulin resistant index in PCOS group. The findings of this study suggest that ghrelin/obestatin imbalance may play a role in pathogenesis of adolescent PCOS.


Subject(s)
Ghrelin/blood , Polycystic Ovary Syndrome/blood , Abnormalities, Multiple , Adolescent , Blood Glucose/analysis , Case-Control Studies , Energy Metabolism , Facies , Fasting , Female , Follicle Stimulating Hormone/blood , Homeostasis , Humans , Hypothyroidism , Insulin/blood , Insulin Resistance , Luteinizing Hormone/blood , Pituitary Hormones, Anterior/deficiency , Pituitary Hormones, Anterior/metabolism , Polycystic Ovary Syndrome/physiopathology , Retrospective Studies , Transcription Factor Pit-1/deficiency , Transcription Factor Pit-1/metabolism
4.
Sci Rep ; 10(1): 3069, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32080237

ABSTRACT

Low blood phosphate (Pi) reduces muscle function in hypophosphatemic disorders. Which Pi transporters are required and whether hormonal changes due to hypophosphatemia contribute to muscle function is unknown. To address these questions we generated a series of conditional knockout mice lacking one or both house-keeping Pi transporters Pit1 and Pit2 in skeletal muscle (sm), using the postnatally expressed human skeletal actin-cre. Simultaneous conditional deletion of both transporters caused skeletal muscle atrophy, resulting in death by postnatal day P13. smPit1-/-, smPit2-/- and three allele mutants are fertile and have normal body weights, suggesting a high degree of redundance for the two transporters in skeletal muscle. However, these mice show a gene-dose dependent reduction in running activity also seen in another hypophosphatemic model (Hyp mice). In contrast to Hyp mice, grip strength is preserved. Further evaluation of the mechanism shows reduced ERK1/2 activation and stimulation of AMP kinase in skeletal muscle from smPit1-/-; smPit2-/- mice consistent with energy-stress. Similarly, C2C12 myoblasts show a reduced oxygen consumption rate mediated by Pi transport-dependent and ERK1/2-dependent metabolic Pi sensing pathways. In conclusion, we here show that Pit1 and Pit2 are essential for normal myofiber function and survival, insights which may improve management of hypophosphatemic myopathy.


Subject(s)
Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Transcription Factor Pit-1/metabolism , Alleles , Animals , Cell Line , Cell Survival , Electron Transport , Energy Metabolism , Hand Strength , Mice, Knockout , Models, Biological , Muscle Cells/metabolism , Necrosis , Oxygen Consumption , Phosphates/metabolism , Sodium-Phosphate Cotransporter Proteins, Type III/deficiency , Transcription Factor Pit-1/deficiency
5.
J Clin Res Pediatr Endocrinol ; 12(4): 444-449, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32248673

ABSTRACT

Pycnodysostosis is a rare autosomal recessive osteosclerotic bone disorder associated with short stature and multiple bony abnormalities. Growth hormone (GH) deficiency may contribute to short stature in about 50% of patients. Available literature has rarely reported other pituitary hormone deficiencies in pyknodysostosis. Though the management remains conservative, recombinant human GH (rhGH) has been tried in selected patients. Here we present a case of pycnodysostosis which was evaluated for associated co-morbidities and found to have multiple pituitary hormone deficiencies. A 7-year-old girl was referred to our centre for evaluation of short stature. On examination, she had frontal and occipital bossing, limited mouth opening, hyperdontia with multiple carries, short and stubby digits and short stature. Investigation revealed dense sclerotic bones with frontal and occipital bossing, non-fusion of sutures with obtuse mandibular angle, non-pneumatised sinuses, small 'J' shaped sella turcica, acro-osteolysis of digits and absent medullary cavities. Cathepsin-K gene mutation analysis confirmed the diagnosis of pycnodysostosis. She was screened for associated co-morbidities and was found to have concomitant GH deficiency. Treatment with rhGH brought about an increase of 1 standard deviation score in height over 2 years and also unmasked central hypothyroidism at three months necessitating thyroxine replacement.


Subject(s)
Abnormalities, Multiple/drug therapy , Human Growth Hormone/administration & dosage , Hypothyroidism/drug therapy , Pituitary Hormones, Anterior/deficiency , Pituitary Hormones/deficiency , Pycnodysostosis/drug therapy , Thyroxine/administration & dosage , Transcription Factor Pit-1/deficiency , Abnormalities, Multiple/pathology , Child , Facies , Female , Humans , Hypothyroidism/complications , Hypothyroidism/pathology , Prognosis , Pycnodysostosis/complications , Pycnodysostosis/pathology
6.
Eur J Med Genet ; 62(11): 103570, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30414530

ABSTRACT

We report a patient with congenital complex pituitary hormone deficiency (CPHD) with intestinal malrotation and anal atresia. We identified a de novo heterozygous mutation, c.664T > G (p.Cys222Gly), in the FOXA2 gene in this individual. This missense mutation had the potential to affect the DNA binding properties of the FOXA2 protein based on a protein structure prediction. Since a CPHD patient with another missense mutation and one other case with an entire gene deletion have also been reported, we speculated that a haploinsufficiency of the FOXA2 gene might be a genetic etiology for this disorder. Phenotypic similarities and differences among these three cases are also discussed.


Subject(s)
Abnormalities, Multiple/genetics , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Hepatocyte Nuclear Factor 3-beta/genetics , Hypothyroidism/genetics , Pituitary Hormones, Anterior/deficiency , Transcription Factor Pit-1/deficiency , Abnormalities, Multiple/physiopathology , Facies , Heterozygote , Humans , Hypothyroidism/physiopathology , Mutation, Missense/genetics , Pituitary Hormones, Anterior/genetics , Protein Conformation , Transcription Factor Pit-1/genetics
7.
Mol Endocrinol ; 20(6): 1378-90, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16556738

ABSTRACT

Mutations in the transcription factors PIT1 (pituitary transcription factor 1) and PROP1 (prophet of Pit1) lead to pituitary hormone deficiency and hypopituitarism in mice and humans. To determine the basis for this, we performed histological analysis of Pit1- and Prop1-deficient dwarf mouse pituitaries throughout fetal and postnatal development. Pit1-deficient mice first exhibit pituitary hypoplasia after birth, primarily caused by reduced cell proliferation, although there is some apoptosis. To determine whether altered development of the vascular system contributes to hypopituitarism, we examined vascularization from embryonic d 14.5 and throughout development. No obvious differences in vascularization are evident in developing Pit1-deficient pituitaries. In contrast, the Prop1-deficient mouse pituitaries are poorly vascularized and dysmorphic, with a striking elevation in apoptosis. At postnatal d 11, apoptosis-independent caspase-3 activation occurs in thyrotropes and somatotropes of normal but not mutant pituitaries. This suggests that Prop1 and/or Pit1 may be necessary for caspase-3 expression. These studies provide further insight as to the mechanisms of Prop1 and Pit1 action in mice.


Subject(s)
Cell Proliferation , Hypopituitarism/etiology , Pituitary Hormones/deficiency , Animals , Animals, Newborn , Apoptosis , Base Sequence , Caspase 3 , Caspases/metabolism , Cell Differentiation , DNA Primers/genetics , Disease Models, Animal , Enzyme Activation , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypopituitarism/genetics , Hypopituitarism/metabolism , Hypopituitarism/pathology , Mice , Mice, Mutant Strains , Pituitary Gland/blood supply , Pituitary Gland/embryology , Pituitary Gland/growth & development , Pituitary Gland/pathology , Pregnancy , Transcription Factor Pit-1/deficiency , Transcription Factor Pit-1/genetics , Vascular Endothelial Growth Factor A/genetics
8.
Int J Mol Med ; 35(2): 358-66, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25434367

ABSTRACT

Since the tissue of children with combined pituitary hormone deficiency (CPHD) is not readily accessible, a new focus in children with CPHD is the blood-based expression profiling of non-protein coding genes, such as microRNAs (miRNAs or miRs), which regulate gene expression by inhibiting the translation of mRNAs. In this study, to address this, we identified potential miRNA signatures for CPHD by comparing genome-wide miRNA expression profiles in the serum of children with CPHD vs. normal (healthy) controls. Human embryonic kidney 293T cells were transfected with miR-593 or miR-511 oligonucleotides. Potential target gene expression was validated by western blot analysis for proteins and by miR-593 or miR-511 reporter assay using PROP1 gene 3'-untranslated region (3'-UTR) reporter. The miR-593 and miR-511 levels in the serum of 103 children with CPHD were assessed using the reverse transcription-quantitative polymerase chain reaction (RT-qPCR) method. We found 23 upregulated and 19 downregulated miRNAs with abnormal expression in children with CPHD compared with the normal controls using miRNA microarray analysis and RT-qPCR. miR-593 and miR-511 targeted the 3'-UTR of the PROP1 gene and attenuated the expression of PROP1. The levels of miR-593 and miR-511 in the serum of children with CPHD were increased compared with those in the control subjects. According to Youden's index, the sensitivity was 82.54 and 84.86%, and the specificity was 98.15 and 91.36% for miR-593 and miR-511, respectively. The various levels of specific miRNAs, particularly miR-593 and miR-511 whose direct target is the PROP1 gene, may serve as a non-invasive diagnostic biomarkers for children with CPHD.


Subject(s)
Abnormalities, Multiple/metabolism , Gene Expression Regulation , Homeodomain Proteins/biosynthesis , Hypothyroidism/metabolism , MicroRNAs/metabolism , Pituitary Hormones, Anterior/deficiency , Transcription Factor Pit-1/deficiency , 3' Untranslated Regions , Abnormalities, Multiple/genetics , Adolescent , Biomarkers/metabolism , Child , Facies , Female , Genome-Wide Association Study , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Hypothyroidism/genetics , Male , MicroRNAs/genetics , Pituitary Hormones, Anterior/genetics , Pituitary Hormones, Anterior/metabolism , Transcription Factor Pit-1/genetics , Transcription Factor Pit-1/metabolism
9.
J Clin Invest ; 122(10): 3635-46, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22945632

ABSTRACT

Sex-determining region Y (SRY) box 2 (SOX2) haploinsufficiency causes a form of hypopituitarism in humans that is characterized by gonadotrophin deficiency known as hypogonadotrophic hypogonadism. Here, we conditionally deleted Sox2 in mice to investigate the pathogenesis of hypogonadotrophic hypogonadism. First, we found that absence of SOX2 in the developing Rathke pouch of conditional embryos led to severe anterior lobe hypoplasia with drastically reduced expression of the pituitary-specific transcription factor POU class 1 homeobox 1 (POU1F1) as well as severe disruption of somatotroph and thyrotroph differentiation. In contrast, corticotrophs, rostral-tip POU1F1-independent thyrotrophs, and, interestingly, lactotrophs and gonadotrophs were less affected. Second, we identified a requirement for SOX2 in normal proliferation of periluminal progenitors; in its absence, insufficient precursors were available to produce all cell lineages of the anterior pituitary. Differentiated cells derived from precursors exiting cell cycle at early stages, including corticotrophs, rostral-tip thyrotrophs, and gonadotrophs, were generated, while hormone-producing cells originating from late-born precursors, such as somatotrophs and POU1F1-dependent thyrotrophs, were severely reduced. Finally, we found that 2 previously characterized patients with SOX2 haploinsufficiency and associated hypogonadotrophic hypogonadism had a measurable response to gonadotropin-releasing hormone (GnRH) stimulation, suggesting that it is not the absence of gonadotroph differentiation, but rather the deficient hypothalamic stimulation of gonadotrophs, that underlies typical hypogonadotrophic hypogonadism.


Subject(s)
Hypogonadism/genetics , Hypothalamo-Hypophyseal System/physiology , SOXB1 Transcription Factors/physiology , Animals , Cell Differentiation , Cell Lineage , Female , Gene Expression Regulation, Developmental , Gonadotropin-Releasing Hormone/therapeutic use , Heterozygote , Homeodomain Proteins/genetics , Humans , Hypogonadism/drug therapy , Hypogonadism/physiopathology , Mice , Mice, Knockout , Organogenesis/genetics , Organogenesis/physiology , Pituitary Gland, Anterior/abnormalities , Pituitary Gland, Anterior/embryology , Pituitary Gland, Anterior/metabolism , Pituitary Gland, Anterior/pathology , Repressor Proteins/genetics , SOXB1 Transcription Factors/deficiency , SOXB1 Transcription Factors/genetics , Somatotrophs/pathology , Thyrotrophs/pathology , Transcription Factor Pit-1/deficiency
10.
Aging Cell ; 11(4): 668-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22577861

ABSTRACT

Previous work has shown that primary skin-derived fibroblasts from long-lived pituitary dwarf mutants resist the lethal effects of many forms of oxidative and nonoxidative stress. We hypothesized that increased autophagy may protect fibroblasts of Pit-1(dw/dw) (Snell dwarf) mice from multiple forms of stress. We found that dwarf-derived fibroblasts had higher levels of autophagy, using LC3 and p62 as markers, in response to amino acid deprivation, hydrogen peroxide, and paraquat. Fibroblasts from dwarf mice also showed diminished phosphorylation of mTOR, S6K, and 4EBP1, consistent with the higher levels of autophagy in these cells after stress. Similar results were also observed in fibroblasts from mutant mice lacking growth hormone receptor (GHRKO mice) after amino acid withdrawal. Our results suggested that increased autophagy, regulated by TOR-dependent processes, may contribute to stress resistance in fibroblasts from long-lived mutant mice.


Subject(s)
Autophagy/genetics , Longevity/genetics , Longevity/physiology , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autophagy/drug effects , Autophagy/physiology , Carrier Proteins/metabolism , Cell Cycle Proteins , Culture Media , Eukaryotic Initiation Factors , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Hydrogen Peroxide/toxicity , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Mice, Mutant Strains , Oxidative Stress , Paraquat/toxicity , Phosphoproteins/metabolism , Phosphorylation , Receptors, Somatotropin/deficiency , Receptors, Somatotropin/genetics , Receptors, Somatotropin/physiology , Ribosomal Protein S6 Kinases/metabolism , Transcription Factor Pit-1/deficiency , Transcription Factor Pit-1/genetics , Transcription Factor Pit-1/physiology
11.
J Clin Invest ; 121(1): 113-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21123951

ABSTRACT

The pituitary-specific transcriptional factor-1 (PIT-1, also known as POU1F1), is an essential factor for multiple hormone-secreting cell types. A genetic defect in the PIT-1 gene results in congenital growth hormone (GH), prolactin (PRL), and thyroid-stimulating hormone (TSH) deficiency. Here, we investigated 3 cases of adult-onset combined GH, PRL, and TSH deficiencies and found that the endocrinological phenotype in each was linked to autoimmunity directed against the PIT-1 protein. We detected anti-PIT-1 antibody along with various autoantibodies in the patients' sera. An ELISA-based screening revealed that this antibody was highly specific to the disease and absent in control subjects. Immunohistochemical analysis revealed that PIT-1-, GH-, PRL-, and TSH-positive cells were absent in the pituitary of patient 2, who also had a range of autoimmune endocrinopathies. These clinical manifestations were compatible with the definition of autoimmune polyendocrine syndrome (APS). However, the main manifestations of APS-I--hypoparathyroidism and Candida infection--were not observed and the pituitary abnormalities were obviously different from the hypophysitis associated with APS. These data suggest that these patients define a unique "anti-PIT-1 antibody syndrome," related to APS.


Subject(s)
Autoantibodies/blood , Human Growth Hormone/deficiency , Polyendocrinopathies, Autoimmune/immunology , Prolactin/deficiency , Thyrotropin/deficiency , Transcription Factor Pit-1/antagonists & inhibitors , Transcription Factor Pit-1/immunology , Adult , Aged , Antibody Specificity , Humans , Immunohistochemistry , Male , Pituitary Gland/metabolism , Polyendocrinopathies, Autoimmune/blood , Polyendocrinopathies, Autoimmune/metabolism , Transcription Factor Pit-1/deficiency
12.
Genetics ; 189(2): 665-73, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21840860

ABSTRACT

Thyroid hormone has pleiotropic effects on cochlear development, and genomic variation influences the severity of associated hearing deficits. DW/J-Pou1f1dw/dw mutant mice lack pituitary thyrotropin, which causes severe thyroid hormone deficiency and profound hearing impairment. To assess the genetic complexity of protective effects on hypothyroidism-induced hearing impairment, an F1 intercross was generated between DW/J-Pou1f1dw/+ carriers and an inbred strain with excellent hearing derived from Mus castaneus, CAST/EiJ. Approximately 24% of the (DW/J×CAST/EiJ) Pou1f1dw/dw F2 progeny had normal hearing. A genome scan revealed a locus on chromosome 2, named modifier of dw hearing, or Mdwh, that rescues hearing despite persistent hypothyroidism. This chromosomal region contains the modifier of tubby hearing 1 (Moth1) locus that encodes a protective allele of the microtubule-associated protein MTAP1A. DW/J-Pou1f1dw/+ carriers were crossed with the AKR strain, which also carries a protective allele of Mtap1a, and we found that AKR is not protective for hearing in the (DW/J×AKR) Pou1f1dw/dw F2 progeny. Thus, protective alleles of Mtap1a are not sufficient to rescue DW/J-Pou1f1dw/dw hearing. We expect that identification of protective modifiers will enhance our understanding of the mechanisms of hypothyroidism-induced hearing impairment.


Subject(s)
Genes, Modifier/genetics , Hearing Loss/genetics , Hypothyroidism/complications , Transcription Factor Pit-1/genetics , Alleles , Animals , Base Sequence , Chromosome Mapping , Chromosomes, Mammalian/genetics , Crosses, Genetic , Female , Genetic Predisposition to Disease/genetics , Hearing Loss/etiology , Male , Mice , Mice, Inbred AKR , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Mutant Strains , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Polymorphism, Single Nucleotide , Quantitative Trait Loci/genetics , Transcription Factor Pit-1/deficiency
13.
Pharm Res ; 24(12): 2239-48, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17929148

ABSTRACT

PURPOSE: To investigate the in vitro in vivo correlation of a sustained release formulation for human growth hormone (hGH) based on hydroxyethyl methacrylated dextran (dex-HEMA) microspheres in Pit-1 deficient Snell dwarf mice and in healthy human volunteers. MATERIALS AND METHODS: A hGH-loaded microsphere formulation was developed and tested in Snell dwarf mice (pharmacodynamic study) and in healthy human volunteers (pharmacokinetic study). RESULTS: Single subcutaneous administration of the microspheres in mice resulted in a good correlation between hGH released in vitro and in vivo effects for the hGH-loaded microsphere formulation similar to daily injected hGH indicating a retained bioactivity. Testing the microspheres in healthy volunteers showed an increase (over 7-8 days) in hGH serum concentrations (peak concentrations: 1-2.5 ng/ml). A good in vitro in vivo correlation was obtained between the measured and calculated (from in vitro release data) hGH serum concentrations. Moreover, an increased serum concentration of biomarkers (insulin-like growth factor-I (IGF-I), IGF binding protein-3 (IGFBP-3) was found again indicating that bioactive hGH was released from the microspheres. CONCLUSIONS: Good in vitro in vivo correlations were obtained for hGH-loaded dex-HEMA microspheres, which is an important advantage in predicting the effect of the controlled drug delivery product in a clinical situations.


Subject(s)
Dextrans/chemistry , Drug Carriers , Dwarfism/drug therapy , Human Growth Hormone/pharmacology , Methacrylates/chemistry , Microspheres , Aged , Animals , Biomarkers/blood , Body Size/drug effects , Body Weight/drug effects , Chemistry, Pharmaceutical , Delayed-Action Preparations , Disease Models, Animal , Drug Compounding , Dwarfism/genetics , Dwarfism/physiopathology , Human Growth Hormone/administration & dosage , Human Growth Hormone/blood , Human Growth Hormone/chemistry , Human Growth Hormone/pharmacokinetics , Humans , Injections, Subcutaneous , Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor Binding Proteins/blood , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Mutant Strains , Middle Aged , Models, Biological , Particle Size , Solubility , Transcription Factor Pit-1/deficiency , Transcription Factor Pit-1/genetics , Transcription Factor Pit-1/metabolism
14.
Horm Res ; 64(4): 175-9, 2005.
Article in English | MEDLINE | ID: mdl-16210857

ABSTRACT

Mutations of the PIT1/POU1F1 gene are responsible for a rare variant of anterior hypopituitarism, including deficiency of growth hormone, prolactin and thyrotropin. In 8 ethnically diverse POU1F1-deficient patients (4 different mutations) with normal circulating levels of cortisol and adrenocorticotropic hormone, and with spontaneous onset and progression of puberty, we observed an absence or delay of adrenarche (median circulating dehydroepiandrosterone-sulfate -6.2 SD); in each of the 4 postmenarcheal females, pubarche (i.e. appearance of pubic hair) was also absent or delayed. The absence/delay of adrenarche in POU1F1-deficient patients and the absence/delay of pubarche in POU1F1-deficient females suggest that a POU1F1-dependent factor contributes to the normal development of adrenarche and female pubarche.


Subject(s)
Adrenarche/physiology , Transcription Factor Pit-1/deficiency , Adolescent , Adrenocorticotropic Hormone/blood , Adult , Child , Dehydroepiandrosterone Sulfate/blood , Female , Humans , Hydrocortisone/blood , Hypopituitarism/etiology , Hypopituitarism/physiopathology , Male , Mutation , Puberty
SELECTION OF CITATIONS
SEARCH DETAIL