17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Central precocious puberty in a boy with pseudohypoparathyroidism type Ia due to a novel GNAS mutation

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction Pseudohypoparathyroidism (PHP) is characterized by hypocalcemia, hyperphosphatemia, and elevated PTH levels owing to the resistance of target tissue to PTH. PHP Ia is associated with the phenotype of Albright hereditary osteodystrophy, including short stature, obesity, round face, and brachydactyly. PHP Ia is caused by an inactivating mutation in the GNAS gene on the maternal allele, which encodes the α-subunit of the stimulatory guanine nucleotide-binding protein (1). Pubertal development in male patients with PHP Ia has not been fully elucidated. Among female patients with PHP Ia, some have been observed to show delayed puberty due to resistance to LH and FSH (2). Although two boys with PHP Ia and peripheral precocious puberty have been reported (3), no reports have described PHP Ia with central precocious puberty. Here, we report a boy with central precocious puberty and PHP Ia due to a novel GNAS mutation. Case Report The proband was an 11-yr-old boy, who was the first child of healthy Japanese parents. He was born after an uneventful pregnancy and delivery. Neither significant hypocalcemia nor precocious puberty were reported in first-degree relatives. At 10 yr of age, he had afebrile convulsions for 5 min due to hypocalcemia (5.3 mg/dL, reference: 8.2–10.2 mg/dL). Hyperphosphatemia (11.1 mg/dL, reference: 2.5–4.6 mg/dL) and elevated serum intact PTH levels (363 pg/mL, reference: 10–65 pg/mL) were also detected. The serum 25-hydroxyvitamin D level was 16.2 ng/mL (reference: ≥ 20.0 ng/mL). His height was 138 cm (+ 0.2 SD), weight was 38.5 kg (+ 0.7 SD), face was rounded, and the fourth and fifth metacarpals were shortened. The Wechsler Intelligence Scale for Children-IV showed a Full Scale Intelligence Quotient of 61, indicating intellectual disability. We clinically diagnosed the patient as having PHP Ia. On admission, at 10 yr and 1 mo of age, voice breaking was noticed. The right and left testicular volumes were 8 and 6 mL, respectively. The stretched penile length was 9.5 cm, and his genital hair development was characterized as Tanner stage II. A growth spurt occurred at 9 yr of age, and his bone age was assessed to be 13 yr, indicating accelerated growth and maturation. Testosterone (2.5 ng/mL, reference for adults: 1.42–9.23 ng/mL) and stimulated gonadotropin levels (LH: 18.0 mIU/mL; FSH: 7.0 mIU/mL) after administration of GnRH were consistent with central precocious puberty. No abnormal findings, except calcifications in the basal ganglia, were detected using magnetic resonance imaging of the head. We started treatment with sc injection of leuprorelin to suppress pubertal progression. Leuprorelin treatment suppressed testosterone (0.074 ng/mL) and gonadotropin (LH: 0.6 mIU/mL; FSH: 0.4 mIU/mL) levels close to a prepubertal range. Mutation Analysis After receiving approval from the institutional review board of Yokohama Municipal Citizen’s Hospital and obtaining informed consent from the proband’s mother, we extracted genomic DNA from the peripheral blood samples of the proband. We amplified exons 2–13 and their splice sites in the GNAS gene and performed direct sequencing. A heterozygous frameshift variant, c.568dupT, p.Tyr190Leufs*20, in the GNAS gene (NM_000516; Fig. 1 Fig. 1. Partial sequence of exon 7 in the GNAS gene. This figure shows a chromatogram of the proband who had a heterozygous mutation, c.568dupT, p.Tyr190Leufs*20, denoted by an arrow. ) was identified. This variant was not found in the Human Genetic Variation Database or the Exome Aggregation Consortium Database. Discussion We identified a novel GNAS frameshift mutation in a boy clinically diagnosed as having PHP Ia with central precocious puberty. To the best of our knowledge, this is the first report of PHP Ia with central precocious puberty. The association between inactive GNAS mutation and central precocious puberty is unclear. GNAS is expressed in many cell types but has not been reported to be involved in the gonadotropin inhibitory system. Signal transduction of the gonadotropin inhibitory hormone receptor was not regulated by the stimulatory G protein, but by the inhibitory G protein (4). Precocious puberty has not been reported in cases of nearby frameshift mutations, such as c.565_568delGACT (5). It is unlikely that such truncated GNAS proteins preserving approximately 190 amino acids in the amino terminus are constitutively active even in a tissue-specific manner. This case showed that central precocious puberty could occur in patients with PHP Ia, who show less resistance to gonadotropin when gonadotropin secretion is accelerated early. However, the specific cause for this occurrence in these patients is still unclear. In this case, central precocious puberty secondary to peripheral precocious puberty is highly improbable for the following two reasons. First, the testosterone and gonadotropin levels were suppressed following leuprorelin treatment. Second, it is unlikely that the function of the truncated GNAS protein in our patient would depend on temperature. Peripheral precocious puberty was only reported in a case of PHP Ia with the mutant GNAS protein, p.Ala366Ser, that showed both gain and loss of functions in a temperature-dependent manner (3). Thus, patients with PHP Ia can develop not only peripheral precocious puberty with a mutation-specific cause but also central precocious puberty with other causes. In summary, we report a boy with central precocious puberty who had PHP Ia due to a novel GNAS mutation. Conflict of Interests Tomonobu Hasegawa has the following financial relationships to disclose: Research funding from Novo Nordisk Pharma Ltd. and JCR Pharmaceuticals Co., Ltd.

          Related collections

          Most cited references4

          • Record: found
          • Abstract: found
          • Article: not found

          Rapid GDP release from Gs alpha in patients with gain and loss of endocrine function.

          Luteinizing hormone stimulates testicular Leydig cells to produce testosterone by binding to a receptor that activates the G protein Gs and adenylyl cyclase. Testotoxicosis is a form of precocious puberty in which the Leydig cells secrete testosterone in the absence of luteinizing hormone, often due to constitutive activation of the luteinizing hormone receptor and (indirectly) Gs (refs 1-4). Here we study two unrelated boys suffering from a paradoxical combination of testotoxicosis and pseudohypoparathyroidism type Ia (PHP-Ia), a condition marked by resistance to hormones acting through cyclic AMP (parathyroid hormone and thyroid-stimulating hormone) as well as a 50% decrease in erythrocyte Gs activity (the remaining 50% is due to the normal Gs allele). In both patients, a mutation in the gene encoding the Gs alpha-subunit replace alanine at position 366 with serine. We show that this alpha s-A366S mutation constitutively activates adenylyl cyclase in vitro, causing hormone-independent cAMP accumulation when expressed in cultured cells, and accounting for the testotoxicosis phenotype (as cAMP stimulates testosterone secretion). Although alpha s-A366S is quite stable at testis temperature, it is rapidly degraded at 37 degrees C explaining the PHP-Ia phenotype caused by loss of Gs activity. In vitro experiments indicate that accelerated release of GDP causes both the constitutive activity and the thermolability of alpha s-A366S.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Molecular Mechanisms of Gonadotropin-Inhibitory Hormone (GnIH) Actions in Target Cells and Regulation of GnIH Expression

            Since gonadotropin-inhibitory hormone (GnIH) was discovered in 2000 as the first hypothalamic neuropeptide that actively inhibits gonadotropin release, researches conducted for the last 18 years have demonstrated that GnIH acts as a pronounced negative regulator of reproduction. Inhibitory effect of GnIH on reproduction is mainly accomplished at hypothalamic-pituitary levels; gonadotropin-releasing hormone (GnRH) neurons and gonadotropes are major targets of GnIH action based on the morphological interaction with GnIH neuronal fibers and the distribution of GnIH receptor. Here, we review molecular studies mainly focusing on the signal transduction pathway of GnIH in target cells, GnRH neurons, and gonadotropes. The use of well-defined cellular model systems allows the mechanistic study of signaling pathway occurring in target cells by demonstrating the direct cause-and-effect relationship. The insights gained through studying molecular mechanism of GnIH action contribute to deeper understanding of the mechanism of how GnIH communicates with other neuronal signaling systems to control our reproductive function. Reproductive axis closely interacts with other endocrine systems, thus GnIH expression levels would be changed by adrenal and thyroid status. We also briefly review molecular studies investigating the regulatory mechanisms of GnIH expression to understand the role of GnIH as a mediator between adrenal, thyroid and gonadal axes.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Analysis of GNAS1 and overlapping transcripts identifies the parental origin of mutations in patients with sporadic Albright hereditary osteodystrophy and reveals a model system in which to observe the effects of splicing mutations on translated and untranslated messenger RNA.

              Albright hereditary osteodystrophy (AHO) is caused by heterozygous deactivating GNAS1 mutations. There is a parent-of-origin effect. Maternally derived mutations are usually associated with resistance to parathyroid hormone termed "pseudohypoparathyroidism type Ia." Paternally derived mutations are associated with AHO but usually normal hormone responsiveness, known as "pseudo-pseudohypoparathyroidism." These observations can be explained by tissue-specific GNAS1 imprinting. Regulation of the genomic region that encompasses GNAS1 is complex. At least three upstream exons that splice to exon 2 of GNAS1 and that are imprinted have been reported. NESP55 is exclusively maternally expressed, whereas exon 1A and XL alphas are exclusively paternally expressed. We set out to identify the parental origin of GNAS1 mutations in patients with AHO by searching for their mutation in the overlapping transcripts. This information would be of value in patients with sporadic disease, for predicting their endocrine phenotype and planning follow-up. In doing so, we identified mutations that resulted in nonsense-mediated decay of the mutant Gs alpha transcript but that were detectable in NESP55 messenger RNA (mRNA), probably because they lie within its 3' untranslated region. Analysis of the NESP55 transcripts revealed the creation of a novel splice site in one patient and an unusual intronic mutation that caused retention of the intron in a further patient, neither of which could be detected by analysis of the Gs alpha complementary DNA. This cluster of overlapping transcripts represents a useful model system in which to analyze the effects that mutant sequence has on mRNA-in particular, splicing-and the mechanisms of nonsense-mediated mRNA decay.
                Bookmark

                Author and article information

                Journal
                Clin Pediatr Endocrinol
                Clin Pediatr Endocrinol
                CPE
                Clinical Pediatric Endocrinology
                The Japanese Society for Pediatric Endocrinology
                0918-5739
                1347-7358
                16 April 2020
                2020
                : 29
                : 2
                : 89-90
                Affiliations
                [1 ] Department of Pediatrics, Yokohama Municipal Citizen’s Hospital, Kanagawa, Japan
                [2 ] Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
                Author notes
                Corresponding Author: Tomonobu Hasegawa, MD, PhD, Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
                Tomohiro Ishii, MD, PhD, Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
                Article
                2019-0030
                10.1297/cpe.29.89
                7160456
                873e21bf-c7fc-4bf9-a149-58660a3c9af0
                2020©The Japanese Society for Pediatric Endocrinology

                This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives (by-nc-nd) License. (CC-BY-NC-ND 4.0: http://creativecommons.org/licenses/by-nc-nd/4.0/ ).

                History
                : 04 November 2019
                : 25 December 2019
                Categories
                Mutation-in-Brief

                Comments

                Comment on this article