12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Novel AVPR2 variant in a male infant with nephrogenic diabetes insipidus who showed delayed head control

      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 Congenital nephrogenic diabetes insipidus (NDI) is a rare disease caused by genetic mutations in AVPR2 or AQP2 (1). AVPR2 is located at the Xq28 locus, and it encodes arginine vasopressin receptor 2 (AVPR2). Mutations in AVPR2 have been associated with X-linked NDI. AQP2 is located at the 12q13.12 locus, and it encodes the water transporter aquaporin-2. Mutations in AQP2 result in autosomal NDI. Here, we describe a male infant with a novel AVPR2 variant who was referred to our hospital due to delayed head control. Case Report A 5-mo-old boy was referred to the Department of Child Neurology in Okayama University Hospital (Okayama, Japan) due to delayed head control. He was born to non-consanguineous parents. His birth weight was 3630 g (+ 1.39 SD) and birth length was 50.8 cm (+ 0.86 SD). He showed no signs of asphyxia. A recurrent fever of unknown origin was observed beginning at 2 mo of age. Poor weight gain was observed at 3 mo of age. Notably, he did not take any medications. At his first visit to our hospital, the patient’s length was 62.6 cm (–1.55 SD) and weight was 6190 g (–1.82 SD). His heart rate was 128 bpm, and his body temperature was 36.8°C. He was dehydrated, and his head control was incomplete. Laboratory examination revealed that his sodium level and serum osmolarity were elevated, and his urinary osmolarity and specific gravity were noticeably low. Head magnetic resonance imaging scanning revealed that high posterior lobe intensity was absent in T1-weighted images. However, the pituitary stalk was intact, and no other abnormalities were identified. These findings were indicative of diabetes insipidus. Water deprivation test with vasopressin challenge test resulted in no urine osmolarity increase and no urine volume decrease. Moreover, the AVP level at admission was extremely high (114 pg/mL; reference range; 0.0–4.2 pg/mL). Based on these findings, we diagnosed the patient with congenital NDI. His urine volume decreased, his weight increased, and head control was achieved after initiation of hydrochlorothiazide. Assessment of the patient’s family history revealed that his mother had polydipsia and polyuria from infancy until the present day (Fig. 1A Fig. 1. A: Family tree of our patient (arrow). Black square and black circle indicate family members with polydipsia and polyuria, respectively. Arrow indicates our patient. B: Results of mutation analysis. Our patient showed hemizygous duplication of four base pairs (c.990_993 dup CAGC; single bold line indicates CAGC). His older brother did not show this duplication. His mother was heterozygous for the wild-type allele and the four-base pair duplication (dotted line). ). She also had increased levels of sodium of unknown origin during each of her three pregnancies. Furthermore, the patient’s maternal grandfather, maternal great-grandfather, and maternal aunt had histories of polyuria and polydipsia. We suspected that our patient had familial NDI and, thus, conducted a genetic analysis. Mutational Analysis Genetic analysis was approved by the ethical committee of Okayama University Hospital and conducted in accordance with the 1975 Declaration of Helsinki and subsequent amendments. Informed consent for genetic analysis was obtained from the patient’s mother. We identified a hemizygous four-base pair duplication in exon 3 of AVPR2 (c.990_993 dup CAGC, Fig. 1B). This four-base pair duplication results in early termination (p.Val332Gln Fs26X). This variant was previously unreported, and it is not present in the 1000 Genomes Project databases (http://www.internationalgenome.org/1000-genomes-browers), the Human Genetic Variation Database (http://www.hgvd.genome.med.kyoto-u.ac.jp), or the Exome Aggregation Consortium Server (http://exac.broadinstitute.org). This variant is considered to be likely pathogenic based on ACMG criteria (PS1, PM2 and PM4). We suspected that the patient’s mother was heterozygous for the wild-type allele and the four-base pair duplication. Thus, an amplicon obtained from the mother was cloned into a PCR® 4-TOPO® vector using the TOPO® TA Cloning® Kit for Sequencing (Thermo Fisher Scientific, Waltham, MA, USA), and each transformant was, subsequently, sequenced. The results confirmed heterozygosity for the wild-type allele and the four-base pair duplication. Since the patient’s mother suspected that the patient’s older brother had polydipsia, we also analyzed the patient’s older brother, However, the patient’s brother did not carry the four-base pair duplication. Notably, we did not analyze AQP2 in our patient, and we did not perform genetic analysis for his father, older sister, maternal aunt, maternal grandfather, or maternal great-grandfather. Discussion We identified a novel AVPR2 variant in a patient with familial congenital NDI. Duplication variants are relatively rare among X-linked NDI patients: insertion AVPR2 variants were found in only about 5% of large cohort (1) and Japanese patients (2). There are five classes of loss of function mutations in AVPR2 (1). Class I mutations result in unstable mRNA, which undergoes nonsense-mediated RNA decay. Class II mutations cause misfolding of receptors, and these mutant receptors remain in the endoplasmic reticulum. Class III mutations also cause receptor misfolding. Class III receptors reach the plasma membrane and react with AVP, but subsequent interactions with G proteins and cAMP production are impaired. Class IV mutations also result in misfolded receptors that reach the plasma membrane, but result in incomplete AVP binding. Class V mutations cause mis-sorting to incorrect cellular compartments. A duplication of four bases in exon 3 of AVPR2 causes early termination. However, this is the final exon, which might avoid nonsense-mediated RNA decay (3). Residue 332 is located in an intracellular domain that does not bind to AVP. Therefore, the variant in our patient may lose its function via a non-class I mechanism. Functional analysis is needed to clarify the pathophysiology of this variant. Congenital NDI is generally identified by the presence of DI symptoms like polyuria, polydipsia, fever, and poor weight gain (2). The patient whose case is reported here was referred to the department of Child Neurology in our hospital because of delayed head control, poor weight gain, and fever. After treating his dehydration, he gained head control and his development proceeded normally. We consider delayed motor development in infancy to be an important clinical symptom of congenital NDI. The patient’s mother had a heterozygous AVPR2 variant. She demonstrated a high sodium level during each of her pregnancies, but her sodium levels were normal before pregnancy and after birth for all children. In gestational DI, urinary volume increases during pregnancy because of increased clearance of AVP by placental vasopressinase, a cystine aminopeptidase that strongly degrades AVP (4). Although inactivation of the X chromosome was not analyzed in the patient’s mother, AVP resistance and degradation by vasopressinase, might have contributed to her high sodium levels during pregnancy. In conclusion, we identified a novel AVPR2 variant in a patient with familial congenital NDI. Common symptoms of DI were observed, including polyuria, polydipsia, poor weight gain, and fever, and delayed motor development was also observed during infancy. Conflict of Interest The authors declare that they have no conflict of interest.

          Related collections

          Most cited references4

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

          Nonsense-mediated decay approaches the clinic.

          Nonsense-mediated decay (NMD) eliminates mRNAs containing premature termination codons and thus helps limit the synthesis of abnormal proteins. New results uncover a broader role of NMD as a pathway that also affects the expression of wild-type genes and alternative-splice products. Because the mechanisms by which NMD operates have received much attention, we discuss here the emerging awareness of the impact of NMD on the manifestation of human genetic diseases. We explore how an understanding of NMD accounts for phenotypic differences in diseases caused by premature termination codons. Specifically, we consider how the protective function of NMD sometimes benefits heterozygous carriers and, in contrast, sometimes contributes to a clinical picture of protein deficiency by inhibiting expression of partially functional proteins. Potential 'NMD therapeutics' will therefore need to strike a balance between the general physiological benefits of NMD and its detrimental effects in cases of specific genetic mutations.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Nephrogenic diabetes insipidus: essential insights into the molecular background and potential therapies for treatment.

            The water channel aquaporin-2 (AQP2), expressed in the kidney collecting ducts, plays a pivotal role in maintaining body water balance. The channel is regulated by the peptide hormone arginine vasopressin (AVP), which exerts its effects through the type 2 vasopressin receptor (AVPR2). Disrupted function or regulation of AQP2 or the AVPR2 results in nephrogenic diabetes insipidus (NDI), a common clinical condition of renal origin characterized by polydipsia and polyuria. Over several years, major research efforts have advanced our understanding of NDI at the genetic, cellular, molecular, and biological levels. NDI is commonly characterized as hereditary (congenital) NDI, arising from genetic mutations in the AVPR2 or AQP2; or acquired NDI, due to for exmple medical treatment or electrolyte disturbances. In this article, we provide a comprehensive overview of the genetic, cell biological, and pathophysiological causes of NDI, with emphasis on the congenital forms and the acquired forms arising from lithium and other drug therapies, acute and chronic renal failure, and disturbed levels of calcium and potassium. Additionally, we provide an overview of the exciting new treatment strategies that have been recently proposed for alleviating the symptoms of some forms of the disease and for bypassing G protein-coupled receptor signaling.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Clinical Overview of Nephrogenic Diabetes Insipidus Based on a Nationwide Survey in Japan

              Background Nephrogenic diabetes insipidus (NDI) is a rare disease whose complications include polyuria, renal dysfunction, growth disorder and mental retardation. The details of NDI’s clinical course have been unclear. To address this uncertainty, we performed a large investigation of the clinical course of NDI in Japan. Methods Between December 2009 and March 2011, we provided a primary questionnaire to 26,282 members of the Japan Endocrine Society, the Japanese Urological Association, the Japanese Society for Pediatric Endocrinology, the Japanese Society for Pediatric Nephrology, the Japanese Society of Nephrology, the Japanese Society of Neurology and the Japanese Society of Pediatric Urology. In addition, we provided a secondary questionnaire to 121 members who reported experience with cases of NDI. We asked about patient’s age at onset, diagnosis, complications, effect of treatment and patient’s genotype. Results We enrolled 173 patients with NDI in our study. Of these NDI patients, 143 were congenital and 30 were acquired. Of the 173, 73 patients (42%) experienced urologic complications. Among the 143 with congenital NDI, 20 patients (14%) had mental retardation. Patients with NDI mainly received thiazide diuretics, and some patients responded to treatment with desmopressin acetate (DDAVP). Gene analyses were performed in 87 patients (61%) with congenital NDI, revealing that 65 patients had an arginine vasopressin receptor type 2 (AVPR2) gene mutation and that 8 patients (9.2%) had an aquaporin 2 (AQP2) gene mutation. Patients with the AVPR2 mutation (D85N) generally showed a mild phenotype, and we found that DDAVP was generally an effective treatment for NDI among these patients. Conclusion We suggest that adequate diagnosis and treatment are the most important factors for improving prognoses. We further suggest that gene analysis should be performed for optimal treatment selection and the early detection of NDI among siblings.
                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
                19 October 2019
                2019
                : 28
                : 4
                : 155-158
                Affiliations
                [1 ] Department of Pediatrics, Okayama University Hospital, Okayama, Japan
                [2 ] Advanced Critical Care and Emergency Center, Okayama University Hospital, Okayama, Japan
                [3 ] Department of Pediatrics, Okayama University Graduate School of Medicine Dentistry, and Pharmaceutical Sciences, Okayama, Japan
                [4 ] Department of Child Neurology, Okayama University Hospital, Okayama, Japan
                [5 ] Department of Child Neurology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
                Article
                2019-0018
                10.1297/cpe.28.155
                6801361
                2eced34b-e769-4cb6-8f24-f93d0f454d4e
                2019©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
                : 13 May 2019
                : 02 August 2019
                Categories
                Mutation-in-Brief

                developmental delay,polyuria,polydipsia,fever,poor weight gain

                Comments

                Comment on this article