Gitelman's syndrome: a pathophysiological and clinical update
详细信息    查看全文
  • 作者:Farid Nakhoul (1) (5)
    Nakhoul Nakhoul (1)
    Evgenia Dorman (3)
    Liron Berger (3)
    Karl Skorecki (3) (4)
    Daniella Magen (2) (3) (4)
  • 关键词:Gitelman’s syndrome ; Hypokalemia–hypomagnesemia
  • 刊名:Endocrine
  • 出版年:2012
  • 出版时间:February 2012
  • 年:2012
  • 卷:41
  • 期:1
  • 页码:53-57
  • 全文大小:288KB
  • 参考文献:1. N.V.A.M. Knoers, E.N. Levtchenko, Gitelman syndrome. Orphanet J. Rare Dis. 3, 2,21-26 (2008) CrossRef
    2. N.V.A.M. Knoers, Nine inherited forms of renal hypomagnesemia: an update. Pediatr. Nephrol. 24, 697-05 (2009) CrossRef
    3. M. Roser, N. Eibl, B. Eisenhaber, J. Seringer, M. Nagel, S. Nagorka, F.C. Luft, U. Frei, M. Gollasch, Gitelman Syndrome. Hypertension 53, 893-97 (2009) CrossRef
    4. H.W. Seyberth, An improved terminology and classification of Bartter-like Syndrome. Nat. Clin. Pract. Nephrol. 4, 560-77 (2008) CrossRef
    5. D.N. Cruz, A.J. Shaher, M.J. Bia, R.P. Lifton, D.B. Simon, Gitelman syndrome revisited: an evaluation of symptoms and health related quality of life. Kidney Int. 59, 710-17 (2001) CrossRef
    6. A.J. Shaher, Inherited primary renal tubular hypokalemic alkalosis: a review of Gitelman and Bartter syndromes. Am. J. Med. Sci. 322, 316-32 (2001) CrossRef
    7. E. Riveira-Munoz, Q. Chang, R.J. Bindels, O. Devuyst, Gitelman’s syndrome: towards genotype-phenotype correlations? Pediatr. Nephrol. 22, 326-32 (2007) CrossRef
    8. E. Coto, G. Arriba, M. García-Castro, F. Santos, A.I. Corao, M. Díaz, M. Sánchez Heras, M.A. Basterrechea, S. Tallón, V. Alvarez, Clinical and analytical findings in Gitelman’s syndrome associated with homozygosity for the c.1925 G?>?A / SLC12A3 mutation. Am. J. Nephrol. 30, 218-21 (2009) CrossRef
    9. I. Zelikovic, R. Szargal, A. Hawash, V. Labay, I. Hatib, N. Cohen, F. Nakhoul, A novel mutation in the chloride channel gene CLCNKB as a cause of Gitelman and Bartter syndrome. Kidney Int. 63, 24-2 (2003) CrossRef
    10. R. Vargas-Poussou, K. Dahan, D. Kahila, A. Venisse, E. Riveira-Munoz, H. Debaix, B. Gristar, F. Bridoux, R. Unwin, B. Moulin, X. Jeunemaitre, Spectrum of mutations in Gitelman Syndrome. J. Am. Soc. Nephrol. 22, 693-03 (2011) CrossRef
    11. A.S. Balavoine, P. Bataille, P. Vanhille, R. Azar et al., Phenotype-genotype correlation and follow-up in adult patients with hypokalemia of renal origin suggesting Gitelman syndrome. Eur. J. Endocrinol. 165, 665-73 (2011) CrossRef
    12. A. Noriko, T. Nakayama, Y. Tehira, A. Haketa, M. Yabuki, T. Sekiyama, C. Nakane, H. Mano, K. Matsumoto, Two novel genotypes of the thiazide-sensitive Na-Cl cotransporter (SLC12A3) gene in patients with Gitelman’s syndrome. Endocrine 31, 149-53 (2007) CrossRef
    13. F. Tammaro, A. Bettinelli, D. Cattarelli et al., Early appearance of hypokalemia in Gitelman syndrome. Pediatr. Nephrol. 25, 2179-182 (2010) CrossRef
    14. M. Naesens, P. Steels, R. Verberckmoes, Y. Vanrenterghem, D. Kuypers, Bartter’s and Gitelman’s syndromes: from gene to clinic. Nephron Physiol. 96, 65-8 (2004) CrossRef
    15. G. Graziani, C. Fedeli, L. Moroni, L. Cosmai, S. Badalamenti, C. Ponticelli, Gitelman syndrome: pathophysiological and clinical aspects. QJM 103, 741-48 (2010) CrossRef
    16. R. Tyler Miller, Genetic disorders of NaCl transport in the distal convolute tubule. Nephron Physiol. 118, 15-0 (2011) CrossRef
    17. T. Nijenhuis, V. Vallon, A.W.C.M. van der Kemp, J. Loffing, J.G.J. Hoenderop, R.J.M. Bindels, Enhanced passive Ca++ reabsorption and reduced Mg++ channel abundance explains thiazide-induced hypocalciuria and hypomagnesemia. J. Clin. Invest. 115(6), 1651-658 (2005) CrossRef
    18. K. Aoki, T. Tajima, Y. Yabushita, A. Nakamura, U. Nezu, M. Takahashi, M. Kimura, Y. Terauchi, A novel initial codon mutation of the thiazide-sensitive Na-Cl Cotransporter gene in a Japanese patients with Gitelman’s Syndrome. Endocr. J. 55(3), 557-60 (2008) CrossRef
    19. L. Shao, L. Liu, Z. Miao, H. Ren, W. Wang, Y. Lang, S. Yue, N. Chen, A novel SLC12A3 splicing mutation skipping of two exons and preliminary screening for alternative splice variants in human kidney. Am. J. Nephrol. 28, 900-07 (2008) CrossRef
    20. Y.F. Lo, K. Nozu, K. Lijima, T. Morishita, C.C. Huang, S.S. Yang, H.K. Sytwu, Y.W. Fang, M.H. Tseng, S.H. Lin, Recurrent deep intronic mutations in the SLC12A3 gene responsible for Gitelman’s syndrome. Clin. J. Am. Soc. Nephrol. 6, 630-39 (2011) CrossRef
    21. N. Jeck, M. Konrad, S. Webers, K.E. Bonzel, H.W. Seyberth, Mutations in the chloride channel gene, / CLCNKB, leading to a mixed Bartter-Gitelman phenotype. Pediatr. Res. 48, 754-58 (2000) CrossRef
    22. M. Enya, Y. Kanoh, T. Mune, M. Ishizawa, H. Sarui, M. Yamamoto, N. Takeda, K. Yasuda, J. Takeda, Depressive state and paresthesia dramatically improved by intravenous MgSO4 in Gitelman’s Syndrome. Intern. Med. 43(5), 410-14 (2000) CrossRef
  • 作者单位:Farid Nakhoul (1) (5)
    Nakhoul Nakhoul (1)
    Evgenia Dorman (3)
    Liron Berger (3)
    Karl Skorecki (3) (4)
    Daniella Magen (2) (3) (4)

    1. Nephrology & Hypertension Division, Faculty of Medicine, Baruch-Padeh Poryia Medical Center, Lower Galilee, Israel
    5. Faculty of Medicine, Bar-Ilan University, Tel-Aviv, Israel
    3. Laboratory of Molecular Medicine, Technion-Haifa, Israel
    4. Rambam Health Care Campus, Faculty of Medicine, Technion-Haifa, Israel
    2. Pediatric Nephrology Unit, Technion-Haifa, Israel
文摘
Gitelman’s syndrome (GS), also known as familial hypokalemic hypomagnesemia, is a rare autosomal recessive hereditary salt-losing tubulopathy, characterized by hypokalemic metabolic alkalosis, hypomagnesemia, and hypocalciuria, which is usually caused by mutations in the SLC12A3 gene encoding the thiazide-sensitive sodium chloride contrasporter. Because 18-0% of suspected GS patients carry only one SLC12A3 mutant allele, large genomic rearrangements must account for unidentified mutations. The clinical manifestations of GS are highly variable in terms of age at presentation, severity of symptoms, and biochemical abnormalities. Molecular analysis in our sibling’s patients revealed compound heterozygous mutations in the coding region of SLC12A3 as underlying their disease. Such compound heterozygosity can result in disease phenotype for such loss of function mutations in the absence of homozygosis through consanguineous inheritance of mutant alleles, identical by descent. Missense mutations account for approximately 70% of the mutations in GS, and there is a predisposition to large rearrangements caused by the presence of repeated sequences within the SLC12A3. We report two adult male siblings of Jewish origin with late onset GS, who presented in their fifth decade of life with muscle weakness, hypokalemia, hypomagnesaemia, and metabolic alkalosis. Rapid clinical and biochemical improvement was achieved by replacement therapy with potassium and magnesium.
NGLC 2004-2010.National Geological Library of China All Rights Reserved.
Add:29 Xueyuan Rd,Haidian District,Beijing,PRC. Mail Add: 8324 mailbox 100083
For exchange or info please contact us via email.