Skip to main content

Advertisement

Log in

Rare diseases in clinical endocrinology: a taxonomic classification system

  • Consensus Statement
  • Published:
Journal of Endocrinological Investigation Aims and scope Submit manuscript

Abstract

Purpose

Rare endocrine–metabolic diseases (REMD) represent an important area in the field of medicine and pharmacology. The rare diseases of interest to endocrinologists involve all fields of endocrinology, including rare diseases of the pituitary, thyroid and adrenal glands, paraganglia, ovary and testis, disorders of bone and mineral metabolism, energy and lipid metabolism, water metabolism, and syndromes with possible involvement of multiple endocrine glands, and neuroendocrine tumors. Taking advantage of the constitution of a study group on REMD within the Italian Society of Endocrinology, consisting of basic and clinical scientists, a document on the taxonomy of REMD has been produced.

Methods and results

This document has been designed to include mainly REMD manifesting or persisting into adulthood. The taxonomy of REMD of the adult comprises a total of 166 main disorders, 338 including all variants and subtypes, described into 11 tables.

Conclusions

This report provides a complete taxonomy to classify REMD of the adult. In the future, the creation of registries of rare endocrine diseases to collect data on cohorts of patients and the development of common and standardized diagnostic and therapeutic pathways for each rare endocrine disease is advisable. This will help planning and performing intervention studies in larger groups of patients to prove the efficacy, effectiveness, and safety of a specific treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Abbreviations

ACA Abs:

Adrenal cortex antibodies

AFP:

Alpha fetoprotein

ALP:

Alkaline phosphatase

Apo B:

Apolipoprotein B

AVP:

Arginine vasopressin

BAP:

Bone alkaline phosphatase

BB-CK:

Brain-specific creatine kinase isoenzyme

β-HCG:

β-Human chorionic gonadotropin

Ca:

Calcium

cAMP:

Cyclic adenosine monophosphate

CaSR:

Calcium-sensing receptor

CEA:

Carcino-embryonic antigen

CgA:

Chromogranin A

CK:

Creatin kinase

DDAVP:

Desmopressin

DHEA:

Dehydroepiandrosterone

DHEA-s:

Dehydroepiandrosterone-sulfate

DHT:

Dihydrotestosterone

DIT:

Di-iodinated tyrosine

DOC:

Deoxycorticosterone

FGF23:

Fibroblast growth factor 23

FPG:

Fasting plasma glucose

GAD Abs:

Glutamic acid decarboxylase antibodies

GIST:

Gastro-intestinal stromal tumors

Gs:

G-protein alpha subunit

hCG:

Human chorionic gonadotropin

HDL:

High-density lipoprotein

5-HIAA:

5-Hydroxyindoleacetic acid

5-HT:

5-Hydroxytryptophan

IA2 Abs:

Tyrosine phosphatase antibodies

IDL:

Intermediate-density lipoprotein

IGF-1:

Insulin-like growth factor

K:

Potassium

LDL:

Low-density lipoprotein

LPL:

Lipoprotein lipase

Mg:

Magnesium

MIT:

Mono-iodinated tyrosine

MPC Abs:

Melanin-producing cells antibodies

Na:

Sodium

NSE:

Neuron-specific enolase

21OH Abs:

21 hydroxylase antibodies

17-OHP:

17 OH progesterone

17α-OH Abs:

17 alpha-hydroxylase antibodies

P:

Phosphorus

PCA Abs:

Parietal cell antibodies

PP:

Pancreatic polypeptide

PRL:

Prolactin

PTHrP:

Parathyroid hormone-related protein

SCC Abs:

Side-chain cleavage enzyme antibodies

SS:

Somatostatin

StCA Abs:

Steroid-producing cell antibodies

TmP/GFR:

Renal tubular reabsorption of phosphate

tTGA Abs:

Tissue transglutaminase antibodies

TPO Abs:

Thyroid peroxidase antibodies

UFC:

Urinary free cortisol

Ur. Ca:

Urinary calcium

Ur. K:

Urinary potassium

Ur. Na:

Urinary sodium

Ur. P:

Urinary phosphorus

VIP:

Vasoactive intestinal polypeptide

VLCFA:

Very long chain fatty acids

References

  1. Orphan Drug Act amendment (1983) http://history.nih.gov/research/downloads/PL97-414.pdf.

  2. Rare Disease Act (2002) https://www.govtrack.us/congress/bills/107/hr4013/text

  3. Institute of Medicine (2010) Rare diseases and orphan products: accelerating research and development. The National Academies Press, Washington DC

    Google Scholar 

  4. European Union, legislation in Regulation EC No 141/2000 on a Decision Act of the European Parliament and of the Council No 1295/1999/EC. http://ec.europa.eu/health/files/eudralex/vol-1/reg_2000_141/reg_2000_141_en.pdf

  5. European Union Committee of Expert on Rare Disease (2013) Report on the state of the art of rare disease activities in Europe. Part I. pp 1–78. http://www.eucerd.eu/upload/file/Reports/2013ReportStateofArtRDActivities.pdf

  6. European Organisation for Rare Diseases (EURORDIS) (2005) Rare diseases: understanding this public health priority. pp 1–14. http://www.eurordis.org/IMG/pdf/princeps_document-EN.pdf

  7. European Union Committee of Experts on Rare Diseases (2012) Report on the state of the art of rare disease activities in Europe of The European Union Committee Of experts on rare diseases. http://www.eucerd.eu/?post_type=document&p=1378

  8. Prevalence of rare diseases: Bibliographic data, Orphanet Report Series, Rare Diseases collection, Number 1: Listed in alphabetical order of disease or group of diseases (2013) http://www.orpha.net/orphacom/cahiers/docs/GB/Prevalence_of_rare_diseases_by_alphabetical_list.pdf

  9. Griggs RC, Batshaw M, Dunkle M, Gopal-Srivastava R, Kaye E, Krischer J, Nguyen T, Paulus K, Merkel PA, Rare Diseases Clinical Research Network (2009) Clinical research for rare disease: opportunities, challenges, and solutions. Mol Genet Metab 96:20–26

    CAS  PubMed Central  PubMed  Google Scholar 

  10. 5th European Conference on rare Diseases European reference networks and Center of Expertise for rare Diseases (2010) pp 1–75

  11. Office of Rare Disease Research. Global rare disease patient registry and data repository background. (2013) [cited 2013 March 1]

  12. Daneshvari S, Youssof S, Kroth PJ (2013) The NIH Office of Rare Diseases Research Patient Registry Standard: a report from the University of New Mexico’s Oculopharyngeal Muscular Dystrophy Patient Registry: AMIA Annu Symp Proc 269–277

  13. Bellgard M, Beroud C, Parkinson K, Harris T, Ayme S, Baynam G, Weeramanthri T, Dawkins H, Hunter A (2014) Correction: dispelling myths about rare disease registry system development. Source Code Biol Med 9:4

    PubMed Central  PubMed  Google Scholar 

  14. Landais P, Messiaen C, Rath A, Le Mignot L, Dufour E, Ben Said M, Jais JP, Toubiana L, Baujat G, Bourdon-Lanoy E, Gérard-Blanluet M, Bodemer C, Salomon R, Aymé S, Le Merrer M, Verloes A (2010) CEMARA task force. CEMARA an information system for rare diseases. Stud Health Technol Inform 160:481–485

    PubMed  Google Scholar 

  15. Aymé S, Rodwell C (2014) The European Union Committee of Experts on Rare Diseases: three productive years at the service of the rare disease community. Orphanet J Rare Dis 9:30

    PubMed Central  PubMed  Google Scholar 

  16. Aymé S, Kole A, Groft S (2008) Empowerment of patients: lessons from the rare diseases community. Lancet 371:2048–2051

    PubMed  Google Scholar 

  17. Kole A, Faurisson F (2010) Rare diseases social epidemiology: analysis of inequalities. Adv Exp Med Biol 686:223–250

    PubMed  Google Scholar 

  18. Taru Taruscio D, Gentile AE, De Santis M, Ferrelli RM, Posada de la Paz M, Hens M, Huizer J, Fregonese L, Stefanov R, Bottarelli V, Weinman A, Le Cam Y, Gavhed D, Mincarone P, Bushby K, Frazzica RG, Donati C, Vittozzi L, Jessop E (2013) EUROPLAN: a project to support the development of national plans on rare diseases in Europe. Public Health Genomics 16:278–287

    Google Scholar 

  19. National Organization for Rare Diseases (NORD) of the United States. http://www.rarediseases.org

  20. Pastores GM, Gupta P (2013) Orphan drug development. Pediatr Endocrinol Rev 11:64–67

    PubMed  Google Scholar 

  21. Hamosh A, Scott AF, Amberger J, Valle D, McKusick VA (2000) Online Mendelian inheritance in man (OMIM). Hum Mutat 15:57–61

    CAS  PubMed  Google Scholar 

  22. Gray KA, Daugherty LC, Gordon SM, Seal RL, Wright MW, Bruford EA (2013) Genenames.org: the HGNC resources in 2013. Nucleic Acids Res 41:D545–D552

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Rath A, Olry A, Dhombres F, Brandt MM, Urbero B, Ayme S (2012) Representation of rare diseases in health information systems: the Orphanet approach to serve a wide range of end users. Hum Mutat 33:803–808

    PubMed  Google Scholar 

  24. Melmed S, Colao A, Barkan A, Molitch M, Grossman AB, Kleinberg D, Clemmons D, Chanson P, Laws E, Schlechte J, Vance ML, Ho K, Giustina A, Acromegaly Consensus Group (2009) Guidelines for acromegaly management: an update. J Clin Endocrinol Metab 94:1509–1517

    CAS  PubMed  Google Scholar 

  25. Beck-Peccoz P, Persani L, Mannavola D, Campi I (2009) Pituitary tumours: TSH-secreting adenomas. Best Pract Res Clin Endocrinol Metab 23:597–606

    CAS  PubMed  Google Scholar 

  26. Castinetti F, Morange I, Conte-Devolx B, Brue T (2012) Cushing’s disease. Orphanet J Rare Dis 7:41

    PubMed Central  PubMed  Google Scholar 

  27. Chanson P, Brochier S (2005) Non-functioning pituitary adenomas. J Endocrinol Invest 28:93–99

    CAS  PubMed  Google Scholar 

  28. Dahlqvist P, Koskinen LO, Brännström T, Hägg E (2010) Testicular enlargement in a patient with a FSH-secreting pituitary adenoma. Endocrine 37:289–293

    CAS  PubMed  Google Scholar 

  29. Martucci F, Trivellin G, Korbonits M (2012) Familial isolated pituitary adenomas: an emerging clinical entity. J Endocrinol Invest 11:1003–1014

    Google Scholar 

  30. Mitchell AL, Dwyer A, Pitteloud N, Quinton R (2011) Genetic basis and variable phenotypic expression of Kallmann syndrome: towards a unifying theory. Trends Endocrinol Metab 22:249–258

    CAS  PubMed  Google Scholar 

  31. Moore SJ, Green JS, Fan Y, Bhogal AK, Dicks E, Fernandez BA, Stefanelli M, Murphy C, Cramer BC, Dean JC, Beales PL, Katsanis N, Bassett AS, Davidson WS, Parfrey PS (2005) Clinical and genetic epidemiology of Bardet–Biedl syndrome in Newfoundland: a 22-year prospective, population-based, cohort study. Am J Med Genet A 132:352–360

    PubMed Central  Google Scholar 

  32. Feigerlová E, Diene G, Oliver I, Gennero I, Salles JP, Arnaud C, Tauber M (2010) Elevated insulin-like growth factor-I values in children with Prader–Willi syndrome compared with growth hormone (GH) deficiency children over two years of GH treatment. J Clin Endocrinol Metab 95:4600–4608

    PubMed  Google Scholar 

  33. Marshall JD, Bronson RT, Collin GB, Nordstrom AD, Maffei P, Paisey RB, Carey C, Macdermott S, Russell-Eggitt I, Shea SE, Davis J, Beck S, Shatirishvili G, Mihai CM, Hoeltzenbein M, Pozzan GB, Hopkinson I, Sicolo N, Naggert JK, Nishina PM (2005) New Alstrom syndrome phenotypes based on the evaluation of 182 cases. Arch Intern Med 165:675–683

    PubMed  Google Scholar 

  34. Miyata I, Vallette-Kasic S, Saveanu A, Takeuchi M, Yoshikawa H, Tajima A, Tojo K, Reynaud R, Gueydan M, Enjalbert A, Tajima N, Eto Y, Brue T (2006) Identification and functional analysis of the novel S179R POU1F1 mutation associated with combined pituitary hormone deficiency. J Clin Endoc Metab 91:4981–4987

    CAS  Google Scholar 

  35. De Graaff LCG, Pagon RA, Adam MP, Bird TD, Dolan CR, Fong CT, Smith RJH, Stephens K (2000) PROP1-related combined pituitary hormone deficiency. GeneReviewsSeattle (WA): University of Washington, Seattle; 1993–2014 Dec 07 (updated 2011 Oct 06)

  36. Bhangoo APS, Hunter CS, Savage JJ, Anhalt H, Pavlakis S, Walvoord EC, Ten S, Rhodes SJ (2006) Clinical case seminar: a novel LHX3 mutation presenting as combined pituitary hormonal deficiency. J Clin Endoc Metab 91:747–753

    CAS  Google Scholar 

  37. Castinetti F, Saveanu A, Reynaud R, Quentien MH, Buffin A, Brauner R, Kaffel N, Albarel F, Guedj AM, El Kholy M, Amin M, Enjalbert A, Barlier A, Brue T (2008) A novel dysfunctional LHX4 mutation with high phenotypical variability in patients with hypopituitarism. J Clin Endoc Metab 93:2790–2799

    CAS  Google Scholar 

  38. Webb EA, Dattani MT (2010) Septo-optic dysplasia. Eur J Hum Genet 18:393–397

    PubMed Central  PubMed  Google Scholar 

  39. Mullis PE (2007) Genetics of growth hormone deficiency. Endocr Metab Clin N Am 36:17–36

    CAS  Google Scholar 

  40. Casey M, Vaughan CJ, He J, Hatcher CJ, Winter JM, Weremowicz S, Montgomery K, Kucherlapati R, Morton CC, Basson CT (2000) Mutations in the protein kinase A R1-alpha regulatory subunit cause familial cardiac myxomas and Carney complex. J Clin Invest 106:R31–R38

    CAS  PubMed Central  PubMed  Google Scholar 

  41. Force American Thyroid Association Guidelines Task, Kloos RT, Eng C, Evans DB, Francis GL, Gagel RF, Gharib H, Moley JF, Pacini F, Ringel MD, Schlumberger M, Wells SA Jr (2009) Medullary thyroid cancer: management guidelines of the American Thyroid Association. Thyroid 19:565–612

    Google Scholar 

  42. Wells SA Jr, Pacini F, Robinson BG, Santoro M (2013) Multiple endocrine neoplasia type 2 and familial medullary thyroid carcinoma: an update. J Clin Endocrinol Metab 98:3149–3164

    CAS  PubMed  Google Scholar 

  43. Romei C, Mariotti S, Fugazzola L, Taccaliti A, Pacini F, Opocher G, Mian C, Castellano M, degli Uberti E, Ceccherini I, Cremonini N, Seregni E, Orlandi F, Ferolla P, Puxeddu E, Giorgino F, Colao A, Loli P P, Bondi F, Cosci B, Bottici V, Cappai A, Pinna G, Persani L, Verga U, Boscaro M, Castagna MG, Cappelli C, Zatelli MC MC, Faggiano A, Francia G, Brandi ML ML, Falchetti A, Pinchera A, Elisei R, ItaMEN network (2010) Multiple endocrine neoplasia type 2 syndromes (MEN 2): results from the ItaMEN network analysis on the prevalence of different genotypes and phenotypes. Eur J Endocrinol 163:301–308

    CAS  PubMed  Google Scholar 

  44. Ain KB (1998) Anaplastic thyroid carcinoma: behavior, biology, and therapeutic approaches. Thyroid 8:715–726

    CAS  PubMed  Google Scholar 

  45. Carney JA, Gordon H, Carpenter PC, Shenoy BV (1985) The complex of myxomas, spotty pigmentation, and endocrine overactivity. Medicine (Baltimore) 64:270–283

    CAS  Google Scholar 

  46. Blumenthal GM, Dennis PA (2008) PTEN hamartoma tumor syndromes. Eur J Hum Gen 16:1289–1300

    CAS  Google Scholar 

  47. Half E, Bercovich D, Rozen P (2009) Familial adenomatous polyposis. Orphanet J Rare Dis 12(4):22

    Google Scholar 

  48. Khan A, Smellie J, Nutting C, Harrington K, Newbold K (2010) Familial nonmedullary thyroid cancer: a review of the genetics. Thyroid 20:795–801

    CAS  PubMed  Google Scholar 

  49. Peretz A, Leiberman E, Kapelushnik J, Hershkovitz E (2004) Thyroglossal duct carcinoma in children: case presentation and review of the literature. Thyroid 14:777–785

    PubMed  Google Scholar 

  50. Refetoff S, DeWind LT, DeGroot LJ (1967) Familial syndrome combining deaf-mutism, stuppled epiphyses, goiter and abnormally high PBI: possible target organ refractoriness to thyroid hormone. Clin Endocrinol Metab 27:279–294

    CAS  Google Scholar 

  51. Bochukova E, Schoenmakers N, Agostini M, Schoenmakers E, Rajanayagam O, Keogh JM, Henning E, Reinemund J, Gevers E, Sarri M, Downes K, Offiah A, Albanese A, Halsall D, Schwabe JW, Bain M, Lindley K, Muntoni F, Vargha-Khadem F, Dattani M, Farooqi IS, Gurnell M, Chatterjee K (2012) A mutation in the thyroid hormone receptor alpha gene. N Engl J Med 366:243–249

    CAS  PubMed  Google Scholar 

  52. Schwartz CE, Ulmer J, Brown A, Pancoast I, Goodman HO, Stevenson RE (1990) Allan–Herndon syndrome. II. Linkage to DNA markers in Xq21. Am J Hum Genet 47:454–458

    CAS  PubMed Central  PubMed  Google Scholar 

  53. Bamforth JS, Hughes I, Lazarus J, John R (1986) Congenital anomalies associated with hypothyroidism. Arch Dis Child 61:608–609

    CAS  PubMed Central  PubMed  Google Scholar 

  54. Sunthornthepvarakui T, Gottschalk ME, Hayashi Y, Refetoff S (1995) Brief report: resistance to thyrotropin caused by mutations in the thyrotropin-receptor gene. N Engl J Med 332:155–160

    CAS  PubMed  Google Scholar 

  55. Everett LA, Glaser B, Beck JC, Idol JR, Buchs A, Heyman M, Adawi F, Hazani E, Nassir E, Baxevanis AD, Sheffield VC, Green ED (1997) Pendred syndrome is caused by mutations in a putative sulphate transporter gene (PDS). Nat Genet 17:411–422

    CAS  PubMed  Google Scholar 

  56. Macchia PE, Lapi P, Krude H, Pirro MT, Missero C, Chiovato L, Souabni A, Baserga M, Tassi V, Pinchera A, Fenzi G, Gruters A, Busslinger M, Di Lauro R (1998) PAX8 mutations associated with congenital hypothyroidism caused by thyroid dysgenesis. Nat Genet 19:83–86

    CAS  PubMed  Google Scholar 

  57. Devriendt K, Vanhole C, Matthijs G, de Zegher F (1998) Deletion of thyroid transcription factor-1 gene in an infant with neonatal thyroid dysfunction and respiratory failure. (Lett) New Eng J Med 338:1317–1318

    CAS  Google Scholar 

  58. Schussler GC (2000) The thyroxine-binding proteins. Thyroid 10:141–149

    CAS  PubMed  Google Scholar 

  59. Fujiwara H, Tatsumi K, Miki K, Harada T, Miyai K, Takai S, Amino N (1997) Congenital hypothyroidism caused by a mutation in the Na(+)/I(−) symporter. (Letter) Nat Genet 16:124–125. (Note: Erratum: Nat Genet 17:122 only, 1997)

  60. Abramowicz MJ, Targovnik HM, Varela V, Cochaux P, Krawiec L, Pisarev MA, Propato FVE, Juvenal G, Chester HA, Vassart G (1992) Identification of a mutation in the coding sequence of the human thyroid peroxidase gene causing congenital goiter. J Clin Invest 90:1200–1204

    CAS  PubMed Central  PubMed  Google Scholar 

  61. Ieiri T, Cochaux P, Targovnik HM, Suzuki M, Shimoda SI, Perret J, Vassart G (1991) A 3-prime splice site mutation in the thyroglobulin gene responsible for congenital goiter with hypothyroidism. J Clin Invest 88:1901–1905

    CAS  PubMed Central  PubMed  Google Scholar 

  62. Moreno JC, Klootwijk W, van Toor H, Pinto G, D’Alessandro M, Leger A, Goudie D, Polak M, Gruters A, Visser TJ (2008) Mutations in the iodotyrosine deiodinase gene and hypothyroidism. New Eng J Med 358:1811–1818

    CAS  PubMed  Google Scholar 

  63. Moreno JC, Bikker H, Kempers MJE, van Trotsenburg ASP, Baas F, de Vijlder JJM, Vulsma T, Ris-Stalpers C (2002) Inactivating mutations in the gene for thyroid oxidase 2 (THOX2) and congenital hypothyroidism. New Eng J Med 347:95–100

    CAS  PubMed  Google Scholar 

  64. Van Dijk FS, Pals G, Van Rijn RR, Nikkels PG, Cobben JM (2010) Classification of Osteogenesis Imperfecta revisited. Eur J Med Genet 53:1–5

    PubMed  Google Scholar 

  65. Marcucci G, Masi L, Carossino AM, Franchi A, Capanna R, Sinigaglia L, Brandi ML (2013) Cystic bone angiomatosis: a case report treated with aminobisphosphonates and review of the literature. Calcif Tissue Int 95:9761–9763

    Google Scholar 

  66. Kiran DN, Anupama A (2011) Vanishing bone disease: a review. J Oral Maxillofac Surg 69:199–203

    CAS  PubMed  Google Scholar 

  67. Liberman UA, Marx SJ (2001) Vitamin D and other calciferols. In: Scriver CR, Beaudet AL, Sly WS, Valle D (eds) The metabolic and molecular bases of inherited disease, vol II, 8th edn. McGraw-Hill, New York, pp 4223–4240

  68. Casella SJ, Reiner BJ, Chen TC, Holick MF, Harrison HE (1994) A possible genetic defect in 25-hydroxylation as a cause of rickets. J Pediatr 124:929–932

    CAS  PubMed  Google Scholar 

  69. Hewison M, Rut AR, Kristjansson K, Walker RE, Dillon MJ, Hughes MR, O’Riordan JL (1993) Tissue resistance to 1,25-dihydroxyvitamin D without a mutation of the vitamin D receptor gene. Clin Endocrinol 39:663–670

    CAS  Google Scholar 

  70. Masi L, Gozzini A, Franchi A, Campanacci D, Amedei A, Falchetti A, Franceschelli F, Marcucci G, Tanini A, Capanna R, Brandi ML (2009) A novel recessive mutation of fibroblast growth factor-23 in tumoral calcinosis. J Bone Jt Surg Am 91:1190–1198

    CAS  Google Scholar 

  71. Hershkovitz D, Gross Y, Nahum S, Yehezkel S, Sarig O, Uitto J, Sprecher E (2011) Functional characterization of SAMD9, a protein deficient in normophosphatemic familial tumoral calcinosis. J Invest Dermatol 131:662–669

    CAS  PubMed Central  PubMed  Google Scholar 

  72. Bastepe M, Jüppner H (2008) Inherited hypophosphatemic disorders in children and the evolving mechanisms of phosphate regulation. Rev Endocr Metab Disord 9:171–180

    PubMed  Google Scholar 

  73. ADHR Consortium (2000) Autosomal dominant hypophosphataemic rickets is associated with mutations in FGF23. Nat Genet 26:345–348

    Google Scholar 

  74. White KE, Cabral JM, Davis SI, Fishburn T, Evans WE, Ichikawa S, Fields J, Yu X, Shaw NJ, McLellan NJ, McKeown C, Fitzpatrick D, Yu K, Ornitz DM, Econs MJ (2005) Mutations that cause osteoglophonic dysplasia define novel roles for FGFR1 in bone elongation. J Hum Genet 76:361–367

    CAS  Google Scholar 

  75. Brownstein CA, Adler F, Nelson-Williams C, Iijima J, Li P, Imura A, Nabeshima Y, Reyes-Mugica M, Carpenter TO, Lifton RP (2008) A translocation causing increasing α-Klotho level results in hypophosphatemic rickets and hyperparathyroidism. Proc Natl Acad Sci 105:3455–3460

    CAS  PubMed Central  PubMed  Google Scholar 

  76. Ihde LL, Forrester DM, Gottsegen CJ, Masih S, Patel DB, Vachon LA, White EA, Matcuk GR Jr (2011) Sclerosing bone dysplasias: review and differentiation from other causes of osteosclerosis. Radiographics 31:1865–1882

    PubMed  Google Scholar 

  77. Pangrazio A, Boudin E, Piters E, Damante G, Lo Iacono N, D’Elia AV, Vezzoni P, Van Hul W, Villa A, Sobacchi C (2011) Identification of the first deletion in the LRP5 gene in a patient with autosomal dominant osteopetrosis type I. Bone 49:568–571

    CAS  PubMed Central  PubMed  Google Scholar 

  78. Benichou OD, Laredo JD, de Vernejoul MC (2000) Type II autosomal dominant osteopetrosis (Albers-Schonberg disease): clinical and radiological manifestations in 42 patients. Bone 26:87–93

    CAS  PubMed  Google Scholar 

  79. Xue Y, Cai T, Shi S, Wang W, Zhang Y, Mao T, Duan X (2011) Clinical and animal research findings in pycnodysostosis and gene mutations of cathepsin K from 1996 to 2011. Orphanet J Rare Dis 10:6–20

    Google Scholar 

  80. de Vernejoul MC (2008) Sclerosing bone disorders. Best Pract Res Clin Rheumatol 22:71–83

    PubMed  Google Scholar 

  81. Sheppard WM, Shprintzen RJ, Tatum SA, Woods CI (2003) Craniometaphyseal dysplasia: a case report and review of medical and surgical management. Int J Pediatr Otorhinolaryngol 67:687–693

    PubMed  Google Scholar 

  82. Gagliardi GG, Mahan KT (2010) Melorheostosis: a literature review and case report with surgical considerations. J Foot Ankle Surg 49:80–85

    PubMed  Google Scholar 

  83. Whyte MP (2008) Enzyme defects and the skeleton. In: Rosen C (ed) Primer on the metabolic bone diseases and disorders of mineral metabolism, 7th edn. American Society for Bone and Mineral Research, Washington, D.C., pp 454–458

    Google Scholar 

  84. Hughes AE, Ralston SH, Marken J, Bell C, MacPherson H, Wallace RG, van Hul W, Whyte MP, Nakatsuka K, Hovy L, Anderson DM (2000) Mutations in TNFRSF11A, affecting the signal peptide of RANK, cause familial expansile osteolysis. Nat Genet 24:45–48

    CAS  PubMed  Google Scholar 

  85. Horn D, Krawitz P, Mannhardt A, Korenke GC, Meinecke P (2011) Hyperphosphatasia-mental retardation syndrome due to PIGV mutations: expanded clinical spectrum. Am J Med Genet A 155A:1917–1922

    PubMed  Google Scholar 

  86. Krawitz PM, Murakami Y, Hecht J, Krüger U, Holder SE, Mortier GR, Delle Chiaie B, De Baere E, Thompson MD, Roscioli T, Kielbasa S, Kinoshita T, Mundlos S, Robinson PN, Horn D (2012) Mutations in PIGO, a member of the GPI-anchor-synthesis pathway, cause hyperphosphatasia with mental retardation. Am J Hum Genet 91:146–151

    CAS  PubMed Central  PubMed  Google Scholar 

  87. Hansen L, Tawamie H, Murakami Y, Mang Y, ur Rehman S, Buchert R, Schaffer S, Muhammad S, Bak M, Nöthen MM, Bennett EP, Maeda Y, Aigner M, Reis A, Kinoshita T, Tommerup N, Baig SM, Abou Jamra R (2013) Hypomorphic mutations in PGAP2, encoding a GPI-anchor-remodeling protein, cause autosomal-recessive intellectual disability. Am J Hum Genet 92:575–583

    CAS  PubMed Central  PubMed  Google Scholar 

  88. Whyte MP, Obrecht SE, Finnegan PM, Jones JL, Podgornik MN, McAlister WH, Mumm S (2002) Osteoprotegerin deficiency and juvenile Paget’s disease. N Engl J Med 347:175–184

    CAS  PubMed  Google Scholar 

  89. Salpea P, Stratakis CA (2014) Carney complex and McCune Albright syndrome: an overview of clinical manifestations and human molecular genetics. Mol Cell Endocrinol 386:85–91

    CAS  PubMed  Google Scholar 

  90. Waaijer CJ, Winter MG, Reijnders CM, de Jong D, John Ham S, Bovée JV, Szuhai K (2013) Intronic deletion and duplication proximal of the EXT1 gene: a novel causative mechanism for multiple osteochondromas. Genes Chromosomes Cancer 52:431–436

    CAS  PubMed  Google Scholar 

  91. Francannet C, Cohen-Tanugi A, Le Merrer M, Munnich A, Bonaventure J, Legeai-Mallet L (2001) Genotype–phenotype correlation in hereditary multiple exostoses. J Med Genet 38:430–434

    CAS  PubMed Central  PubMed  Google Scholar 

  92. Bocciardi R, Bordo D, Di Duca M, Di Rocco M, Ravazzolo R (2009) Mutational analysis of the ACVR1 gene in Italian patients affected with fibrodysplasia ossificans progressiva: confirmations and advancements. Eur J Hum Genet 17:311–318

    CAS  PubMed Central  PubMed  Google Scholar 

  93. Marcocci C, Cetani F (2011) Clinical practice. Primary hyperparathyroidism. N Engl J Med 365:2389–2397

    CAS  PubMed  Google Scholar 

  94. Pepe J, Cipriani C, Pilotto R, De Lucia F, Castro C, Lenge L, Russo S, Guarnieri V, Scillitani A, Carnevale V, D’Erasmo E, Romagnoli E, Minisola S (2011) Sporadic and hereditary primary hyperparathyroidism. J Endocrinol Invest 34:40–44

    CAS  PubMed  Google Scholar 

  95. Thakker RV, Newey PJ, Walls GV, Bilezikian J, Dralle H, Ebeling PR, Melmed S, Sakurai A, Tonelli F, Brandi ML (2012) Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). J Clin Endocrinol Metab 97:2990–3011

    CAS  PubMed  Google Scholar 

  96. Costa-Guda J, Arnold A (2013) Genetic and epigenetic changes in sporadic endocrine tumors: Parathyroid tumors. Mol Cell Endocrinol 386:S0303–S7207

    PubMed  Google Scholar 

  97. Marx SJ (2011) Hyperparathyroid genes: sequences reveal answers and questions. Endocr Pract 3:18–27

    Google Scholar 

  98. Pannett AAJ, Thakker RV (2001) Somatic mutations in MEN type 1 tumors, consistent with the Knudson ‘two-hit’ hypothesis. J Clin Endocrinol Metab 86:4371–4374

    CAS  PubMed  Google Scholar 

  99. Costa-Guda J, Arnold A (2014) Genetic and epigenetic changes in sporadic endocrine tumors: Parathyroid tumors. Mol Cell Endocrinol 386:46–54

    CAS  PubMed  Google Scholar 

  100. Maher ER, Richard S, Gardie B, Couvé S, Gad S (2013) Von Hippel–Lindau: how a rare disease illuminates cancer biology. Semin Cancer Biol 23:26–37

    Google Scholar 

  101. Cetani F, Pardi E, Borsari S, Viacava P, Dipollina G, Cianferotti L, Ambrogini E, Gazzerro E, Colussi G, Berti P, Miccoli P, Pinchera A, Marcocci C (2004) Genetic analyses of the HRPT2 gene in primary hyperparathyroidism: germline and somatic mutations in familial and sporadic parathyroid tumors. J Clin Endocrinol Metab 89:5583–5591

    CAS  PubMed  Google Scholar 

  102. Egbuna OI, Brown EM (2008) Hypercalcaemic and hypocalcaemic conditions due to calcium-sensing receptor mutations. Best Pract Res Clin Rheum 22:129–148

    CAS  Google Scholar 

  103. Marcocci C, Cetani F, Rubin MR, Silverberg SJ, Pinchera A, Bilezikian J (2008) Parathyroid carcinoma. J Bone Miner Res 23:1869–1880

    CAS  PubMed Central  PubMed  Google Scholar 

  104. Hannan FM, Thakker RV (2013) Calcium-sensing receptor (CaSR) mutations and disorders of calcium, electrolyte and water metabolism. Best Pract Res Clin Endocrinol Metab 27:359–371

    CAS  PubMed  Google Scholar 

  105. Nesbit MA, Hannan FM, Howles SA, Babinsky VN, Head RA, Cranston T, Rust N, Hobbs MR, Heath H 3rd, Thakker RV (2013) Mutations affecting G-protein subunit α11 in hypercalcemia and hypocalcemia. N Engl J Med 368:2476–2486

    CAS  PubMed Central  PubMed  Google Scholar 

  106. Hendy GN, Cole DE (2013) Ruling in a suspect: the role of AP2S1 mutations in familial hypocalciuric hypercalcemia type 3. J Clin Endocrinol Metab 98:4666–4669

    CAS  PubMed Central  PubMed  Google Scholar 

  107. Pallais JC, Kifor O, Chen YB, Slovik D, Brown EM (2004) Acquired hypocalciuric hypercalcemia due to autoantibodies against the calcium-sensing receptor. N Engl J Med 22(351):362–369

    Google Scholar 

  108. Fujino T, Watanabe T, Yamaguchi K, Nagasaki K, Onishi E, Iwamoto I, Dozono H, Nagata Y (1992) The development of hypercalcemia in a patient with an ovarian tumor producing parathyroid hormone-related protein. Cancer 70:2845–2850

    CAS  PubMed  Google Scholar 

  109. Botea V, Edelson GW, Munasinghe RL (2003) Hyperparathyroidism, hypercalcemia, and calcified brain metastatic lesions in a patient with small cell carcinoma demonstrating positive immunostain for parathyroid hormone. Endocr Pract 9:40–44

    PubMed  Google Scholar 

  110. Rizzoli R, Pache JC, Didierjean L, Bürger A, Bonjour JP (1994) A thymoma as a cause of true ectopic hyperparathyroidism. J Clin Endocrinol Metab 79:912–915

    CAS  PubMed  Google Scholar 

  111. Brown EM (2009) Anti-parathyroid and anti-calcium sensing receptor antibodies in autoimmune hypoparathyroidism. Endocrinol Metab Clin N Am 38:437–445

    CAS  Google Scholar 

  112. Perheentupa J (2006) Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. J Clin Endocrinol Metab 91:2843–2850

    CAS  PubMed  Google Scholar 

  113. Arnold A, Horst SA, Gardella TJ, Baba H, Levine MA, Kronenberg HM (1990) Mutation of the signal peptide-encoding region of the preproparathyroid hormone gene in familial isolated hypoparathyroidism. J Clin Invest 86:1084–1087

    CAS  PubMed Central  PubMed  Google Scholar 

  114. Pollak MR, Brown EM, Estep HL, McLaine PN, Kifor O, Park J, Hebert SC, Seidman CE, Seidman JG (1994) Autosomal dominant hypocalcaemia caused by a Ca(2+)-sensing receptor gene mutation. Nat Genet 8:303–307

    CAS  PubMed  Google Scholar 

  115. Thakker RV, Davies KE, Whyte MP, Wooding C, Riordan JL (1990) Mapping the gene causing X-linked recessive idiopathic hypoparathyroidism to Xq26–Xq27 by linkage studies. J Clin Invest 86:40–45

    CAS  PubMed Central  PubMed  Google Scholar 

  116. Yagi H, Furutani Y, Hamada H, Sasaki T, Asakawa S, Minoshima S, Ichida F, Joo K, Kimura M, Imamura S, Kamatani N, Momma K, Takao A, Nakazawa M, Shimizu N, Matsuoka R (2003) Role of TBX1 in human del22q11.2 syndrome. Lancet 362:1366–1373

    CAS  PubMed  Google Scholar 

  117. Watanabe T, Mochizuki H, Kohda N, Minamitani K, Minagawa M, Yasuda T, Niimi H (1998) Autosomal dominant familial hypoparathyroidism, sensorineural deafness, and renal dysplasia. Eur J Endocrinol 139:631–634

    CAS  PubMed  Google Scholar 

  118. Kenny FM, Linarelli L (1966) Dwarfism and cortical thickening of tubular bones. Transient hypocalcemia in a mother and son. Am J Dis Child 111:201–207

    CAS  PubMed  Google Scholar 

  119. Marsden D, Nyhan WL, Sakati NO (1994) Syndrome of hypoparathyroidism with growth hormone deficiency and multiple minor anomalies. Am J Med Genet 52:334–338

    CAS  PubMed  Google Scholar 

  120. Baruteau J, Levade T, Redonnet-Vernhet I, Mesli S, Bloom MC, Broué P (2009) Hypoketotic hypoglycemia with myolysis and hypoparathyroidism: an unusual association in medium chain acyl-CoA desydrogenase deficiency (MCADD). J Pediatr Endocrinol Metab 22:1175–1177

    CAS  PubMed  Google Scholar 

  121. Montagna P, Gallassi R, Medori R, Govoni E, Zeviani M, Di Mauro S, Lugaresi E, Andermann F (1998) MELAS syndrome: characteristic migrainous and epileptic features and maternal transmission. Neurology 38:751–754

    Google Scholar 

  122. Spiekerkoetter U, Sun B, Khuchua Z, Bennett MJ, Strauss AW (2003) Molecular and phenotypic heterogeneity in mitochondrial trifunctional protein deficiency due to beta-subunit mutations. Hum Mutat 21:598–607

    CAS  PubMed  Google Scholar 

  123. Matsubara Y, Kraus JP, Yang-Feng TL, Francke U, Rosenberg LE, Tanaka K (1986) Molecular cloning of cDNAs encoding rat and human medium-chain acyl-CoA dehydrogenase and assignment of the gene to human chromosome 1. Proc Nat Acad 83:6543–6547

    CAS  Google Scholar 

  124. Mantovani G, Spada A (2006) Mutations in the Gs alpha gene causing hormone resistance. Bes Prac Res Clin Endocr Metab 20:501–513

    CAS  Google Scholar 

  125. Brandi ML (2011) Genetics of hypoparathyroidism and pseudohypoparathyroidism. J Endocrinol Invest 34:27–34

    CAS  PubMed  Google Scholar 

  126. Farfel Z, Bourne HR, Iiri T (1999) The expanding spectrum of G protein diseases. N Engl J Med 340:1012–1020

    CAS  PubMed  Google Scholar 

  127. Wilson LC, Oude Luttikhuis MEM, Clayton PT, Fraser WD, Trembath RC (1994) Parental origin of Gs-alpha gene mutations in Albright’s hereditary osteodystrophy. J Med Genet 31:835–839

    CAS  PubMed Central  PubMed  Google Scholar 

  128. Calvi LM, Schipani E (2000) The PTH/PTHrP receptor in Jansen’s metaphyseal chondrodysplasia. J Endocrinol Invest 23:545–554

    CAS  PubMed  Google Scholar 

  129. Tanaka H (2010) Cytokines in bone diseases. FGF receptor signaling and achondroplasia/hypochondroplasi. Clin Calcium 20:1490–1496

    PubMed  Google Scholar 

  130. Superti-Furga A, Spranger J, Nishimura G (2012) Enchondromatosis revisited: new classification with molecular basis. Am J Med Genet C Semin Med Genet 15:154–164

    Google Scholar 

  131. Huang-Doran I, Sleigh A, Rochford JJ, O’Rahilly S, Savage DB (2010) Lipodystrophy: metabolic insights from a rare disorder. Endocrinol 207:245–255

    CAS  Google Scholar 

  132. Garg A (2004) Acquired and inherited lipodystrophies. New Eng J Med 350:1220–1234

    CAS  PubMed  Google Scholar 

  133. Garavelli L, D’Apice MR, Rivieri F, Bertoli M, Wischmeijer A, Gelmini C, De Nigris V, Albertini E, Rosato S, Virdis R, Bacchini E, Dal Zotto R, Banchini G, Iughetti L, Bernasconi S, Superti-Furga A, Novelli GA (2009) Mandibuloacral dysplasia type A in childhood. J Med Genet A 149A:2258–2264

    CAS  Google Scholar 

  134. Meas T, Laloi-Michelin M, Virally M, Ambonville C, Kevorkian JP, Guillausseau PJ (2010) Mitochondrial diabetes: clinical features, diagnosis and management. Rev Med Interne 31:216–221

    CAS  PubMed  Google Scholar 

  135. Marais AD, Solomon GA, Blom DJ (2014) Dysbetalipoproteinaemia: a mixed hyperlipidaemia of remnant lipoproteins due to mutations in apolipoprotein E. Crit Rev Clin Lab Sci 51:46–62

    CAS  PubMed  Google Scholar 

  136. Wilcken Bridget (2010) Fatty acid oxidation disorders: outcome and long-term prognosis. J Inherit Metab Dis 33:501–506

    CAS  PubMed  Google Scholar 

  137. Thuillier L, Rostane H, Droin V, Demaugre F, Brivet M, Kadhom N, Prip-Buus C, Gobin S, Saudubray JM, Bonnefont JP (2003) Correlation between genotype, metabolic data, and clinical presentation in carnitine palmitoyltransferase 2 (CPT2) deficiency. Hum Mutat 21:493–501

    CAS  PubMed  Google Scholar 

  138. Lamhonwah AM, Olpin SE, Pollitt RJ, Vianey-Saban C, Divry P, Guffon N, Besley GT, Onizuka R, De Meirleir LJ, Cvitanovic-Sojat L, Baric I, Dionisi-Vici C, Fumic K, Maradin M, Tein I (2002) Novel OCTN2 mutations: no genotype–phenotype correlations: early carnitine therapy prevents cardiomyopathy. Am J Med Genet 111:271–284

    PubMed  Google Scholar 

  139. Bougnères PF, Saudubray JM, Marsac C, Bernard O, Odièvre M, Girard J (1981) Fasting hypoglycemia resulting from hepatic carnitine palmitoyl transferase deficiency. J Pediatr 98:742–746

    PubMed  Google Scholar 

  140. Shirao K, Okada S, Tajima G, Tsumura M, Hara K, Yasunaga S, Ohtsubo M, Hata I, Sakura N, Shigematsu Y, Takihara Y, Kobayashi M (2010) Molecular pathogenesis of a novel mutation, G108D, in short-chain acyl-CoA dehydrogenase identified in subjects with short-chain acyl-CoA dehydrogenase deficiency. Hum Genet 127:619–628

    CAS  PubMed  Google Scholar 

  141. Lee J, Hegele RA (2014) Abetalipoproteinemia and homozygous hypobetalipoproteinemia: a framework for diagnosis and management. J Inherit Metab Dis 37:333–339

    CAS  PubMed  Google Scholar 

  142. Fab Schiffmann R (2009) Fabry disease. Pharmacol Ther 122:65–77

    Google Scholar 

  143. Merkel M, Eckel RH, Goldberg IJ (2002) Lipoprotein lipase: genetics, lipid uptake, and regulation. J Lipid Res 43:1997–2006

    CAS  PubMed  Google Scholar 

  144. Mehta A (2006) Epidemiology and natural history of Gaucher’s disease. Eur J Intern Med 17:S2–S5

    CAS  PubMed  Google Scholar 

  145. Hicks J, Wartchow E, Mierau G (2011) Glycogen storage diseases: a brief review and update on clinical features, genetic abnormalities, pathologic features, and treatment. Ultrastruct Pathol 35:183–196

    PubMed  Google Scholar 

  146. Ligon AH, Moore SD, Parisi MA, Mealiffe ME, Harris DJ, Ferguson HL, Quade BJ, Morton CC (2005) Constitutional rearrangement of the architectural factor HMGA2: a novel human phenotype including overgrowth and lipomas. Am J Hum Genet 76:340–348

    CAS  PubMed Central  PubMed  Google Scholar 

  147. Kaczirek K, Niederle B (2004) Nesidioblastosis: an old term and a new understanding. World J Surg 28:1227–1230

    PubMed  Google Scholar 

  148. Austin MA, Hutter CM, Zimmern RL, Humphries SE (2004) Genetic causes of monogenic heterozygous familial hypercholesterolemia: a HuGE prevalence review. Am J Epidemiol 160:407–420

    PubMed  Google Scholar 

  149. Muenzer J (2011) Overview of the mucopolysaccharidose. Rheumatology 50:v4–v12

    CAS  PubMed  Google Scholar 

  150. Mahoney CP, Weinberger E, Bryant C, Ito M, Jameson JL, Ito M (2002) Effects of aging on vasopressin production in a kindred with autosomal dominant neurohypophyseal diabetes insipidus due to the delta-E47 neurophysin mutation. J Clin Endocrinol Metab 87:870–876

    CAS  PubMed  Google Scholar 

  151. Forssman H (1955) Two different mutations of the X-chromosome causing diabetes insipidus. Am J Hum Genet 7:21–27

    CAS  PubMed Central  PubMed  Google Scholar 

  152. Hofmann S, Philbrook C, Gerbitz KD, Bauer MF (2003) Wolfram syndrome: structural and functional analyses of mutant and wild-type wolframin, the WFS1 gene product. Hum Molec Genet 12:2003–2012

    CAS  PubMed  Google Scholar 

  153. Holtzman EJ, Harris HW Jr, Kolakowski LF Jr, Guay-Woodford LM, Botelho B, Ausiello DA (1993) Brief report: a molecular defect in the vasopressin V2-receptor gene causing nephrogenic diabetes insipidus. N Engl J Med. 328:1534–1537

    CAS  PubMed  Google Scholar 

  154. Deen PM, Croes H, van Aubel RA, Ginsel LA, van Os CH (1995) Water channels encoded by mutant aquaporin-2 genes in nephrogenic diabetes insipidus are impaired in their cellular routing. J Clin Invest 95:2291–2296

    CAS  PubMed Central  PubMed  Google Scholar 

  155. Feldman BJ, Rosenthal SM, Vargas GA, Fenwick RG, Huang EA, Matsuda-Abedini M, Lustig RH, Mathias RS, Portale AA, Miller WL, Gitelman SE (2005) Nephrogenic syndrome of inappropriate antidiuresis. New Eng J Med 352:1884–1890

    CAS  PubMed  Google Scholar 

  156. Fardella CE, Mosso L, Gomez-Sanchez C, Cortes P, Gomez L, Pinto M, Huete A, Oestreicher E, Foradow A, Montero J (2000) Primary hyperaldosteronism in essential hypertensives: prevalence, biochemical profile, and molecular biology. J Clin Endocrinol Metab 85:1863–1867

    CAS  PubMed  Google Scholar 

  157. Lafferty AR, Torpy DJ, Stowasser M, Taymans SE, Lin JP, Huggard P, Gordon RD, Stratakis CA (2000) A novel genetic locus for low renin hypertension: familial hyperaldosteronism type II maps to chromosome 7 (7p22). J Med Genet 37:831–835

    CAS  PubMed Central  PubMed  Google Scholar 

  158. Choi M, Scholl UI, Yue P, Bjorklund P, Zhao B, Nelson-Williams C, Ji W, Cho Y, Patel A, Men CJ, Lolis E, Wisgerhof MV, Geller DS, Mane S, Hellman P, Westin G, Akerstrom G, Wang W, Carling T, Lifton RP (2011) K+ channel mutations in adrenal aldosterone-producing adenomas and hereditary hypertension. Science 331:768–772

    CAS  PubMed Central  PubMed  Google Scholar 

  159. Fava C, Montagnana M, Rosberg L, Burri P, Almgren P, Jonsson A, Wanby P, Lippi G, Minuz P, Hulthen LU, Aurell M, Melander O (2008) Subjects heterozygous for genetic loss of function of the thiazide-sensitive cotransporter have reduced blood pressure. Hum Mol Genet 17:413–418

    CAS  PubMed  Google Scholar 

  160. Deschenes G, Burguet A, Guyot C, Hubert P, Garabedian M, Dechaux M, Loirat C, Broyer M (1993) Forme antenatale de syndrome de Bartter. Ann Pediat 40:95–101

    CAS  PubMed  Google Scholar 

  161. Finer G, Shalev H, Birk OS, Galron D, Jeck N, Sinai-Treiman L, Landau D (2003) Transient neonatal hyperkalemia in the antenatal (ROMK defective) Bartter syndrome. J Pediat 142:318–323

    CAS  PubMed  Google Scholar 

  162. Nozu K, Inagaki T, Fu XJ, Nozu Y, Kaito H, Kanda K, Sekine T, Igarashi T, Nakanishi K, Yoshikawa N, Iijima K, Matsuo M (2008) Molecular analysis of digenic inheritance in Bartter syndrome with sensorineural deafness. J Med Genet 45:182–186

    CAS  PubMed  Google Scholar 

  163. Arant BS, Brackett NC Jr, Young RB, Still WJS (1970) Case studies of siblings with juxtaglomerular hyperplasia and secondary aldosteronism associated with severe azotemia and renal rickets—Bartter’s syndrome or disease? Pediatrics 46:344–361

    CAS  PubMed  Google Scholar 

  164. Shimkets RA, Warnock DG, Bositis CM, Nelson-Williams C, Hansson JH, Schambelan M, Gill JR Jr, Ulick S, Milora RV, Findling JW, Canessa CM, Rossier BC, Lifton RP (1994) Liddle’s syndrome: heritable human hypertension caused by mutations in the beta subunit of the epithelial sodium channel. Cell 79:407–414

    CAS  PubMed  Google Scholar 

  165. Mansfield TA, Simon DB, Farfel Z, Bia M, Tucci JR, Lebel M, Gutkin M, Vialettes B, Christofills MA, Kauppinen-Makelin R, Mayan H, Risch N, Lifton RP (1997) Multilocus linkage of familial hyperkalaemia and hypertension, pseudohypoaldosteronism type II, to chromose 1q31-42 and 17p11-q21. Nat Genet 16:202–205

    CAS  PubMed  Google Scholar 

  166. Ulick S, Levine LS, Gunczler P, Zanconato G, Ramirez C, Rauh W, Rosler A, Bradlow HL, New MI (1979) A syndrome of apparent mineralocorticoid excess asociated with defects in the peripheral metabolism of cortisol. J Clin Endocrinol Metab 49:757–764

    CAS  PubMed  Google Scholar 

  167. Gimenez-Roqueplo AP, Dahia PL, Robledo M (2012) An update on the genetics of paraganglioma, pheochromocytoma, and associated hereditary syndromes. Horm Metab Res 44:328–333

    CAS  PubMed  Google Scholar 

  168. Bausch B, Borozdin W, Mautner VF, Hoffmann MM, Boehm D, Robledo M, Cascon A, Harenberg T, Schiavi F, Pawlu C, Peczkowska M, Letizia C, Calvieri S, Arnaldi G, Klingenberg-Noftz RD, Reisch N, Fassina A, Brunaud L, Walter MA, Mannelli M, MacGregor G, Palazzo FF, Barontini M, Walz MK, Kremens B, Brabant G, Pfäffle R, Koschker AC, Lohoefner F, Mohaupt M, Gimm O, Jarzab B, McWhinney SR, Opocher G, Januszewicz A, Kohlhase J, Eng C, Neumann HP, European-American Phaeochromocytoma Registry Study Group (2007) Germline NF1 mutational spectra and loss-of-heterozygosity analyses in patients with pheochromocytoma and neurofibromatosis type 1. J Clin Endocrinol Metab 92:2784–2792

    CAS  PubMed  Google Scholar 

  169. Yao L, Schiavi F, Cascon A, Qin Y, Inglada-Pérez L, King EE, Toledo RA, Ercolino T, Rapizzi E, Ricketts CJ, Mori L, Giacchè M, Mendola A, Taschin E, Boaretto F, Loli P, Iacobone M, Rossi GP, Biondi B, Lima-Junior JV, Kater CE, Bex M, Vikkula M, Grossman AB, Gruber SB, Barontini M, Persu A, Castellano M, Toledo SP, Maher ER, Mannelli M, Opocher G, Robledo M, Dahia PL (2010) Spectrum and prevalence of FP/TMEM127 gene mutations in pheochromocytomas and paragangliomas. JAMA 304:2611–2619

    CAS  PubMed  Google Scholar 

  170. Burnichon N, Cascón A, Schiavi F, Morales NP, Comino-Méndez I, Abermil N, Inglada-Pérez L, de Cubas AA, Amar L, Barontini M, de Quirós SB, Bertherat J, Bignon YJ, Blok MJ, Bobisse S, Borrego S, Castellano M, Chanson P, Chiara MD, Corssmit EP, Giacchè M, de Krijger RR, Ercolino T, Girerd X, Gómez-García EB, Gómez-Graña A, Guilhem I, Hes FJ, Honrado E, Korpershoek E, Lenders JW, Letón R, Mensenkamp AR, Merlo A, Mori L, Murat A, Pierre P, Plouin PF, Prodanov T, Quesada-Charneco M, Qin N, Rapizzi E, Raymond V, Reisch N, Roncador G, Ruiz-Ferrer M, Schillo F, Stegmann AP, Suarez C, Taschin E, Timmers HJ, Tops CM, Urioste M, Beuschlein F, Pacak K, Mannelli M, Dahia PL, Opocher G, Eisenhofer G, Gimenez-Roqueplo AP, Robledo M (2012) MAX mutations cause hereditary and sporadic pheochromocytoma and paraganglioma. Clin Cancer Res 18:2828–2837

    CAS  PubMed  Google Scholar 

  171. Gimenez-Roqueplo AP, Caumont-Prim A, Houzard C, Hignette C, Hernigou A, Halimi P, Niccoli P, Leboulleux S, Amar L, Borson-Chazot F, Cardot-Bauters C, Delemer B, Chabolle F, Coupier I, Libé R, Peitzsch M, Peyrard S, Tenenbaum F, Plouin PF, Chatellier G, Rohmer V (2013) Imaging work-up for screening of paraganglioma and pheochromocytoma in SDHx mutation carriers: a multicenter prospective study from the PGL. EVA Investigators. J Clin Endocrinol Metab 98:E162–E173

    CAS  PubMed  Google Scholar 

  172. Hao HX, Khalimonchuk O, Schraders M, Dephoure N, Bayley JP, Kunst H, Devilee P, Cremers CW, Schiffman JD, Bentz BG, Gygi SP, Winge DR, Kremer H, Rutter J (2009) SDH5, a gene required for flavination of succinate dehydrogenase, is mutated in paraganglioma. Science 325:1139–1142

    CAS  PubMed  Google Scholar 

  173. Niemann S, Müller U (2000) Mutations in SDHC cause autosomal dominant paraganglioma, type 3. Nat Genet 26:268–270

    CAS  PubMed  Google Scholar 

  174. Carney JA, Stratakis CA (2002) Familial paraganglioma and gastric stromal sarcoma: a new syndrome distinct from the Carney triad. Am J Med Genet 108:132–139

    PubMed  Google Scholar 

  175. Carney JA (1999) Gastric stromal sarcoma, pulmonary chondroma, and extra-adrenal paraganglioma (Carney Triad): natural history, adrenocortical component, an possible familial occurrence. Mayo Clin Proc 74:543–552

    CAS  PubMed  Google Scholar 

  176. White PC, Speiser PW (2000) Congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Endocr Rev 21:245–291

    CAS  PubMed  Google Scholar 

  177. Joehrer K, Geley S, Strasser-Wozak EM, Azziz R, Wollmann HA, Schmitt K, Kofler R, White PC (1997) CYP11B1 mutations causing non-classic adrenal hyperplasia due to 11 β-hydroxylase deficiency. Hum Mol Genet 6:1829–1834

    CAS  PubMed  Google Scholar 

  178. Auchus RJ (2001) The genetics, pathophysiology, and management of human deficiencies of P450c17. Endocrinol Metab Clin N Am 30:101–119

    CAS  Google Scholar 

  179. Simard J, Ricketts ML, Gingras S, Soucy P, Feltus FA, Melner MH (2005) Molecular biology of the 3β-hydroxysteroid dehydrogenase/δ5–δ4 isomerase gene family. Endocr Rev 26:525–582

    CAS  PubMed  Google Scholar 

  180. Arlt W, Walker EA, Draper N, Ivison HE, Ride JP, Hammer F, Chalder SM, Borucka-Mankiewicz M, Hauffa BP, Malunowicz EM, Stewart PM, Shackleton CHL (2004) Congenital adrenal hyperplasia caused by mutant P450 oxidoreductase and human androgen synthesis: analytical study. Lancet 363:2128–2135

    CAS  PubMed  Google Scholar 

  181. Bose HS, Sugawara T, Strauss JF, Miller WL (1996) The pathophysiology and genetics of congenital lipoid adrenal hyperplasia. International congenital lipoid adrenal hyperplasia consortium. N Engl J Med 335:1870–1878

    CAS  PubMed  Google Scholar 

  182. Kim CJ, Lin L, Huang N, Quigley CA, AvRuskin TW, Achermann JC, Miller WL (2008) Severe combined adrenal and gonadal deficiency caused by novel mutations in the cholesterol side chain cleavage enzyme, P450scc. J Clin Endocrinol Metab 93:696–702

    CAS  PubMed Central  PubMed  Google Scholar 

  183. Bertagna X, Guignat L (2013) Approach to the Cushing’s disease patient with persistent/recurrent hypercortisolism after pituitary surgery. J Clin Endocrinol Metab 98:1307–1318

    CAS  PubMed  Google Scholar 

  184. Stratakis CA, Bertherat J, Carney JA (2004) Mutation of perinatal myosin heavy chain. N Engl J Med 351:2556–2558

    CAS  PubMed  Google Scholar 

  185. Libé R, Horvath A, Vezzosi D, Fratticci A, Coste J, Perlemoine K, Ragazzon B, Guillaud-Bataille M, Groussin L, Clauser E, Raffin-Sanson ML, Siegel J, Moran J, Drori-Herishanu L, Faucz FR, Lodish M, Nesterova M, Bertagna X, Bertherat J, Stratakis CA (2011) Frequent phosphodiesterase 11A gene (PDE11A) defects in patients with Carney complex (CNC) caused by PRKAR1A mutations: PDE11A may contribute to adrenal and testicular tumors in CNC as a modifier of the phenotype. J Clin Endocrinol Metab 96:E208–E214

    PubMed Central  PubMed  Google Scholar 

  186. Birch JM, Alston RD, McNally RJ, Evans DG, Kelsey AM, Harris M, Eden OB, Varley JM (2001) Relative frequency and morphology of cancers in carriers of germline TP53 mutations. Oncogene 20:4621–4628

    CAS  PubMed  Google Scholar 

  187. Vahteristo P, Tamminen A, Karvinen P, Eerola H, Eklund C, Aaltonen LA, Blomqvist C, Aittomaki K, Nevanlinna H (2001) p53, CHK2, and CHK1 genes in Finnish families with Li–Fraumeni syndrome: further evidence of CHK2 in inherited cancer predisposition. Cancer Res 61:5718–5722

    CAS  PubMed  Google Scholar 

  188. Bachinski LL, Olufemi SE, Zhou X, Wu CC, Yip L, Shete S, Lozano G, Amos CI, Strong LC, Krahe R (2005) Genetic mapping of a third Li–Fraumeni syndrome predisposition locus to human chromosome 1q23. Cancer Res 65:427–431

    CAS  PubMed  Google Scholar 

  189. Eggermann T, Algar E, Lapunzina P, Mackay D, Maher ER, Mannens M, Netchine I, Prawitt D, Riccio A, Temple IK, Weksberg R (2014) Clinical utility gene card for: Beckwith–Wiedemann syndrome. Eur J Hum Genet. doi:10.1038/ejhg.2013.132

    Google Scholar 

  190. Christopoulos S, Bourdeau I, Lacroix A (2005) Clinical and subclinical ACTH-independent macronodular adrenal hyperplasia and aberrant hormone receptors. Horm Res 64:119–131

    CAS  PubMed  Google Scholar 

  191. Tadjine M, Lampron A, Ouadi L, Horvath A, Stratakis CA, Bourdeau I (2008) Detection of somatic beta-catenin mutations in primary pigmented nodular adrenocortical disease (PPNAD). Clin Endocrinol (Oxf) 69:367–373

    CAS  Google Scholar 

  192. Arlt W, Allolio B (2003) Adrenal insufficiency. Lancet 361:1881–1893

    CAS  PubMed  Google Scholar 

  193. Lalli E, Sassone-Corsi P (1999) DAX-1 and the adrenal cortex. Curr Opin Endocrinol Metab 6:185–190

    CAS  Google Scholar 

  194. Laureti S, Casucci G, Santeusanio F, Angeletti G, Aubourg P, Brunetti P (1996) X-linked adrenoleukodystrophy is a frequent cause of idiopathic Addison’s disease in young adult male patients. J Clin Endocrinol Metab 81:470–474

    CAS  PubMed  Google Scholar 

  195. Metherell LA, Chapple JP, Cooray S, David A, Becker C, Rüschendorf F, Naville D, Begeot M, Khoo B, Nürnberg P, Huebner A, Cheetham ME, Clarket AJL (2005) Mutations in MRAP, encoding a new interacting partner of the ACTH receptor, cause familial glucocorticoid deficiency type 2. Nat Genet 37:166–170

    CAS  PubMed  Google Scholar 

  196. Gicquel C, Le Bouc Y, Luton JP, Bertagna X (1998) Pathogenesis and treatment of adrenocortical carcinoma. Curr Opin Endocrinol Diab 5:189–196

    CAS  Google Scholar 

  197. Gonzalez L, Witchel S-F (2012) The patient with Turner syndrome: puberty and medical management concerns. Fertil Steril 98:780–786

    PubMed Central  PubMed  Google Scholar 

  198. Agarwal P, Philip R, Gutch M, Gupta KK (2013) The other side of Turner’s: Noonan’s syndrome. Indian J Endocrinol Metab 17:794–798

    PubMed Central  PubMed  Google Scholar 

  199. Rafique S, Sterling EW, Nelson LM (2012) A new approach to primary ovarian insufficiency. Obstet Gynecol Clinc N Am 39:567–586

    Google Scholar 

  200. Kousta E, Papathanasiou A, Skordis N (2010) Sex determination and disorders of sex development according to the revised nomenclature and classification in 46, XX individuals. Hormones (Athens) 9:218–231

    Google Scholar 

  201. Young RH, Scully RE (1985) Ovarian Sertoli–Leydig cell tumors. A clinicopathological analysis of 207 cases. Am J Surg Pathol 9:543–569

    CAS  PubMed  Google Scholar 

  202. Young RH, Dickersin GR, Scully RE (1984) Juvenile granulosa cell tumor of the ovary. A clinicopathological analysis of 125 cases. Am J Surg Pathol 8:575–596

    CAS  PubMed  Google Scholar 

  203. Stettner AR, Hartenbach EM, Schink JC, Huddart R, Becker J, Pauli R, Long R, Laxova R (1999) A Familial ovarian germ cell cancer: report and review. J Med Genet 7:43–46

    Google Scholar 

  204. Vorona E, Zitzmann M, Gromoll J, Schüring AN, Nieschlag E (2007) Clinical, endocrinological, and epigenetic features of the 46, XX male syndrome, compared with 47, XXY Klinefelter patients. J Clin Endocrinol Metab 92:3458–3465

    CAS  PubMed  Google Scholar 

  205. Udd B, Krahe R (2012) The myotonic dystrophies: molecular, clinical, and therapeutic challenges. Lancet Neurol 11:891–905

    CAS  PubMed  Google Scholar 

  206. Latronico AC, Arnhold IJ (2012) Inactivating mutations of the human luteinizing hormone receptor in both sexes. Semin Reprod Med 30:382–386

    CAS  PubMed  Google Scholar 

  207. Latronico AC, Arnhold IJ (2013) Gonadotropin resistance. Endocr Dev 24:25–32

    CAS  PubMed  Google Scholar 

  208. Kemp S, Berger J, Aubourg P (2012) X-linked adrenoleukodystrophy: clinical, metabolic, genetic and pathophysiological aspects. Biochim Biophys Acta 1822:1465–1474

    CAS  PubMed  Google Scholar 

  209. Pirgon Ö, Dündar BN (2012) Vanishing testes: a literature review. J Clin Res Pediatr Endocrinol 4:116–120

    PubMed Central  PubMed  Google Scholar 

  210. Ferlin A, Arredi B, Speltra E, Cazzadore C, Selice R, Garolla A, Lenzi A, Foresta C (2007) Molecular and clinical characterization of Y chromosome microdeletions in infertile men: a 10-year experience in Italy. J Clin Endocrinol Metab 9:762–770

    Google Scholar 

  211. Toledo SPA (1992) Leydig cell hypoplasia leading to two different phenotypes: male pseudohermaphroditism and primary hypogonadism not associated with this. (Lett) Clin Endocr 36:521–522

    CAS  Google Scholar 

  212. Nowaczyk MJ, Irons MB (2012) Smith-Lemli–Opitz syndrome: phenotype, natural history, and epidemiology. Am J Med Genet C Semin Med Genet 160C:250–262

    PubMed  Google Scholar 

  213. King SR, Bhangoo A, Stocco DM (2011) Functional and physiological consequences of StAR deficiency: role in lipoid congenital adrenal hyperplasia. Endocr Dev 20:47–53

    CAS  PubMed  Google Scholar 

  214. Hiort O, Holterhus PM, Werner R, Marschke C, Hoppe U, Partsch CJ, Riepe FG, Achermann JC, Struve D (2005) Homozygous disruption of P450 side-chain cleavage (CYP11A1) is associated with prematurity, complete 46, XY sex reversal, and severe adrenal failure. J Clin Endocrinol Metab 90:538–541

    CAS  PubMed  Google Scholar 

  215. Krone N, Arlt W (2009) Genetics of congenital adrenal hyperplasia. Best Pract Res Clin Endocrinol Metab 23:181–192

    CAS  PubMed  Google Scholar 

  216. George MM, New MI, Ten S, Sultan C, Bhangoo A (2010) The clinical and molecular heterogeneity of 17βHSD-3 enzyme deficiency. Horm Res Paediatr 74:229–240

    CAS  PubMed  Google Scholar 

  217. Costa EM, Domenice S, Sircili MH, Inacio M, Mendonca BB (2012) DSD due to 5α-reductase 2 deficiency—from diagnosis to long term outcome. Semin Reprod Med 30:427–431

    CAS  PubMed  Google Scholar 

  218. Hughes IA, Davies JD, Bunch TI, Pasterski V, Mastroyannopoulou K, MacDougall J (2012) Androgen insensitivity syndrome. Lancet 380:1419–1428

    CAS  PubMed  Google Scholar 

  219. Hughes IA, Werner R, Bunch T, Hiort O (2012) Androgen insensitivity syndrome. Semin Reprod Med 30:432–442

    CAS  PubMed  Google Scholar 

  220. Monks DA, Rao P, Mo K, Johansen JA, Lewis G, Kemp MQ (2008) Androgen receptor and Kennedy disease/spinal bulbar muscular atrophy. Horm Behav 53:729–740

    CAS  PubMed Central  PubMed  Google Scholar 

  221. Massanyi EZ, Dicarlo HN, Migeon CJ, Gearhart JP (2013) Review and management of 46, XY disorders of sex development. J Pediatr Urol 9:368–379

    PubMed  Google Scholar 

  222. Ferraz-de-Souza B, Lin L, Achermann JC (2011) Steroidogenic factor-1 (SF-1, NR5A1) and human disease. Mol Cell Endocrinol 336:198–205

    CAS  PubMed Central  PubMed  Google Scholar 

  223. Mendonca BB, Domenice S, Arnhold IJ, Costa EM (2009) 46, XY disorders of sex development (DSD). Clin Endocrinol (Oxf) 70:173–187

    CAS  Google Scholar 

  224. Foster JW (1996) Mutations in SOX9 cause both autosomal sex reversal and campomelic dysplasia. Acta Paediatr Jpn 38:405–411

    CAS  PubMed  Google Scholar 

  225. Koziell A, Charmandari E, Hindmarsh PC, Rees L, Scambler P, Brook CG (2000) Frasier syndrome, part of the Denys Drash continuum or simply a WT1 gene associated disorder of intersex and nephropathy? Clin Endocrinol (Oxf) 52:519–524

    CAS  Google Scholar 

  226. Baets J, Dierick I, Groote CC, Ende JV, Martin JJ, Geens K, Robberecht W, Nelis E, Timmerman V, Jonghe PD (2009) Peripheral neuropathy and 46XY gonadal dysgenesis: a heterogeneous entity. Neuromuscul Disord 19:172–175

    PubMed  Google Scholar 

  227. Horwich A, Nicol D, Huddart R (2013) Testicular germ cell tumours. BMJ 347:f5526

    PubMed  Google Scholar 

  228. Olivier P, Simoneau-Roy J, Francoeur D, Sartelet H, Parma J, Vassart G, Van Vliet G (2012) Leydig cell tumors in children: contrasting clinical, hormonal, anatomical, and molecular characteristics in boys and girls. J Pediatr 161:1147–1152

    PubMed  Google Scholar 

  229. Gómez García I, Romero Molina M, López-García Moreno A, Buendía González E, Rubio Hidalgo E, Bolufer E, Sampietro Crespo A, Gómez Rodríguez A (2010) Sertoli cell tumor, a rare testicular tumor, our experience and review of the literature. Arch Esp Urol 63:392–395

    PubMed  Google Scholar 

  230. Lau YF (1999) Gonadoblastoma, testicular and prostate cancers, and the TSPY gene. Am J Hum Genet 64:921–927

    CAS  PubMed Central  PubMed  Google Scholar 

  231. Curatolo P, Bombardieri R, Jozwiak S (2008) Tuberous sclerosis. Lancet 372:657–668

    CAS  PubMed  Google Scholar 

  232. Jadhav U, Harris RM, Jameson JL (2011) Hypogonadotropic hypogonadism in subjects with DAX1 mutations. Mol Cell Endocrinol 346:65–73

    CAS  PubMed Central  PubMed  Google Scholar 

  233. Rothenbuhler A, Stratakis CA (2010) Clinical and molecular genetics of Carney complex. Best Pract Res Clin Endocrinol Metab 24:389–399

    CAS  PubMed  Google Scholar 

  234. Weiler FG, Dias-da-Silva MR, Lazaretti-Castro M (2012) Autoimmune polyendocrine syndrome type 1: case report and review of literature. Arq Bras Endocrinol Metabol 56:54–66

    PubMed  Google Scholar 

  235. Proust-Lemoine E, Saugier-Veber P, Wémeau JL (2012) Polyglandular autoimmune syndrome type I. Presse Med 41:e651–e662

    PubMed  Google Scholar 

  236. Betterle C, Zanchetta R (2003) Update on autoimmune polyendocrine syndromes (APS). Acta Biomed 74:9–33

    PubMed  Google Scholar 

  237. Michels AW, Gottlieb PA (2010) Autoimmune polyglandular syndromes. Nat Rev Endocrinol 6:270–277

    CAS  PubMed  Google Scholar 

  238. Ghaferi AA, Chojnacki KA, Long WD, Cameron JL, Yeo CJ (2008) Pancreatic VIPomas: subject review and one institutional experience. J Gastrointest Surg 12:382–393

    PubMed  Google Scholar 

  239. Kanakis G, Kaltsas G (2012) Biochemical markers for gastroenteropancreatic neuroendocrine tumours (GEP-NETs). Best Pract Res Clin Gastroenterol 26:791–802

    CAS  PubMed  Google Scholar 

  240. Nesi G, Marcucci T, Rubio CA, Brandi ML, Tonelli F (2008) Somatostatinoma: clinico-pathological features of three cases and literature reviewed. J Gastroenterol Hepatol 23:521–526

    CAS  PubMed  Google Scholar 

  241. Eldor R, Glaser B, MeravFraenkel M, Doviner V, Salmon A, Gross DJ (2011) Glucagonoma and the glucagonoma syndrome—cumulative experience with an elusive endocrine tumour. Clin Endocrinol 74:593–598

    Google Scholar 

  242. Balachandran A, Tamm EP, Bhosale PR, Patnana M, Vikram R, Fleming JB, Katz MH, Charnsangavej C (2013) Pancreatic neuroendocrine neoplasms: diagnosis and management. Abdom Imaging 38:342–357

    PubMed  Google Scholar 

  243. Amador Cano A, García F, Espinoza A, Bezies N, Herrera H, De Leija Portilla J (2013) Nonfunctional neuroendocrine tumor of the pancreas: case report and review of the literature. Int J Surg Case Rep 4:225–228

    CAS  PubMed Central  PubMed  Google Scholar 

  244. Faggiano A, Ferolla P, Grimaldi F, Campana D, Manzoni M, Davì MV, Bianchi A, Valcavi R, Papini E, Giuffrida D, Ferone D, Fanciulli G, Arnaldi G, Franchi GM, Francia G, Fasola G, Crinò L, Pontecorvi A, Tomassetti P, Colao A (2012) Natural history of gastro-entero-pancreatic and thoracic neuroendocrine tumors. Data from a large prospective and retrospective Italian epidemiological study: the NET management study. J Endocrinol Invest 35:817–823

    CAS  PubMed  Google Scholar 

  245. Gibril F, Jensen RT (2005) Advances in valuation and management of gastrinoma in patients with Zollinger–Ellison syndrome. Curr Gastroenterol Rep 7:114–121

    PubMed  Google Scholar 

  246. O’Toole D, Delle Fave G, Jensen RT (2012) Gastric and duodenal neuroendocrine tumours. Best Pract Res Clin Gastroenterol 26:719–735

    PubMed  Google Scholar 

  247. Fraenkel M, Kim MK, Tonelli F, Giudici F, Giusti F, Brand ML (2012) Gastroenteropancreatic neuroendocrine tumors in multiple endocrine neoplasia Type 1. Cancers 4:504–522

    Google Scholar 

  248. Cryer PE, Axelrod L, Grossman AB, Heller SR, Montori VM, Seaquist ER, Service FJ (2009) Evaluation and management of adult hypoglycemic disorders: an Endocrine Society Clinical practice guideline. J Clin Endocrinol Metab 94:709–728

    CAS  PubMed  Google Scholar 

  249. Strosberg J (2012) Neuroendocrine tumours of the small intestine. Best Practice Res Clin Gastroenterol 26:755–773

    CAS  Google Scholar 

  250. Mandair D, Caplin ME (2012) Colonic and rectal NET’s. Best Practice Res Clin Gastroenterol 26:775–789

    Google Scholar 

  251. Francis JM, Kiezun A, Ramos AH, Serra S, SekharPedamallu C, Qian ZR, Banck MS, Kanwar R, Kulkarni AA, Karpathakis A, Manzo V, Contractor T, Philips J, Nickerson E, Pho N, Hooshmand SM, Brais LK, Lawrence MS, Pugh T, McKenna A, Sivachenko A, Cibulskis K, Carter SL, Ojesina AI, Freeman S, Jones RT, Voet D, Auclair D, Saksena G, Onofrio R, Shefler E, Sougnez C, Grimsby J, Green L, Lennon N, Meyer T, Caplin M, Chung DC, Beutler AS, Ogino S, Thirlwell C, Shivdasani R, Asa SL, Harris CR, Getz G, Kulke M, Meyerson M (2013) Somatic mutation of CDKN1B in small intestine neuroendocrine tumors. Nat Genet 45:12

    Google Scholar 

  252. Chopin-Laly X, Walter T, Hervieu V, Poncet G, Adham M, Guibal A, Chayvialle JA, Lombard-Bohas C, Scoazec GY (2013) Neuroendocrine neoplasms of the jejunum: a heterogeneous group with distinctive proximal and distal subsets. Virchows Arch 462:489–499

    PubMed  Google Scholar 

  253. Albores-Saavedra J, Batich K, Chable-Montero F, Sagy N, Schwartz AM, Henson DE (2010) Merkel cell carcinoma demographics, morphology, and survival based on 3870 cases: a population based study. J Cutan Pathol 37:20–27

    PubMed  Google Scholar 

Download references

Acknowledgments

This work was made possible through an unrestricted grant from F.I.R.M.O. Fondazione Raffaella Becagli.

Conflict of interest

All authors have no conflict of interest to disclose.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to G. Marcucci.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Marcucci, G., Cianferotti, L., Beck-Peccoz, P. et al. Rare diseases in clinical endocrinology: a taxonomic classification system. J Endocrinol Invest 38, 193–259 (2015). https://doi.org/10.1007/s40618-014-0202-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40618-014-0202-6

Keywords

Navigation