Skip to main content

Molecular Diagnosis of Genodermatoses

  • Protocol
  • First Online:
Molecular Dermatology

Part of the book series: Methods in Molecular Biology ((MIMB,volume 961))

Abstract

The progress of molecular genetics helps clinicians to prove or exclude a suspected diagnosis for a vast and yet increasing number of genodermatoses. This leads to precise genetic counselling, prenatal diagnosis and preimplantation genetic haplotyping for many inherited skin conditions. It is also helpful in such occasions as phenocopy, late onset and incomplete penetrance, uniparental disomy, mitochondrial inheritance and pigmentary mosaicism. Molecular methods of two genodermatoses are explained in detail, i.e. genodermatoses with skin fragility and neurofibromatosis type 1.

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

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 159.00
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. McKusick VA (2007) Mendelian Inheritance in Man and its online version, OMIM. Am J Hum Genet 80:588–604

    Article  PubMed  CAS  Google Scholar 

  2. Wells RS, Kerr CB (1966) Clinical features of autosomal dominant and sex-linked ichthyosis in an English population. Br Med J 1:947–950

    Article  PubMed  CAS  Google Scholar 

  3. Wells RS (1966) Ichthyosis. Br Med J 2:1504–1506

    Article  PubMed  CAS  Google Scholar 

  4. Shen MH, Harper PS, Upadhyaya M (1996) Molecular genetics of neurofibromatosis type 1 (NF1). J Med Genet 33:2–17

    Article  PubMed  CAS  Google Scholar 

  5. Williams VC, Lucas J, Babcock MA, Gutmann DH, Korf B, Maria BL (2009) Neurofibromatosis type 1 revisited. Pediatrics 123:124–133

    Article  PubMed  Google Scholar 

  6. Bolling MC, Veenstra MJ, Jonkman MF, Diercks GF, Curry CJ, Fisher J, Pas HH, Bruckner AL (2010) Lethal acantholytic epidermolysis bullosa due to a novel homozygous deletion in DSP: expanding the phenotype and implications for desmoplakin function in skin and heart. Br J Dermatol 162:1388–1394

    Article  PubMed  CAS  Google Scholar 

  7. Hobbs RP, Han SY, van der Zwaag PA, Bolling MC, Jongbloed JD, Jonkman MF, Getsios S, Paller AS, Green KJ (2010) Insights from a desmoplakin mutation identified in lethal acantholytic epidermolysis bullosa. J Invest Dermatol 130:2680–2683

    Article  PubMed  CAS  Google Scholar 

  8. Jonkman MF, Pasmooij AM, Pasmans SG, van den Berg MP, Ter Horst HJ, Timmer A, Pas HH (2005) Loss of desmoplakin tail causes lethal acantholytic epidermolysis bullosa. Am J Hum Genet 77:653–660

    Article  PubMed  CAS  Google Scholar 

  9. Shimizu H, Hammami-Hauasli N, Hatta N, Nishikawa T, Bruckner-Tuderman L (1999) Compound heterozygosity for silent and dominant glycine substitution mutations in COL7A1 leads to a marked transient intracytoplasmic retention of procollagen VII and a moderately severe dystrophic epidermolysis bullosa phenotype. J Invest Dermatol 113:419–421

    Article  PubMed  CAS  Google Scholar 

  10. Sprecher E, Ishida-Yamamoto A, Becker OM, Marekov L, Miller CJ, Steinert PM, Neldner K, Richard G (2001) Evidence for novel functions of the keratin tail emerging from a mutation causing ichthyosis hystrix. J Invest Dermatol 116:511–519

    Article  PubMed  CAS  Google Scholar 

  11. Lee JY, Pulkkinen L, Liu HS, Chen YF, Uitto J (1997) A glycine-to-arginine substitution in the triple-helical domain of type VII collagen in a family with dominant dystrophic epidermolysis bullosa pruriginosa. J Invest Dermatol 108:947–949

    Article  PubMed  CAS  Google Scholar 

  12. Wessagowit V, Ashton GH, Mohammedi R, Salas-Alanis JC, Denyer JE, Mellerio JE, Eady RA, McGrath JA (2001) Three cases of de novo dominant dystrophic epidermolysis bullosa associated with the mutation G2043R in COL7A1. Clin Exp Dermatol 26:97–99

    Article  PubMed  CAS  Google Scholar 

  13. Lachmann HJ, Booth DR, Booth SE, Bybee A, Gilbertson JA, Gillmore JD, Pepys MB, Hawkins PN (2002) Misdiagnosis of hereditary amyloidosis as AL (primary) amyloidosis. N Engl J Med 346:1786–1791

    Article  PubMed  CAS  Google Scholar 

  14. Hund E, Linke RP, Willig F, Grau A (2001) Transthyretin-associated neuropathic amyloidosis. Pathogenesis and treatment. Neurology 56:431–435

    Article  PubMed  CAS  Google Scholar 

  15. Perfetto F, Moggi-Pignone A, Livi R, Tempestini A, Bergesio F, Matucci-Cerinic M (2010) Systemic amyloidosis: a challenge for the rheumatologist. Nat Rev Rheumatol 6:417–429

    Article  PubMed  CAS  Google Scholar 

  16. Heathcote K, Syrris P, Carter ND, Patton MA (2000) A connexin 26 mutation causes a syndrome of sensorineural hearing loss and palmoplantar hyperkeratosis (MIM 148350). J Med Genet 37:50–51

    Article  PubMed  CAS  Google Scholar 

  17. Reid FM, Vernham GA, Jacobs HT (1994) A novel mitochondrial point mutation in a maternal pedigree with sensorineural deafness. Hum Mutat 3:243–247

    Article  PubMed  CAS  Google Scholar 

  18. Robinson WP (2000) Mechanisms leading to uniparental disomy and their clinical consequences. Bioessays 22:452–459

    Article  PubMed  CAS  Google Scholar 

  19. Fassihi H, Wessagowit V, Ashton GH, Moss C, Ward R, Denyer J, Mellerio JE, McGrath JA (2005) Complete paternal uniparental isodisomy of chromosome 1 resulting in Herlitz junctional epidermolysis bullosa. Clin Exp Dermatol 30:71–74

    Article  PubMed  CAS  Google Scholar 

  20. Fassihi H, Lu L, Wessagowit V, Ozoemena LC, Jones CA, Dopping-Hepenstal PJ, Foster L, Atherton DJ, Mellerio JE, McGrath JA (2006) Complete maternal isodisomy of chromosome 3 in a child with recessive dystrophic epidermolysis bullosa but no other phenotypic abnormalities. J Invest Dermatol 126:2039–2043

    Article  PubMed  CAS  Google Scholar 

  21. Taibjee SM, Bennett DC, Moss C (2004) Abnormal pigmentation in hypomelanosis of Ito and pigmentary mosaicism: the role of pigmentary genes. Br J Dermatol 151:269–282

    Article  PubMed  CAS  Google Scholar 

  22. Horn D, Rommeck M, Sommer D, Korner H (1997) Phylloid pigmentary pattern with mosaic trisomy 13. Pediatr Dermatol 14:278–280

    Article  PubMed  CAS  Google Scholar 

  23. Lipsker D, Flory E, Wiesel ML, Hanau D, de la Salle H (2008) Between light and dark, the chimera comes out. Arch Dermatol 144:327–330

    Article  PubMed  Google Scholar 

  24. Scherer SW, Lee C, Birney E, Altshuler DM, Eichler EE, Carter NP, Hurles ME, Feuk L (2007) Challenges and standards in integrating surveys of structural variation. Nat Genet 39:S7–S15

    Article  PubMed  CAS  Google Scholar 

  25. Fassihi H, Liu L, Renwick PJ, Braude PR, McGrath JA (2010) Development and successful clinical application of preimplantation genetic haplotyping for Herlitz junctional epidermolysis bullosa. Br J Dermatol 162:1330–1336

    Article  PubMed  CAS  Google Scholar 

  26. Rohlin A, Wernersson J, Engwall Y, Wiklund L, Bjork J, Nordling M (2009) Parallel sequencing used in detection of mosaic mutations: comparison with four diagnostic DNA screening techniques. Hum Mutat 30:1012–1020

    Article  PubMed  CAS  Google Scholar 

  27. Wheeler DA, Srinivasan M, Egholm M, Shen Y, Chen L, McGuire A, He W, Chen YJ, Makhijani V, Roth GT, Gomes X, Tartaro K, Niazi F, Turcotte CL, Irzyk GP, Lupski JR, Chinault C, Song XZ, Liu Y, Yuan Y, Nazareth L, Qin X, Muzny DM, Margulies M, Weinstock GM, Gibbs RA, Rothberg JM (2008) The complete genome of an individual by massively parallel DNA sequencing. Nature 452:872–876

    Article  PubMed  CAS  Google Scholar 

  28. Craig DW, Pearson JV, Szelinger S, Sekar A, Redman M, Corneveaux JJ, Pawlowski TL, Laub T, Nunn G, Stephan DA, Homer N, Huentelman MJ (2008) Identification of genetic variants using bar-coded multiplexed sequencing. Nat Methods 5:887–893

    Article  PubMed  CAS  Google Scholar 

  29. Suzuki S, Ono N, Furusawa C, Ying BW, Yomo T (2011) Comparison of sequence reads obtained from three next-generation sequencing platforms. PLoS One 6:e19534

    Article  PubMed  CAS  Google Scholar 

  30. Cooper DN, Ball EV, Krawczak M (1998) The human gene mutation database. Nucleic Acids Res 26:285–287

    Article  PubMed  CAS  Google Scholar 

  31. Christiano AM, McGrath JA, Tan KC, Uitto J (1996) Glycine substitutions in the triple-helical region of type VII collagen result in a spectrum of dystrophic epidermolysis bullosa phenotypes and patterns of inheritance. Am J Hum Genet 58:671–681

    PubMed  CAS  Google Scholar 

  32. Dowling GB, Meara RH (1954) Epidermolysis bullosa resembling juvenile dermatitis herpetiformis. Br J Dermatol 66:139–143

    Article  PubMed  CAS  Google Scholar 

  33. Bull MJ, Norins AL, Weaver DD, Weber T, Mitchell M (1983) Epidermolysis bullosa–pyloric atresia. An autosomal recessive syndrome. Am J Dis Child 137:449–451

    PubMed  CAS  Google Scholar 

  34. McGrath JA, Eady RA (1997) The role of immunohistochemistry in the diagnosis of the non-lethal forms of junctional epidermolysis bullosa. J Dermatol Sci 14:68–75

    Article  PubMed  CAS  Google Scholar 

  35. Wessagowit V, Kim SC, Woong Oh S, McGrath JA (2005) Genotype-phenotype correlation in recessive dystrophic epidermolysis bullosa: when missense doesn’t make sense. J Invest Dermatol 124:863–866

    Article  PubMed  CAS  Google Scholar 

  36. Christiano AM, Hoffman GG, Zhang X, Xu Y, Tamai Y, Greenspan DS, Uitto J (1997) Strategy for identification of sequence variants in COL7A1 and a novel 2-bp deletion mutation in recessive dystrophic epidermolysis bullosa. Hum Mutat 10:408–414

    Article  PubMed  CAS  Google Scholar 

  37. Tasanen K, Floeth M, Schumann H, Bruckner-Tuderman L (2000) Hemizygosity for a glycine substitution in collagen XVII: unfolding and degradation of the ectodomain. J Invest Dermatol 115:207–212

    Article  PubMed  CAS  Google Scholar 

  38. Floeth M, Bruckner-Tuderman L (1999) Digenic junctional epidermolysis bullosa: mutations in COL17A1 and LAMB3 genes. Am J Hum Genet 65:1530–1537

    Article  PubMed  CAS  Google Scholar 

  39. Wallace MR, Marchuk DA, Andersen LB, Letcher R, Odeh HM, Saulino AM, Fountain JW, Brereton A, Nicholson J, Mitchell AL et al (1990) Type 1 neurofibromatosis gene: identification of a large transcript disrupted in three NF1 patients. Science 249:181–186

    Article  PubMed  CAS  Google Scholar 

  40. Huson SM, Compston DA, Clark P, Harper PS (1989) A genetic study of von Recklinghausen neurofibromatosis in south east Wales. I. Prevalence, fitness, mutation rate, and effect of parental transmission on severity. J Med Genet 26:704–711

    Article  PubMed  CAS  Google Scholar 

  41. Zlotogora J (1993) Mutations in von Recklinghausen neurofibromatosis: an hypothesis. Am J Med Genet 46:182–184

    Article  PubMed  CAS  Google Scholar 

  42. Jadayel D, Fain P, Upadhyaya M, Ponder MA, Huson SM, Carey J, Fryer A, Mathew CG, Barker DF, Ponder BA (1990) Paternal origin of new mutations in von Recklinghausen neurofibromatosis. Nature 343:558–559

    Article  PubMed  CAS  Google Scholar 

  43. Upadhyaya M, Ruggieri M, Maynard J, Osborn M, Hartog C, Mudd S, Penttinen M, Cordeiro I, Ponder M, Ponder BA, Krawczak M, Cooper DN (1998) Gross deletions of the neurofibromatosis type 1 (NF1) gene are predominantly of maternal origin and commonly associated with a learning disability, dysmorphic features and developmental delay. Hum Genet 102:591–597

    Article  PubMed  CAS  Google Scholar 

  44. Ingordo V, D’Andria G, Mendicini S, Grecucci M, Baglivo A (1995) Segmental neurofibromatosis: is it uncommon or underdiagnosed? Arch Dermatol 131:959–960

    Article  PubMed  CAS  Google Scholar 

  45. Lazaro C, Gaona A, Lynch M, Kruyer H, Ravella A, Estivill X (1995) Molecular characterization of the breakpoints of a 12-kb deletion in the NF1 gene in a family showing germ-line mosaicism. Am J Hum Genet 57:1044–1049

    PubMed  CAS  Google Scholar 

  46. Altarescu G, Brooks B, Kaplan Y, Eldar-Geva T, Margalioth EJ, Levy-Lahad E, Renbaum P (2006) Single-sperm analysis for haplotype construction of de-novo paternal mutations: application to PGD for neurofibromatosis type 1. Hum Reprod 21:2047–2051

    Article  PubMed  CAS  Google Scholar 

  47. Pros E, Fernandez-Rodriguez J, Canet B, Benito L, Sanchez A, Benavides A, Ramos FJ, Lopez-Ariztegui MA, Capella G, Blanco I, Serra E, Lazaro C (2009) Antisense therapeutics for neurofibromatosis type 1 caused by deep intronic mutations. Hum Mutat 30:454–462

    Article  PubMed  CAS  Google Scholar 

  48. Wimmer K, Roca X, Beiglbock H, Callens T, Etzler J, Rao AR, Krainer AR, Fonatsch C, Messiaen L (2007) Extensive in silico analysis of NF1 splicing defects uncovers determinants for splicing outcome upon 5′ splice-site disruption. Hum Mutat 28:599–612

    Article  PubMed  CAS  Google Scholar 

  49. Kehrer-Sawatzki H, Cooper DN (2008) Mosaicism in sporadic neurofibromatosis type 1: variations on a theme common to other hereditary cancer syndromes? J Med Genet 45:622–631

    Article  PubMed  CAS  Google Scholar 

  50. Lopez Correa C, Brems H, Lazaro C, Marynen P, Legius E (2000) Unequal meiotic crossover: a frequent cause of NF1 microdeletions. Am J Hum Genet 66:1969–1974

    Article  PubMed  CAS  Google Scholar 

  51. O’Donovan MR, Freemantle MR, Hull G, Bell DA, Arlett CF, Cole J (1995) Extended-term cultures of human T-lymphocytes: a practical alternative to primary human lymphocytes for use in genotoxicity testing. Mutagenesis 10:189–201

    Article  PubMed  Google Scholar 

  52. Messiaen LM, Callens T, Mortier G, Beysen D, Vandenbroucke I, Van Roy N, Speleman F, Paepe AD (2000) Exhaustive mutation analysis of the NF1 gene allows identification of 95% of mutations and reveals a high frequency of unusual splicing defects. Hum Mutat 15:541–555

    Article  PubMed  CAS  Google Scholar 

  53. Wimmer K, Yao S, Claes K, Kehrer-Sawatzki H, Tinschert S, De Raedt T, Legius E, Callens T, Beiglbock H, Maertens O, Messiaen L (2006) Spectrum of single- and multiexon NF1 copy number changes in a cohort of 1,100 unselected NF1 patients. Genes Chromosomes Cancer 45:265–276

    Article  PubMed  CAS  Google Scholar 

  54. Gissen P, Johnson CA, Morgan NV, Stapelbroek JM, Forshew T, Cooper WN, McKiernan PJ, Klomp LW, Morris AA, Wraith JE, McClean P, Lynch SA, Thompson RJ, Lo B, Quarrell OW, Di Rocco M, Trembath RC, Mandel H, Wali S, Karet FE, Knisely AS, Houwen RH, Kelly DA, Maher ER (2004) Mutations in VPS33B, encoding a regulator of SNARE-dependent membrane fusion, cause arthrogryposis-renal dysfunction-cholestasis (ARC) syndrome. Nat Genet 36:400–404

    Article  PubMed  CAS  Google Scholar 

  55. Hu Z, Bonifas JM, Beech J, Bench G, Shigihara T, Ogawa H, Ikeda S, Mauro T, Epstein EH Jr (2000) Mutations in ATP2C1, encoding a calcium pump, cause Hailey-Hailey disease. Nat Genet 24:61–65

    Article  PubMed  CAS  Google Scholar 

  56. Rodriguez-Viciana P, Tetsu O, Tidyman WE, Estep AL, Conger BA, Cruz MS, McCormick F, Rauen KA (2006) Germline mutations in genes within the MAPK pathway cause cardio-facio-cutaneous syndrome. Science 311:1287–1290

    Article  PubMed  CAS  Google Scholar 

  57. Niihori T, Aoki Y, Narumi Y, Neri G, Cave H, Verloes A, Okamoto N, Hennekam RC, Gillessen-Kaesbach G, Wieczorek D, Kavamura MI, Kurosawa K, Ohashi H, Wilson L, Heron D, Bonneau D, Corona G, Kaname T, Naritomi K, Baumann C, Matsumoto N, Kato K, Kure S, Matsubara Y (2006) Germline KRAS and BRAF mutations in cardio-facio-cutaneous syndrome. Nat Genet 38:294–296

    Article  PubMed  CAS  Google Scholar 

  58. Norgett EE, Hatsell SJ, Carvajal-Huerta L, Cabezas JC, Common J, Purkis PE, Whittock N, Leigh IM, Stevens HP, Kelsell DP (2000) Recessive mutation in desmoplakin disrupts desmoplakin-intermediate filament interactions and causes dilated cardiomyopathy, woolly hair and keratoderma. Hum Mol Genet 9:2761–2766

    Article  PubMed  CAS  Google Scholar 

  59. Sprecher E, Ishida-Yamamoto A, Mizrahi-Koren M, Rapaport D, Goldsher D, Indelman M, Topaz O, Chefetz I, Keren H, O’Brien TJ, Bercovich D, Sprecher E, Ishida-Yamamoto A, Mizrahi-Koren M, Rapaport D, Goldsher D, Indelman M, Topaz O, Chefetz I, Keren H, O’Brien TJ, Bercovich D, Shalev S, Geiger D, Bergman R, Horowitz M, Mandel H (2005) A mutation in SNAP29, coding for a SNARE protein involved in intracellular trafficking, causes a novel neurocutaneous syndrome characterized by cerebral dysgenesis, neuropathy, ichthyosis, and palmoplantar keratoderma. Am J Hum Genet 77:242–251

    Article  PubMed  CAS  Google Scholar 

  60. Lefevre C, Jobard F, Caux F, Bouadjar B, Karaduman A, Heilig R, Lakhdar H, Wollenberg A, Verret JL, Weissenbach J, Ozguc M, Lathrop M, Prud’homme JF, Fischer J (2001) Mutations in CGI-58, the gene encoding a new protein of the esterase/lipase/thioesterase subfamily, in Chanarin-Dorfman syndrome. Am J Hum Genet 69:1002–1012

    Article  PubMed  CAS  Google Scholar 

  61. Derry JM, Gormally E, Means GD, Zhao W, Meindl A, Kelley RI, Boyd Y, Herman GE (1999) Mutations in a delta 8-delta 7 sterol isomerase in the tattered mouse and X-linked dominant chondrodysplasia punctata. jderry@immunex.com. Nat Genet 22:286–290

    Article  PubMed  CAS  Google Scholar 

  62. Kranz C, Jungeblut C, Denecke J, Erlekotte A, Sohlbach C, Debus V, Kehl HG, Harms E, Reith A, Reichel S, Grobe H, Hammersen G, Schwarzer U, Marquardt T (2007) A defect in dolichol phosphate biosynthesis causes a new inherited disorder with death in early infancy. Am J Hum Genet 80:433–440

    Article  PubMed  CAS  Google Scholar 

  63. Konig A, Happle R, Bornholdt D, Engel H, Grzeschik KH (2000) Mutations in the NSDHL gene, encoding a 3beta-hydroxysteroid dehydrogenase, cause CHILD syndrome. Am J Med Genet 90:339–346

    Article  PubMed  CAS  Google Scholar 

  64. Sakuntabhai A, Ruiz-Perez V, Carter S, Jacobsen N, Burge S, Monk S, Smith M, Munro CS, O’Donovan M, Craddock N, Kucherlapati R, Rees JL, Owen M, Lathrop GM, Monaco AP, Strachan T, Hovnanian A (1999) Mutations in ATP2A2, encoding a Ca2+ pump, cause Darier disease. Nat Genet 21:271–277

    Article  PubMed  CAS  Google Scholar 

  65. Maestrini E, Korge BP, Ocana-Sierra J, Calzolari E, Cambiaghi S, Scudder PM, Hovnanian A, Monaco AP, Munro CS (1999) A missense mutation in connexin26, D66H, causes mutilating keratoderma with sensorineural deafness (Vohwinkel’s syndrome) in three unrelated families. Hum Mol Genet 8:1237–1243

    Article  PubMed  CAS  Google Scholar 

  66. Lugassy J, Itin P, Ishida-Yamamoto A, Holland K, Huson S, Geiger D, Hennies HC, Indelman M, Bercovich D, Uitto J, Bergman R, McGrath JA, Richard G, Sprecher E (2006) Naegeli-Franceschetti-Jadassohn syndrome and dermatopathia pigmentosa reticularis: two allelic ectodermal dysplasias caused by dominant mutations in KRT14. Am J Hum Genet 79:724–730

    Article  PubMed  CAS  Google Scholar 

  67. Betz RC, Planko L, Eigelshoven S, Hanneken S, Pasternack SM, Bussow H, Van Den Bogaert K, Wenzel J, Braun-Falco M, Rutten A, Rogers MA, Ruzicka T, Nothen MM, Magin TM, Kruse R (2006) Loss-of-function mutations in the keratin 5 gene lead to Dowling-Degos disease. Am J Hum Genet 78:510–519

    Article  PubMed  CAS  Google Scholar 

  68. Rothnagel JA, Dominey AM, Dempsey LD, Longley MA, Greenhalgh DA, Gagne TA, Huber M, Frenk E, Hohl D, Roop DR (1992) Mutations in the rod domains of keratins 1 and 10 in epidermolytic hyperkeratosis. Science 257:1128–1130

    Article  PubMed  CAS  Google Scholar 

  69. Richard G, Smith LE, Bailey RA, Itin P, Hohl D, Epstein EH Jr, DiGiovanna JJ, Compton JG, Bale SJ (1998) Mutations in the human connexin gene GJB3 cause erythrokeratodermia variabilis. Nat Genet 20:366–369

    Article  PubMed  CAS  Google Scholar 

  70. Macari F, Landau M, Cousin P, Mevorah B, Brenner S, Panizzon R, Schorderet DF, Hohl D, Huber M (2000) Mutation in the gene for connexin 30.3 in a family with erythrokeratodermia variabilis. Am J Hum Genet 67:1296–1301

    PubMed  CAS  Google Scholar 

  71. Hart TC, Stabholz A, Meyle J, Shapira L, Van Dyke TE, Cutler CW, Soskolne WA (1997) Genetic studies of syndromes with severe periodontitis and palmoplantar hyperkeratosis. J Periodontal Res 32:81–89

    Article  PubMed  CAS  Google Scholar 

  72. Choate KA, Lu Y, Zhou J, Choi M, Elias PM, Farhi A, Nelson-Williams C, Crumrine D, Williams ML, Nopper AJ, Bree A, Milstone LM, Lifton RP (2010) Mitotic recombination in patients with ichthyosis causes reversion of dominant mutations in KRT10. Science 330:94–97

    Article  PubMed  CAS  Google Scholar 

  73. Sybert VP, Francis JS, Corden LD, Smith LT, Weaver M, Stephens K, McLean WH (1999) Cyclic ichthyosis with epidermolytic hyperkeratosis: a phenotype conferred by mutations in the 2B domain of keratin K1. Am J Hum Genet 64:732–738

    Article  PubMed  CAS  Google Scholar 

  74. Lefevre C, Bouadjar B, Karaduman A, Jobard F, Saker S, Ozguc M, Lathrop M, Prud’homme JF, Fischer J (2004) Mutations in ichthyin a new gene on chromosome 5q33 in a new form of autosomal recessive congenital ichthyosis. Hum Mol Genet 13:2473–2482

    Article  PubMed  CAS  Google Scholar 

  75. Kelsell DP, Norgett EE, Unsworth H, Teh MT, Cullup T, Mein CA, Dopping-Hepenstal PJ, Dale BA, Tadini G, Fleckman P, Stephens KG, Sybert VP, Mallory SB, North BV, Witt DR, Sprecher E, Taylor AE, Ilchyshyn A, Kennedy CT, Goodyear H, Moss C, Paige D, Harper JI, Young BD, Leigh IM, Eady RA, O’Toole EA (2005) Mutations in ABCA12 underlie the severe congenital skin disease harlequin ichthyosis. Am J Hum Genet 76:794–803

    Article  PubMed  CAS  Google Scholar 

  76. Rothnagel JA, Traupe H, Wojcik S, Huber M, Hohl D, Pittelkow MR, Saeki H, Ishibashi Y, Roop DR (1994) Mutations in the rod domain of keratin 2e in patients with ichthyosis bullosa of Siemens. Nat Genet 7:485–490

    Article  PubMed  CAS  Google Scholar 

  77. Smith FJ, Irvine AD, Terron-Kwiatkowski A, Sandilands A, Campbell LE, Zhao Y, Liao H, Evans AT, Goudie DR, Lewis-Jones S, Arseculeratne G, Munro CS, Sergeant A, O’Regan G, Bale SJ, Compton JG, DiGiovanna JJ, Presland RB, Fleckman P, McLean WH (2006) Loss-of-function mutations in the gene encoding filaggrin cause ichthyosis vulgaris. Nat Genet 38:337–342

    Article  PubMed  CAS  Google Scholar 

  78. Klar J, Schweiger M, Zimmerman R, Zechner R, Li H, Torma H, Vahlquist A, Bouadjar B, Dahl N, Fischer J (2009) Mutations in the fatty acid transport protein 4 gene cause the ichthyosis prematurity syndrome. Am J Hum Genet 85:248–253

    Article  PubMed  CAS  Google Scholar 

  79. van Geel M, van Steensel MA, Kuster W, Hennies HC, Happle R, Steijlen PM, Konig A (2002) HID and KID syndromes are associated with the same connexin 26 mutation. Br J Dermatol 146:938–942

    Article  PubMed  Google Scholar 

  80. Huber M, Rettler I, Bernasconi K, Frenk E, Lavrijsen SP, Ponec M, Bon A, Lautenschlager S, Schorderet DF, Hohl D (1995) Mutations of keratinocyte transglutaminase in lamellar ichthyosis. Science 267:525–528

    Article  PubMed  CAS  Google Scholar 

  81. Lefevre C, Audebert S, Jobard F, Bouadjar B, Lakhdar H, Boughdene-Stambouli O, Blanchet-Bardon C, Heilig R, Foglio M, Weissenbach J, Lathrop M, Prud’homme JF, Fischer J (2003) Mutations in the transporter ABCA12 are associated with lamellar ichthyosis type 2. Hum Mol Genet 12:2369–2378

    Article  PubMed  CAS  Google Scholar 

  82. Lefevre C, Bouadjar B, Ferrand V, Tadini G, Megarbane A, Lathrop M, Prud’homme JF, Fischer J (2006) Mutations in a new cytochrome P450 gene in lamellar ichthyosis type 3. Hum Mol Genet 15:767–776

    Article  PubMed  CAS  Google Scholar 

  83. Hadj-Rabia S, Baala L, Vabres P, Hamel-Teillac D, Jacquemin E, Fabre M, Lyonnet S, De Prost Y, Munnich A, Hadchouel M, Smahi A (2004) Claudin-1 gene mutations in neonatal sclerosing cholangitis associated with ichthyosis: a tight junction disease. Gastroenterology 127:1386–1390

    Article  PubMed  CAS  Google Scholar 

  84. Laiho E, Ignatius J, Mikkola H, Yee VC, Teller DC, Niemi KM, Saarialho-Kere U, Kere J, Palotie A (1997) Transglutaminase 1 mutations in autosomal recessive congenital ichthyosis: private and recurrent mutations in an isolated population. Am J Hum Genet 61:529–538

    Article  PubMed  CAS  Google Scholar 

  85. Jobard F, Lefevre C, Karaduman A, Blanchet-Bardon C, Emre S, Weissenbach J, Ozguc M, Lathrop M, Prud’homme JF, Fischer J (2002) Lipoxygenase-3 (ALOXE3) and 12(R)-lipoxygenase (ALOX12B) are mutated in non-bullous congenital ichthyosiform erythroderma (NCIE) linked to chromosome 17p13.1. Hum Mol Genet 11:107–113

    Article  PubMed  CAS  Google Scholar 

  86. Basel-Vanagaite L, Attia R, Ishida-Yamamoto A, Rainshtein L, Ben Amitai D, Lurie R, Pasmanik-Chor M, Indelman M, Zvulunov A, Saban S, Magal N, Sprecher E, Shohat M (2007) Autosomal recessive ichthyosis with hypotrichosis caused by a mutation in ST14, encoding type II transmembrane serine protease matriptase. Am J Hum Genet 80:467–477

    Article  PubMed  CAS  Google Scholar 

  87. van Steensel MA, van Geel M, Nahuys M, Smitt JH, Steijlen PM (2002) A novel connexin 26 mutation in a patient diagnosed with keratitis-ichthyosis-deafness syndrome. J Invest Dermatol 118:724–727

    Article  PubMed  Google Scholar 

  88. Aten E, Brasz LC, Bornholdt D, Hooijkaas IB, Porteous ME, Sybert VP, Vermeer MH, Vossen RH, van der Wielen MJ, Bakker E, Breuning MH, Grzeschik KH, Oosterwijk JC, den Dunnen JT (2010) Keratosis Follicularis Spinulosa Decalvans is caused by mutations in MBTPS2. Hum Mutat 31:1125–1133

    Article  PubMed  CAS  Google Scholar 

  89. Rickman L, Simrak D, Stevens HP, Hunt DM, King IA, Bryant SP, Eady RA, Leigh IM, Arnemann J, Magee AI, Kelsell DP, Buxton RS (1999) N-terminal deletion in a desmosomal cadherin causes the autosomal dominant skin disease striate palmoplantar keratoderma. Hum Mol Genet 8:971–976

    Article  PubMed  CAS  Google Scholar 

  90. Armstrong DK, McKenna KE, Purkis PE, Green KJ, Eady RA, Leigh IM, Hughes AE (1999) Haploinsufficiency of desmoplakin causes a striate subtype of palmoplantar keratoderma. Hum Mol Genet 8:143–148

    Article  PubMed  CAS  Google Scholar 

  91. Whittock NV, Smith FJ, Wan H, Mallipeddi R, Griffiths WA, Dopping-Hepenstal P, Ashton GH, Eady RA, McLean WH, McGrath JA (2002) Frameshift mutation in the V2 domain of human keratin 1 results in striate palmoplantar keratoderma. J Invest Dermatol 118:838–844

    Article  PubMed  CAS  Google Scholar 

  92. Richard G, Brown N, Ishida-Yamamoto A, Krol A (2004) Expanding the phenotypic spectrum of Cx26 disorders: Bart-Pumphrey syndrome is caused by a novel missense mutation in GJB2. J Invest Dermatol 123:856–863

    Article  PubMed  CAS  Google Scholar 

  93. Fischer J, Bouadjar B, Heilig R, Huber M, Lefevre C, Jobard F, Macari F, Bakija-Konsuo A, Ait-Belkacem F, Weissenbach J, Lathrop M, Hohl D, Prud’homme JF (2001) Mutations in the gene encoding SLURP-1 in Mal de Meleda. Hum Mol Genet 10:875–880

    Article  PubMed  CAS  Google Scholar 

  94. Dierks T, Schmidt B, Borissenko LV, Peng J, Preusser A, Mariappan M, von Figura K (2003) Multiple sulfatase deficiency is caused by mutations in the gene encoding the human C(alpha)-formylglycine generating enzyme. Cell 113:435–444

    Article  PubMed  CAS  Google Scholar 

  95. Cosma MP, Pepe S, Annunziata I, Newbold RF, Grompe M, Parenti G, Ballabio A (2003) The multiple sulfatase deficiency gene encodes an essential and limiting factor for the activity of sulfatases. Cell 113:445–456

    Article  PubMed  CAS  Google Scholar 

  96. Tartaglia M, Mehler EL, Goldberg R, Zampino G, Brunner HG, Kremer H, van der Burgt I, Crosby AH, Ion A, Jeffery S, Kalidas K, Patton MA, Kucherlapati RS, Gelb BD (2001) Mutations in PTPN11, encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome. Nat Genet 29:465–468

    Article  PubMed  CAS  Google Scholar 

  97. Schubbert S, Zenker M, Rowe SL, Boll S, Klein C, Bollag G, van der Burgt I, Musante L, Kalscheuer V, Wehner LE, Nguyen H, West B, Zhang KY, Sistermans E, Rauch A, Niemeyer CM, Shannon K, Kratz CP (2006) Germline KRAS mutations cause Noonan syndrome. Nat Genet 38:331–336

    Article  PubMed  CAS  Google Scholar 

  98. Roberts AE, Araki T, Swanson KD, Montgomery KT, Schiripo TA, Joshi VA, Li L, Yassin Y, Tamburino AM, Neel BG, Kucherlapati RS (2007) Germline gain-of-function mutations in SOS1 cause Noonan syndrome. Nat Genet 39:70–74

    Article  PubMed  CAS  Google Scholar 

  99. Tartaglia M, Pennacchio LA, Zhao C, Yadav KK, Fodale V, Sarkozy A, Pandit B, Oishi K, Martinelli S, Schackwitz W, Ustaszewska A, Martin J, Bristow J, Carta C, Lepri F, Neri C, Vasta I, Gibson K, Curry CJ, Siguero JP, Digilio MC, Zampino G, Dallapiccola B, Bar-Sagi D, Gelb BD (2007) Gain-of-function SOS1 mutations cause a distinctive form of Noonan syndrome. Nat Genet 39:75–79

    Article  PubMed  CAS  Google Scholar 

  100. Pandit B, Sarkozy A, Pennacchio LA, Carta C, Oishi K, Martinelli S, Pogna EA, Schackwitz W, Ustaszewska A, Landstrom A, Bos JM, Ommen SR, Esposito G, Lepri F, Faul C, Mundel P, Lopez Siguero JP, Tenconi R, Selicorni A, Rossi C, Mazzanti L, Torrente I, Marino B, Digilio MC, Zampino G, Ackerman MJ, Dallapiccola B, Tartaglia M, Gelb BD (2007) Gain-of-function RAF1 mutations cause Noonan and LEOPARD syndromes with hypertrophic cardiomyopathy. Nat Genet 39:1007–1012

    Article  PubMed  CAS  Google Scholar 

  101. Cirstea IC, Kutsche K, Dvorsky R, Gremer L, Carta C, Horn D, Roberts AE, Lepri F, Merbitz-Zahradnik T, Konig R, Kratz CP, Pantaleoni F, Dentici ML, Joshi VA, Kucherlapati RS, Mazzanti L, Mundlos S, Patton MA, Silengo MC, Rossi C, Zampino G, Digilio C, Stuppia L, Seemanova E, Pennacchio LA, Gelb BD, Dallapiccola B, Wittinghofer A, Ahmadian MR, Tartaglia M, Zenker M (2010) A restricted spectrum of NRAS mutations causes Noonan syndrome. Nat Genet 42:27–29

    Article  PubMed  CAS  Google Scholar 

  102. Sarkozy A, Carta C, Moretti S, Zampino G, Digilio MC, Pantaleoni F, Scioletti AP, Esposito G, Cordeddu V, Lepri F, Petrangeli V, Dentici ML, Mancini GM, Selicorni A, Rossi C, Mazzanti L, Marino B, Ferrero GB, Silengo MC, Memo L, Stanzial F, Faravelli F, Stuppia L, Puxeddu E, Gelb BD, Dallapiccola B, Tartaglia M (2009) Germline BRAF mutations in Noonan, LEOPARD, and cardiofaciocutaneous syndromes: molecular diversity and associated phenotypic spectrum. Hum Mutat 30:695–702

    Article  PubMed  CAS  Google Scholar 

  103. Martinelli S, De Luca A, Stellacci E, Rossi C, Checquolo S, Lepri F, Caputo V, Silvano M, Buscherini F, Consoli F, Ferrara G, Digilio MC, Cavaliere ML, van Hagen JM, Zampino G, van der Burgt I, Ferrero GB, Mazzanti L, Screpanti I, Yntema HG, Nillesen WM, Savarirayan R, Zenker M, Dallapiccola B, Gelb BD, Tartaglia M (2010) Heterozygous germline mutations in the CBL tumor-suppressor gene cause a Noonan syndrome-like phenotype. Am J Hum Genet 87:250–257

    Article  PubMed  CAS  Google Scholar 

  104. Cordeddu V, Di Schiavi E, Pennacchio LA, Ma’ayan A, Sarkozy A, Fodale V, Cecchetti S, Cardinale A, Martin J, Schackwitz W, Lipzen A, Zampino G, Mazzanti L, Digilio MC, Martinelli S, Flex E, Lepri F, Bartholdi D, Kutsche K, Ferrero GB, Anichini C, Selicorni A, Rossi C, Tenconi R, Zenker M, Merlo D, Dallapiccola B, Iyengar R, Bazzicalupo P, Gelb BD, Tartaglia M (2009) Mutation of SHOC2 promotes aberrant protein N-myristoylation and causes Noonan-like syndrome with loose anagen hair. Nat Genet 41:1022–1026

    Article  PubMed  CAS  Google Scholar 

  105. McKoy G, Protonotarios N, Crosby A, Tsatsopoulou A, Anastasakis A, Coonar A, Norman M, Baboonian C, Jeffery S, McKenna WJ (2000) Identification of a deletion in plakoglobin in arrhythmogenic right ventricular cardiomyopathy with palmoplantar keratoderma and woolly hair (Naxos disease). Lancet 355:2119–2124

    Article  PubMed  CAS  Google Scholar 

  106. Chavanas S, Bodemer C, Rochat A, Hamel-Teillac D, Ali M, Irvine AD, Bonafe JL, Wilkinson J, Taieb A, Barrandon Y, Harper JI, de Prost Y, Hovnanian A (2000) Mutations in SPINK5, encoding a serine protease inhibitor, cause Netherton syndrome. Nat Genet 25:141–142

    Article  PubMed  CAS  Google Scholar 

  107. Paznekas WA, Boyadjiev SA, Shapiro RE, Daniels O, Wollnik B, Keegan CE, Innis JW, Dinulos MB, Christian C, Hannibal MC, Jabs EW (2003) Connexin 43 (GJA1) mutations cause the pleiotropic phenotype of oculodentodigital dysplasia. Am J Hum Genet 72:408–418

    Article  PubMed  CAS  Google Scholar 

  108. Bowden PE, Haley JL, Kansky A, Rothnagel JA, Jones DO, Turner RJ (1995) Mutation of a type II keratin gene (K6a) in pachyonychia congenita. Nat Genet 10:363–365

    Article  PubMed  CAS  Google Scholar 

  109. Smith FJ, McKusick VA, Nielsen K, Pfendner E, Uitto J, McLean WH (1999) Cloning of multiple keratin 16 genes facilitates prenatal diagnosis of pachyonychia congenita type 1. Prenat Diagn 19:941–946

    Article  PubMed  CAS  Google Scholar 

  110. Smith FJ, Jonkman MF, van Goor H, Coleman CM, Covello SP, Uitto J, McLean WH (1998) A mutation in human keratin K6b produces a phenocopy of the K17 disorder pachyonychia congenita type 2. Hum Mol Genet 7:1143–1148

    Article  PubMed  CAS  Google Scholar 

  111. Terrinoni A, Smith FJ, Didona B, Canzona F, Paradisi M, Huber M, Hohl D, David A, Verloes A, Leigh IM, Munro CS, Melino G, McLean WH (2001) Novel and recurrent mutations in the genes encoding keratins K6a, K16 and K17 in 13 cases of pachyonychia congenita. J Invest Dermatol 117:1391–1396

    Article  PubMed  CAS  Google Scholar 

  112. Reis A, Hennies HC, Langbein L, Digweed M, Mischke D, Drechsler M, Schrock E, Royer-Pokora B, Franke WW, Sperling K et al (1994) Keratin 9 gene mutations in epidermolytic palmoplantar keratoderma (EPPK). Nat Genet 6:174–179

    Article  PubMed  CAS  Google Scholar 

  113. Hatsell SJ, Eady RA, Wennerstrand L, Dopping-Hepenstal P, Leigh IM, Munro C, Kelsell DP (2001) Novel splice site mutation in keratin 1 underlies mild epidermolytic palmoplantar keratoderma in three kindreds. J Invest Dermatol 116:606–609

    Article  PubMed  CAS  Google Scholar 

  114. Kimonis V, DiGiovanna JJ, Yang JM, Doyle SZ, Bale SJ, Compton JG (1994) A mutation in the V1 end domain of keratin 1 in non-epidermolytic palmar-plantar keratoderma. J Invest Dermatol 103:764–769

    Article  PubMed  CAS  Google Scholar 

  115. Shamsher MK, Navsaria HA, Stevens HP, Ratnavel RC, Purkis PE, Kelsell DP, McLean WH, Cook LJ, Griffiths WA, Gschmeissner S et al (1995) Novel mutations in keratin 16 gene underly focal non-epidermolytic palmoplantar keratoderma (NEPPK) in two families. Hum Mol Genet 4:1875–1881

    Article  PubMed  CAS  Google Scholar 

  116. Parma P, Radi O, Vidal V, Chaboissier MC, Dellambra E, Valentini S, Guerra L, Schedl A, Camerino G (2006) R-spondin1 is essential in sex determination, skin differentiation and malignancy. Nat Genet 38:1304–1309

    Article  PubMed  CAS  Google Scholar 

  117. Toomes C, James J, Wood AJ, Wu CL, McCormick D, Lench N, Hewitt C, Moynihan L, Roberts E, Woods CG, Markham A, Wong M, Widmer R, Ghaffar KA, Pemberton M, Hussein IR, Temtamy SA, Davies R, Read AP, Sloan P, Dixon MJ, Thakker NS (1999) Loss-of-function mutations in the cathepsin C gene result in periodontal disease and palmoplantar keratosis. Nat Genet 23:421–424

    Article  PubMed  CAS  Google Scholar 

  118. Zhang ZH, Niu ZM, Yuan WT, Zhao JJ, Jiang FX, Zhang J, Chai B, Cui F, Chen W, Lian CH, Xiang LH, Xu SJ, Liu WD, Zheng ZZ, Huang W (2005) A mutation in SART3 gene in a Chinese pedigree with disseminated superficial actinic porokeratosis. Br J Dermatol 152:658–663

    Article  PubMed  CAS  Google Scholar 

  119. Oji V, Eckl KM, Aufenvenne K, Natebus M, Tarinski T, Ackermann K, Seller N, Metze D, Nurnberg G, Folster-Holst R, Schafer-Korting M, Hausser I, Traupe H, Hennies HC (2010) Loss of corneodesmosin leads to severe skin barrier defect, pruritus, and atopy: unraveling the peeling skin disease. Am J Hum Genet 87:274–281

    Article  PubMed  CAS  Google Scholar 

  120. Cassidy AJ, van Steensel MA, Steijlen PM, van Geel M, van der Velden J, Morley SM, Terrinoni A, Melino G, Candi E, McLean WH (2005) A homozygous missense mutation in TGM5 abolishes epidermal transglutaminase 5 activity and causes acral peeling skin syndrome. Am J Hum Genet 77:909–917

    Article  PubMed  CAS  Google Scholar 

  121. Mihalik SJ, Morrell JC, Kim D, Sacksteder KA, Watkins PA, Gould SJ (1997) Identification of PAHX, a Refsum disease gene. Nat Genet 17:185–189

    Article  PubMed  CAS  Google Scholar 

  122. Braverman N, Chen L, Lin P, Obie C, Steel G, Douglas P, Chakraborty PK, Clarke JT, Boneh A, Moser A, Moser H, Valle D (2002) Mutation analysis of PEX7 in 60 probands with rhizomelic chondrodysplasia punctata and functional correlations of genotype with phenotype. Hum Mutat 20:284–297

    Article  PubMed  CAS  Google Scholar 

  123. Braverman N, Steel G, Obie C, Moser A, Moser H, Gould SJ, Valle D (1997) Human PEX7 encodes the peroxisomal PTS2 receptor and is responsible for rhizomelic chondrodysplasia punctata. Nat Genet 15:369–376

    Article  PubMed  CAS  Google Scholar 

  124. Motley AM, Hettema EH, Hogenhout EM, Brites P, ten Asbroek AL, Wijburg FA, Baas F, Heijmans HS, Tabak HF, Wanders RJ, Distel B (1997) Rhizomelic chondrodysplasia punctata is a peroxisomal protein targeting disease caused by a non-functional PTS2 receptor. Nat Genet 15:377–380

    Article  PubMed  CAS  Google Scholar 

  125. Purdue PE, Zhang JW, Skoneczny M, Lazarow PB (1997) Rhizomelic chondrodysplasia punctata is caused by deficiency of human PEX7, a homologue of the yeast PTS2 receptor. Nat Genet 15:381–384

    Article  PubMed  CAS  Google Scholar 

  126. Birnbaum RY, Zvulunov A, Hallel-Halevy D, Cagnano E, Finer G, Ofir R, Geiger D, Silberstein E, Feferman Y, Birk OS (2006) Seborrhea-like dermatitis with psoriasiform elements caused by a mutation in ZNF750, encoding a putative C2H2 zinc finger protein. Nat Genet 38:749–751

    Article  PubMed  CAS  Google Scholar 

  127. Smith FJ, Corden LD, Rugg EL, Ratnavel R, Leigh IM, Moss C, Tidman MJ, Hohl D, Huber M, Kunkeler L, Munro CS, Lane EB, McLean WH (1997) Missense mutations in keratin 17 cause either pachyonychia congenita type 2 or a phenotype resembling steatocystoma multiplex. J Invest Dermatol 108:220–223

    Article  PubMed  CAS  Google Scholar 

  128. Natt E, Westphal EM, Toth-Fejel SE, Magenis RE, Buist NR, Rettenmeier R, Scherer G (1987) Inherited and de novo deletion of the tyrosine aminotransferase gene locus at 16q22.1–q22.3 in a patient with tyrosinemia type II. Hum Genet 77:352–358

    Article  PubMed  CAS  Google Scholar 

  129. Maestrini E, Monaco AP, McGrath JA, Ishida-Yamamoto A, Camisa C, Hovnanian A, Weeks DE, Lathrop M, Uitto J, Christiano AM (1996) A molecular defect in loricrin, the major component of the cornified cell envelope, underlies Vohwinkel’s syndrome. Nat Genet 13:70–77

    Article  PubMed  CAS  Google Scholar 

  130. Rugg EL, McLean WH, Allison WE, Lunny DP, Macleod RI, Felix DH, Lane EB, Munro CS (1995) A mutation in the mucosal keratin K4 is associated with oral white sponge nevus. Nat Genet 11:450–452

    Article  PubMed  CAS  Google Scholar 

  131. McGrath JA, McMillan JR, Shemanko CS, Runswick SK, Leigh IM, Lane EB, Garrod DR, Eady RA (1997) Mutations in the plakophilin 1 gene result in ectodermal dysplasia/skin fragility syndrome. Nat Genet 17:240–244

    Article  PubMed  CAS  Google Scholar 

  132. Lane EB, Rugg EL, Navsaria H, Leigh IM, Heagerty AH, Ishida-Yamamoto A, Eady RA (1992) A mutation in the conserved helix termination peptide of keratin 5 in hereditary skin blistering. Nature 356:244–246

    Article  PubMed  CAS  Google Scholar 

  133. Coulombe PA, Hutton ME, Letai A, Hebert A, Paller AS, Fuchs E (1991) Point mutations in human keratin 14 genes of epidermolysis bullosa simplex patients: genetic and functional analyses. Cell 66:1301–1311

    Article  PubMed  CAS  Google Scholar 

  134. Bonifas JM, Rothman AL, Epstein EH Jr (1991) Epidermolysis bullosa simplex: evidence in two families for keratin gene abnormalities. Science 254:1202–1205

    Article  PubMed  CAS  Google Scholar 

  135. Dong W, Ryynanen M, Uitto J (1993) Identification of a leucine-to-proline mutation in the keratin 5 gene in a family with the generalized Kobner type of epidermolysis bullosa simplex. Hum Mutat 2:94–102

    Article  PubMed  CAS  Google Scholar 

  136. Chan YM, Yu QC, Fine JD, Fuchs E (1993) The genetic basis of Weber-Cockayne epidermolysis bullosa simplex. Proc Natl Acad Sci U S A 90:7414–7418

    Article  PubMed  CAS  Google Scholar 

  137. Chen MA, Bonifas JM, Matsumura K, Blumenfeld A, Epstein EH Jr (1993) A novel three-nucleotide deletion in the helix 2B region of keratin 14 in epidermolysis bullosa simplex: delta E375. Hum Mol Genet 2:1971–1972

    Article  PubMed  CAS  Google Scholar 

  138. Uttam J, Hutton E, Coulombe PA, Anton-Lamprecht I, Yu QC, Gedde-Dahl T Jr, Fine JD, Fuchs E (1996) The genetic basis of epidermolysis bullosa simplex with mottled pigmentation. Proc Natl Acad Sci U S A 93:9079–9084

    Article  PubMed  CAS  Google Scholar 

  139. Gu LH, Kim SC, Ichiki Y, Park J, Nagai M, Kitajima Y (2003) A usual frameshift and delayed termination codon mutation in keratin 5 causes a novel type of epidermolysis bullosa simplex with migratory circinate erythema. J Invest Dermatol 121:482–485

    Article  PubMed  CAS  Google Scholar 

  140. Hovnanian A, Pollack E, Hilal L, Rochat A, Prost C, Barrandon Y, Goossens M (1993) A missense mutation in the rod domain of keratin 14 associated with recessive epidermolysis bullosa simplex. Nat Genet 3:327–332

    Article  PubMed  CAS  Google Scholar 

  141. Koss-Harnes D, Hoyheim B, Anton-Lamprecht I, Gjesti A, Jorgensen RS, Jahnsen FL, Olaisen B, Wiche G, Gedde-Dahl T Jr (2002) A site-specific plectin mutation causes dominant epidermolysis bullosa simplex Ogna: two identical de novo mutations. J Invest Dermatol 118:87–93

    Article  PubMed  CAS  Google Scholar 

  142. Smith FJ, Eady RA, Leigh IM, McMillan JR, Rugg EL, Kelsell DP, Bryant SP, Spurr NK, Geddes JF, Kirtschig G, Milana G, de Bono AG, Owaribe K, Wiche G, Pulkkinen L, Uitto J, McLean WH, Lane EB (1996) Plectin deficiency results in muscular dystrophy with epidermolysis bullosa. Nat Genet 13:450–457

    Article  PubMed  CAS  Google Scholar 

  143. Charlesworth A, Gagnoux-Palacios L, Bonduelle M, Ortonne JP, De Raeve L, Meneguzzi G (2003) Identification of a lethal form of epidermolysis bullosa simplex associated with a homozygous genetic mutation in plectin. J Invest Dermatol 121:1344–1348

    Article  PubMed  CAS  Google Scholar 

  144. Vidal F, Baudoin C, Miquel C, Galliano MF, Christiano AM, Uitto J, Ortonne JP, Meneguzzi G (1995) Cloning of the laminin alpha 3 chain gene (LAMA3) and identification of a homozygous deletion in a patient with Herlitz junctional epidermolysis bullosa. Genomics 30:273–280

    Article  PubMed  CAS  Google Scholar 

  145. Pulkkinen L, Christiano AM, Airenne T, Haakana H, Tryggvason K, Uitto J (1994) Mutations in the gamma 2 chain gene (LAMC2) of kalinin/laminin 5 in the junctional forms of epidermolysis bullosa. Nat Genet 6:293–297

    Article  PubMed  CAS  Google Scholar 

  146. Pulkkinen L, Christiano AM, Gerecke D, Wagman DW, Burgeson RE, Pittelkow MR, Uitto J (1994) A homozygous nonsense mutation in the beta 3 chain gene of laminin 5 (LAMB3) in Herlitz junctional epidermolysis bullosa. Genomics 24:357–360

    Article  PubMed  CAS  Google Scholar 

  147. McGrath JA, Gatalica B, Christiano AM, Li K, Owaribe K, McMillan JR, Eady RA, Uitto J (1995) Mutations in the 180-kD bullous pemphigoid antigen (BPAG2), a hemidesmosomal transmembrane collagen (COL17A1), in generalized atrophic benign epidermolysis bullosa. Nat Genet 11:83–86

    Article  PubMed  CAS  Google Scholar 

  148. McGrath JA, Pulkkinen L, Christiano AM, Leigh IM, Eady RA, Uitto J (1995) Altered laminin 5 expression due to mutations in the gene encoding the beta 3 chain (LAMB3) in generalized atrophic benign epidermolysis bullosa. J Invest Dermatol 104:467–474

    Article  PubMed  CAS  Google Scholar 

  149. Nakano A, Chao SC, Pulkkinen L, Murrell D, Bruckner-Tuderman L, Pfendner E, Uitto J (2002) Laminin 5 mutations in junctional epidermolysis bullosa: molecular basis of Herlitz vs. non-Herlitz phenotypes. Hum Genet 110:41–51

    Article  PubMed  CAS  Google Scholar 

  150. Inoue M, Tamai K, Shimizu H, Owaribe K, Nakama T, Hashimoto T, McGrath JA (2000) A homozygous missense mutation in the cytoplasmic tail of beta4 integrin, G931D, that disrupts hemidesmosome assembly and underlies non-Herlitz junctional epidermolysis bullosa without pyloric atresia? J Invest Dermatol 114:1061–1064

    Article  PubMed  CAS  Google Scholar 

  151. Vidal F, Aberdam D, Miquel C, Christiano AM, Pulkkinen L, Uitto J, Ortonne JP, Meneguzzi G (1995) Integrin beta 4 mutations associated with junctional epidermolysis bullosa with pyloric atresia. Nat Genet 10:229–234

    Article  PubMed  CAS  Google Scholar 

  152. Ruzzi L, Gagnoux-Palacios L, Pinola M, Belli S, Meneguzzi G, D’Alessio M, Zambruno G (1997) A homozygous mutation in the integrin alpha6 gene in junctional epidermolysis bullosa with pyloric atresia. J Clin Invest 99:2826–2831

    Article  PubMed  CAS  Google Scholar 

  153. Christiano AM, Ryynanen M, Uitto J (1994) Dominant dystrophic epidermolysis bullosa: identification of a Gly–>Ser substitution in the triple-helical domain of type VII collagen. Proc Natl Acad Sci U S A 91:3549–3553

    Article  PubMed  CAS  Google Scholar 

  154. Christiano AM, Lee JY, Chen WJ, LaForgia S, Uitto J (1995) Pretibial epidermolysis bullosa: genetic linkage to COL7A1 and identification of a glycine-to-cysteine substitution in the triple-helical domain of type VII collagen. Hum Mol Genet 4:1579–1583

    Article  PubMed  CAS  Google Scholar 

  155. Martinez-Mir A, Liu J, Gordon D, Weiner MS, Ahmad W, Fine JD, Ott J, Gilliam TC, Christiano AM (2002) EB simplex superficialis resulting from a mutation in the type VII collagen gene. J Invest Dermatol 118:547–549

    Article  PubMed  CAS  Google Scholar 

  156. Christiano AM, Bart BJ, Epstein EH Jr, Uitto J (1996) Genetic basis of Bart’s syndrome: a glycine substitution mutation in type VII collagen gene. J Invest Dermatol 106:778–780

    Article  PubMed  CAS  Google Scholar 

  157. Christiano AM, Greenspan DS, Hoffman GG, Zhang X, Tamai Y, Lin AN, Dietz HC, Hovnanian A, Uitto J (1993) A missense mutation in type VII collagen in two affected siblings with recessive dystrophic epidermolysis bullosa. Nat Genet 4:62–66

    Article  PubMed  CAS  Google Scholar 

  158. Ayub M, Basit S, Jelani M, Ur Rehman F, Iqbal M, Yasinzai M, Ahmad W (2009) A homozygous nonsense mutation in the human desmocollin-3 (DSC3) gene underlies hereditary hypotrichosis and recurrent skin vesicles. Am J Hum Genet 85:515–520

    Article  PubMed  CAS  Google Scholar 

  159. Jobard F, Bouadjar B, Caux F, Hadj-Rabia S, Has C, Matsuda F, Weissenbach J, Lathrop M, Prud’homme JF, Fischer J (2003) Identification of mutations in a new gene encoding a FERM family protein with a pleckstrin homology domain in Kindler syndrome. Hum Mol Genet 12:925–935

    Article  PubMed  CAS  Google Scholar 

  160. McLean WH, Irvine AD, Hamill KJ, Whittock NV, Coleman-Campbell CM, Mellerio JE, Ashton GS, Dopping-Hepenstal PJ, Eady RA, Jamil T, Phillips RJ, Shabbir SG, Haroon TS, Khurshid K, Moore JE, Page B, Darling J, Atherton DJ, Van Steensel MA, Munro CS, Smith FJ, McGrath JA (2003) An unusual N-terminal deletion of the laminin alpha3a isoform leads to the chronic granulation tissue disorder laryngo-onycho-cutaneous syndrome. Hum Mol Genet 12:2395–2409

    Article  PubMed  CAS  Google Scholar 

  161. Karamatic Crew V, Burton N, Kagan A, Green CA, Levene C, Flinter F, Brady RL, Daniels G, Anstee DJ (2004) CD151, the first member of the tetraspanin (TM4) superfamily detected on erythrocytes, is essential for the correct assembly of human basement membranes in kidney and skin. Blood 104:2217–2223

    Article  PubMed  CAS  Google Scholar 

  162. Whittock NV, Wan H, Morley SM, Garzon MC, Kristal L, Hyde P, McLean WH, Pulkkinen L, Uitto J, Christiano AM, Eady RA, McGrath JA (2002) Compound heterozygosity for non-sense and mis-sense mutations in desmoplakin underlies skin fragility/woolly hair syndrome. J Invest Dermatol 118:232–238

    Article  PubMed  CAS  Google Scholar 

  163. Hammami-Hauasli N, Raghunath M, Kuster W, Bruckner-Tuderman L (1998) Transient bullous dermolysis of the newborn associated with compound heterozygosity for recessive and dominant COL7A1 mutations. J Invest Dermatol 111:1214–1219

    Article  PubMed  CAS  Google Scholar 

  164. Pepys MB, Hawkins PN, Booth DR, Vigushin DM, Tennent GA, Soutar AK, Totty N, Nguyen O, Blake CC, Terry CJ et al (1993) Human lysozyme gene mutations cause hereditary systemic amyloidosis. Nature 362:553–557

    Article  PubMed  CAS  Google Scholar 

  165. Benson MD, Liepnieks J, Uemichi T, Wheeler G, Correa R (1993) Hereditary renal amyloidosis associated with a mutant fibrinogen alpha-chain. Nat Genet 3:252–255

    Article  PubMed  CAS  Google Scholar 

  166. Maury CP, Alli K, Baumann M (1990) Finnish hereditary amyloidosis. Amino acid sequence homology between the amyloid fibril protein and human plasma gelsoline. FEBS Lett 260:85–87

    Article  PubMed  CAS  Google Scholar 

  167. Saraiva MJ, Birken S, Costa PP, Goodman DS (1984) Amyloid fibril protein in familial amyloidotic polyneuropathy, Portuguese type. Definition of molecular abnormality in transthyretin (prealbumin). J Clin Invest 74:104–119

    Article  PubMed  CAS  Google Scholar 

  168. Arita K, South AP, Hans-Filho G, Sakuma TH, Lai-Cheong J, Clements S, Odashiro M, Odashiro DN, Hans-Neto G, Hans NR, Holder MV, Bhogal BS, Hartshorne ST, Akiyama M, Shimizu H, McGrath JA (2008) Oncostatin M receptor-beta mutations underlie familial primary localized cutaneous amyloidosis. Am J Hum Genet 82:73–80

    Article  PubMed  CAS  Google Scholar 

  169. Coucke PJ, Wessels MW, Van Acker P, Gardella R, Barlati S, Willems PJ, Colombi M, De Paepe A (2003) Homozygosity mapping of a gene for arterial tortuosity syndrome to chromosome 20q13. J Med Genet 40:747–751

    Article  PubMed  CAS  Google Scholar 

  170. Hellemans J, Preobrazhenska O, Willaert A, Debeer P, Verdonk PC, Costa T, Janssens K, Menten B, Van Roy N, Vermeulen SJ, Savarirayan R, Van Hul W, Vanhoenacker F, Huylebroeck D, De Paepe A, Naeyaert JM, Vandesompele J, Speleman F, Verschueren K, Coucke PJ, Mortier GR (2004) Loss-of-function mutations in LEMD3 result in osteopoikilosis, Buschke-Ollendorff syndrome and melorheostosis. Nat Genet 36:1213–1218

    Article  PubMed  CAS  Google Scholar 

  171. Aoki Y, Niihori T, Kawame H, Kurosawa K, Ohashi H, Tanaka Y, Filocamo M, Kato K, Suzuki Y, Kure S, Matsubara Y (2005) Germline mutations in HRAS proto-oncogene cause Costello syndrome. Nat Genet 37:1038–1040

    Article  PubMed  CAS  Google Scholar 

  172. Tassabehji M, Metcalfe K, Hurst J, Ashcroft GS, Kielty C, Wilmot C, Donnai D, Read AP, Jones CJ (1998) An elastin gene mutation producing abnormal tropoelastin and abnormal elastic fibres in a patient with autosomal dominant cutis laxa. Hum Mol Genet 7:1021–1028

    Article  PubMed  CAS  Google Scholar 

  173. Zhang MC, He L, Giro M, Yong SL, Tiller GE, Davidson JM (1999) Cutis laxa arising from frameshift mutations in exon 30 of the elastin gene (ELN). J Biol Chem 274:981–986

    Article  PubMed  CAS  Google Scholar 

  174. Loeys B, Van Maldergem L, Mortier G, Coucke P, Gerniers S, Naeyaert JM, De Paepe A (2002) Homozygosity for a missense mutation in fibulin-5 (FBLN5) results in a severe form of cutis laxa. Hum Mol Genet 11:2113–2118

    Article  PubMed  CAS  Google Scholar 

  175. Hucthagowder V, Sausgruber N, Kim KH, Angle B, Marmorstein LY, Urban Z (2006) Fibulin-4: a novel gene for an autosomal recessive cutis laxa syndrome. Am J Hum Genet 78:1075–1080

    Article  PubMed  CAS  Google Scholar 

  176. Kornak U, Reynders E, Dimopoulou A, van Reeuwijk J, Fischer B, Rajab A, Budde B, Nurnberg P, Foulquier F, Lefeber D, Urban Z, Gruenewald S, Annaert W, Brunner HG, van Bokhoven H, Wevers R, Morava E, Matthijs G, Van Maldergem L, Mundlos S (2008) Impaired glycosylation and cutis laxa caused by mutations in the vesicular H+−ATPase subunit ATP6V0A2. Nat Genet 40:32–34

    Article  PubMed  CAS  Google Scholar 

  177. Guernsey DL, Jiang H, Evans SC, Ferguson M, Matsuoka M, Nightingale M, Rideout AL, Provost S, Bedard K, Orr A, Dube MP, Ludman M, Samuels ME (2009) Mutation in pyrroline-5-carboxylate reductase 1 gene in families with cutis laxa type 2. Am J Hum Genet 85:120–129

    Article  PubMed  CAS  Google Scholar 

  178. Michalickova K, Susic M, Willing MC, Wenstrup RJ, Cole WG (1998) Mutations of the alpha2(V) chain of type V collagen impair matrix assembly and produce ehlers-danlos syndrome type I. Hum Mol Genet 7:249–255

    Article  PubMed  CAS  Google Scholar 

  179. Wenstrup RJ, Langland GT, Willing MC, D’Souza VN, Cole WG (1996) A splice-junction mutation in the region of COL5A1 that codes for the carboxyl propeptide of pro alpha 1(V) chains results in the gravis form of the Ehlers-Danlos syndrome (type I). Hum Mol Genet 5:1733–1736

    Article  PubMed  CAS  Google Scholar 

  180. Nuytinck L, Freund M, Lagae L, Pierard GE, Hermanns-Le T, De Paepe A (2000) Classical Ehlers-Danlos syndrome caused by a mutation in type I collagen. Am J Hum Genet 66:1398–1402

    Article  PubMed  CAS  Google Scholar 

  181. De Paepe A, Nuytinck L, Hausser I, Anton-Lamprecht I, Naeyaert JM (1997) Mutations in the COL5A1 gene are causal in the Ehlers-Danlos syndromes I and II. Am J Hum Genet 60:547–554

    PubMed  Google Scholar 

  182. Richards AJ, Martin S, Nicholls AC, Harrison JB, Pope FM, Burrows NP (1998) A single base mutation in COL5A2 causes Ehlers-Danlos syndrome type II. J Med Genet 35:846–848

    Article  PubMed  CAS  Google Scholar 

  183. Narcisi P, Richards AJ, Ferguson SD, Pope FM (1994) A family with Ehlers-Danlos syndrome type III/articular hypermobility syndrome has a glycine 637 to serine substitution in type III collagen. Hum Mol Genet 3:1617–1620

    Article  PubMed  CAS  Google Scholar 

  184. Zweers MC, Bristow J, Steijlen PM, Dean WB, Hamel BC, Otero M, Kucharekova M, Boezeman JB, Schalkwijk J (2003) Haploinsufficiency of TNXB is associated with hypermobility type of Ehlers-Danlos syndrome. Am J Hum Genet 73:214–217

    Article  PubMed  CAS  Google Scholar 

  185. Superti-Furga A, Gugler E, Gitzelmann R, Steinmann B (1988) Ehlers-Danlos syndrome type IV: a multi-exon deletion in one of the two COL3A1 alleles affecting structure, stability, and processing of type III procollagen. J Biol Chem 263:6226–6232

    PubMed  CAS  Google Scholar 

  186. Hautala T, Heikkinen J, Kivirikko KI, Myllyla R (1993) A large duplication in the gene for lysyl hydroxylase accounts for the type VI variant of Ehlers-Danlos syndrome in two siblings. Genomics 15:399–404

    Article  PubMed  CAS  Google Scholar 

  187. Weil D, Bernard M, Combates N, Wirtz MK, Hollister DW, Steinmann B, Ramirez F (1988) Identification of a mutation that causes exon skipping during collagen pre-mRNA splicing in an Ehlers-Danlos syndrome variant. J Biol Chem 263:8561–8564

    PubMed  CAS  Google Scholar 

  188. Weil D, D’Alessio M, Ramirez F, de Wet W, Cole WG, Chan D, Bateman JF (1989) A base substitution in the exon of a collagen gene causes alternative splicing and generates a structurally abnormal polypeptide in a patient with Ehlers-Danlos syndrome type VII. EMBO J 8:1705–1710

    PubMed  CAS  Google Scholar 

  189. Colige A, Nuytinck L, Hausser I, van Essen AJ, Thiry M, Herens C, Ades LC, Malfait F, Paepe AD, Franck P, Wolff G, Oosterwijk JC, Smitt JH, Lapiere CM, Nusgens BV (2004) Novel types of mutation responsible for the dermatosparactic type of Ehlers-Danlos syndrome (Type VIIC) and common polymorphisms in the ADAMTS2 gene. J Invest Dermatol 123:656–663

    Article  PubMed  CAS  Google Scholar 

  190. Schwarze U, Hata R, McKusick VA, Shinkai H, Hoyme HE, Pyeritz RE, Byers PH (2004) Rare autosomal recessive cardiac valvular form of Ehlers-Danlos syndrome results from mutations in the COL1A2 gene that activate the nonsense-mediated RNA decay pathway. Am J Hum Genet 74:917–930

    Article  PubMed  CAS  Google Scholar 

  191. Dundar M, Muller T, Zhang Q, Pan J, Steinmann B, Vodopiutz J, Gruber R, Sonoda T, Krabichler B, Utermann G, Baenziger JU, Zhang L, Janecke AR (2009) Loss of dermatan-4-sulfotransferase 1 function results in adducted thumb-clubfoot syndrome. Am J Hum Genet 85:873–882

    Article  PubMed  CAS  Google Scholar 

  192. Almeida R, Levery SB, Mandel U, Kresse H, Schwientek T, Bennett EP, Clausen H (1999) Cloning and expression of a proteoglycan UDP-galactose:beta-xylose beta1,4-galactosyltransferase I. A seventh member of the human beta4-galactosyltransferase gene family. J Biol Chem 274:26165–26171

    Article  PubMed  CAS  Google Scholar 

  193. Okajima T, Fukumoto S, Furukawa K, Urano T (1999) Molecular basis for the progeroid variant of Ehlers-Danlos syndrome. Identification and characterization of two mutations in galactosyltransferase I gene. J Biol Chem 274:28841–28844

    Article  PubMed  CAS  Google Scholar 

  194. Schalkwijk J, Zweers MC, Steijlen PM, Dean WB, Taylor G, van Vlijmen IM, van Haren B, Miller WL, Bristow J (2001) A recessive form of the Ehlers-Danlos syndrome caused by tenascin-X deficiency. N Engl J Med 345:1167–1175

    Article  PubMed  CAS  Google Scholar 

  195. Voermans NC, Jenniskens GJ, Hamel BC, Schalkwijk J, Guicheney P, van Engelen BG (2007) Ehlers-Danlos syndrome due to tenascin-X deficiency: muscle weakness and contractures support overlap with collagen VI myopathies. Am J Med Genet A 143A:2215–2219

    Article  PubMed  CAS  Google Scholar 

  196. Dowling O, Difeo A, Ramirez MC, Tukel T, Narla G, Bonafe L, Kayserili H, Yuksel-Apak M, Paller AS, Norton K, Teebi AS, Grum-Tokars V, Martin GS, Davis GE, Glucksman MJ, Martignetti JA (2003) Mutations in capillary morphogenesis gene-2 result in the allelic disorders juvenile hyaline fibromatosis and infantile systemic hyalinosis. Am J Hum Genet 73:957–966

    Article  PubMed  CAS  Google Scholar 

  197. Hanks S, Adams S, Douglas J, Arbour L, Atherton DJ, Balci S, Bode H, Campbell ME, Feingold M, Keser G, Kleijer W, Mancini G, McGrath JA, Muntoni F, Nanda A, Teare MD, Warman M, Pope FM, Superti-Furga A, Futreal PA, Rahman N (2003) Mutations in the gene encoding capillary morphogenesis protein 2 cause juvenile hyaline fibromatosis and infantile systemic hyalinosis. Am J Hum Genet 73:791–800

    Article  PubMed  CAS  Google Scholar 

  198. Wang X, Reid Sutton V, Omar Peraza-Llanes J, Yu Z, Rosetta R, Kou YC, Eble TN, Patel A, Thaller C, Fang P, Van den Veyver IB (2007) Mutations in X-linked PORCN, a putative regulator of Wnt signaling, cause focal dermal hypoplasia. Nat Genet 39:836–838

    Article  PubMed  CAS  Google Scholar 

  199. Sheen VL, Jansen A, Chen MH, Parrini E, Morgan T, Ravenscroft R, Ganesh V, Underwood T, Wiley J, Leventer R, Vaid RR, Ruiz DE, Hutchins GM, Menasha J, Willner J, Geng Y, Gripp KW, Nicholson L, Berry-Kravis E, Bodell A, Apse K, Hill RS, Dubeau F, Andermann F, Barkovich J, Andermann E, Shugart YY, Thomas P, Viri M, Veggiotti P, Robertson S, Guerrini R, Walsh CA (2005) Filamin A mutations cause periventricular heterotopia with Ehlers-Danlos syndrome. Neurology 64:254–262

    Article  PubMed  CAS  Google Scholar 

  200. Hamada T, McLean WH, Ramsay M, Ashton GH, Nanda A, Jenkins T, Edelstein I, South AP, Bleck O, Wessagowit V, Mallipeddi R, Orchard GE, Wan H, Dopping-Hepenstal PJ, Mellerio JE, Whittock NV, Munro CS, van Steensel MA, Steijlen PM, Ni J, Zhang L, Hashimoto T, Eady RA, McGrath JA (2002) Lipoid proteinosis maps to 1q21 and is caused by mutations in the extracellular matrix protein 1 gene (ECM1). Hum Mol Genet 11:833–840

    Article  PubMed  CAS  Google Scholar 

  201. Loeys BL, Chen J, Neptune ER, Judge DP, Podowski M, Holm T, Meyers J, Leitch CC, Katsanis N, Sharifi N, Xu FL, Myers LA, Spevak PJ, Cameron DE, De Backer J, Hellemans J, Chen Y, Davis EC, Webb CL, Kress W, Coucke P, Rifkin DB, De Paepe AM, Dietz HC (2005) A syndrome of altered cardiovascular, craniofacial, neurocognitive and skeletal development caused by mutations in TGFBR1 or TGFBR2. Nat Genet 37:275–281

    Article  PubMed  CAS  Google Scholar 

  202. Loeys BL, Schwarze U, Holm T, Callewaert BL, Thomas GH, Pannu H, De Backer JF, Oswald GL, Symoens S, Manouvrier S, Roberts AE, Faravelli F, Greco MA, Pyeritz RE, Milewicz DM, Coucke PJ, Cameron DE, Braverman AC, Byers PH, De Paepe AM, Dietz HC (2006) Aneurysm syndromes caused by mutations in the TGF-beta receptor. N Engl J Med 355:788–798

    Article  PubMed  CAS  Google Scholar 

  203. Mizuguchi T, Collod-Beroud G, Akiyama T, Abifadel M, Harada N, Morisaki T, Allard D, Varret M, Claustres M, Morisaki H, Ihara M, Kinoshita A, Yoshiura K, Junien C, Kajii T, Jondeau G, Ohta T, Kishino T, Furukawa Y, Nakamura Y, Niikawa N, Boileau C, Matsumoto N (2004) Heterozygous TGFBR2 mutations in Marfan syndrome. Nat Genet 36:855–860

    Article  PubMed  CAS  Google Scholar 

  204. Dietz HC, Cutting GR, Pyeritz RE, Maslen CL, Sakai LY, Corson GM, Puffenberger EG, Hamosh A, Nanthakumar EJ, Curristin SM et al (1991) Marfan syndrome caused by a recurrent de novo missense mutation in the fibrillin gene. Nature 352:337–339

    Article  PubMed  CAS  Google Scholar 

  205. Wimplinger I, Morleo M, Rosenberger G, Iaconis D, Orth U, Meinecke P, Lerer I, Ballabio A, Gal A, Franco B, Kutsche K (2006) Mutations of the mitochondrial holocytochrome c-type synthase in X-linked dominant microphthalmia with linear skin defects syndrome. Am J Hum Genet 79:878–889

    Article  PubMed  CAS  Google Scholar 

  206. Hoffmann K, Muller JS, Stricker S, Megarbane A, Rajab A, Lindner TH, Cohen M, Chouery E, Adaimy L, Ghanem I, Delague V, Boltshauser E, Talim B, Horvath R, Robinson PN, Lochmuller H, Hubner C, Mundlos S (2006) Escobar syndrome is a prenatal myasthenia caused by disruption of the acetylcholine receptor fetal gamma subunit. Am J Hum Genet 79:303–312

    Article  PubMed  CAS  Google Scholar 

  207. Michalk A, Stricker S, Becker J, Rupps R, Pantzar T, Miertus J, Botta G, Naretto VG, Janetzki C, Yaqoob N, Ott CE, Seelow D, Wieczorek D, Fiebig B, Wirth B, Hoopmann M, Walther M, Korber F, Blankenburg M, Mundlos S, Heller R, Hoffmann K (2008) Acetylcholine receptor pathway mutations explain various fetal akinesia deformation sequence disorders. Am J Hum Genet 82:464–476

    Article  PubMed  CAS  Google Scholar 

  208. Morgan NV, Brueton LA, Cox P, Greally MT, Tolmie J, Pasha S, Aligianis IA, van Bokhoven H, Marton T, Al-Gazali L, Morton JE, Oley C, Johnson CA, Trembath RC, Brunner HG, Maher ER (2006) Mutations in the embryonal subunit of the acetylcholine receptor (CHRNG) cause lethal and Escobar variants of multiple pterygium syndrome. Am J Hum Genet 79:390–395

    Article  PubMed  CAS  Google Scholar 

  209. Bonneau D, Huret JL, Godeau G, Couet D, Putterman M, Tanzer J, Babin P, Larregue M (1991) Recurrent ctb(7)(q31.3) and possible laminin involvement in a neonatal cutis laxa with a Marfan phenotype. Hum Genet 87:317–319

    Article  PubMed  CAS  Google Scholar 

  210. Giunta C, Randolph A, Al-Gazali LI, Brunner HG, Kraenzlin ME, Steinmann B (2005) Nevo syndrome is allelic to the kyphoscoliotic type of the Ehlers-Danlos syndrome (EDS VIA). Am J Med Genet A 133A:158–164

    Article  PubMed  Google Scholar 

  211. Kaler SG, Gallo LK, Proud VK, Percy AK, Mark Y, Segal NA, Goldstein DS, Holmes CS, Gahl WA (1994) Occipital horn syndrome and a mild Menkes phenotype associated with splice site mutations at the MNK locus. Nat Genet 8:195–202

    Article  PubMed  CAS  Google Scholar 

  212. Kondo S, Schutte BC, Richardson RJ, Bjork BC, Knight AS, Watanabe Y, Howard E, de Lima RL, Daack-Hirsch S, Sander A, McDonald-McGinn DM, Zackai EH, Lammer EJ, Aylsworth AS, Ardinger HH, Lidral AC, Pober BR, Moreno L, Arcos-Burgos M, Valencia C, Houdayer C, Bahuau M, Moretti-Ferreira D, Richieri-Costa A, Dixon MJ, Murray JC (2002) Mutations in IRF6 cause Van der Woude and popliteal pterygium syndromes. Nat Genet 32:285–289

    Article  PubMed  CAS  Google Scholar 

  213. Bergen AA, Plomp AS, Schuurman EJ, Terry S, Breuning M, Dauwerse H, Swart J, Kool M, van Soest S, Baas F, ten Brink JB, de Jong PT (2000) Mutations in ABCC6 cause pseudoxanthoma elasticum. Nat Genet 25:228–231

    Article  PubMed  CAS  Google Scholar 

  214. Le Saux O, Urban Z, Tschuch C, Csiszar K, Bacchelli B, Quaglino D, Pasquali-Ronchetti I, Pope FM, Richards A, Terry S, Bercovitch L, de Paepe A, Boyd CD (2000) Mutations in a gene encoding an ABC transporter cause pseudoxanthoma elasticum. Nat Genet 25:223–227

    Article  PubMed  CAS  Google Scholar 

  215. Ringpfeil F, Lebwohl MG, Christiano AM, Uitto J (2000) Pseudoxanthoma elasticum: mutations in the MRP6 gene encoding a transmembrane ATP-binding cassette (ABC) transporter. Proc Natl Acad Sci U S A 97:6001–6006

    Article  PubMed  CAS  Google Scholar 

  216. Vanakker OM, Martin L, Gheduzzi D, Leroy BP, Loeys BL, Guerci VI, Matthys D, Terry SF, Coucke PJ, Pasquali-Ronchetti I, De Paepe A (2007) Pseudoxanthoma elasticum-like phenotype with cutis laxa and multiple coagulation factor deficiency represents a separate genetic entity. J Invest Dermatol 127:581–587

    Article  PubMed  CAS  Google Scholar 

  217. Navarro CL, De Sandre-Giovannoli A, Bernard R, Boccaccio I, Boyer A, Genevieve D, Hadj-Rabia S, Gaudy-Marqueste C, Smitt HS, Vabres P, Faivre L, Verloes A, Van Essen T, Flori E, Hennekam R, Beemer FA, Laurent N, Le Merrer M, Cau P, Levy N (2004) Lamin A and ZMPSTE24 (FACE-1) defects cause nuclear disorganization and identify restrictive dermopathy as a lethal neonatal laminopathy. Hum Mol Genet 13:2493–2503

    Article  PubMed  CAS  Google Scholar 

  218. Sood S, Eldadah ZA, Krause WL, McIntosh I, Dietz HC (1996) Mutation in fibrillin-1 and the Marfanoid-craniosynostosis (Shprintzen-Goldberg) syndrome. Nat Genet 12:209–211

    Article  PubMed  CAS  Google Scholar 

  219. Giunta C, Elcioglu NH, Albrecht B, Eich G, Chambaz C, Janecke AR, Yeowell H, Weis M, Eyre DR, Kraenzlin M, Steinmann B (2008) Spondylocheiro dysplastic form of the Ehlers-Danlos syndrome—an autosomal-recessive entity caused by mutations in the zinc transporter gene SLC39A13. Am J Hum Genet 82:1290–1305

    Article  PubMed  CAS  Google Scholar 

  220. Loeys BL, Gerber EE, Riegert-Johnson D, Iqbal S, Whiteman P, McConnell V, Chillakuri CR, Macaya D, Coucke PJ, De Paepe A, Judge DP, Wigley F, Davis EC, Mardon HJ, Handford P, Keene DR, Sakai LY, Dietz HC (2010) Mutations in fibrillin-1 cause congenital scleroderma: stiff skin syndrome. Sci Transl Med 2:23ra20

    Article  PubMed  CAS  Google Scholar 

  221. Martignetti JA, Aqeel AA, Sewairi WA, Boumah CE, Kambouris M, Mayouf SA, Sheth KV, Eid WA, Dowling O, Harris J, Glucksman MJ, Bahabri S, Meyer BF, Desnick RJ (2001) Mutation of the matrix metalloproteinase 2 gene (MMP2) causes a multicentric osteolysis and arthritis syndrome. Nat Genet 28:261–265

    Article  PubMed  CAS  Google Scholar 

  222. Camacho Vanegas O, Bertini E, Zhang RZ, Petrini S, Minosse C, Sabatelli P, Giusti B, Chu ML, Pepe G (2001) Ullrich scleroatonic muscular dystrophy is caused by recessive mutations in collagen type VI. Proc Natl Acad Sci U S A 98:7516–7521

    Article  PubMed  CAS  Google Scholar 

  223. Faivre L, Gorlin RJ, Wirtz MK, Godfrey M, Dagoneau N, Samples JR, Le Merrer M, Collod-Beroud G, Boileau C, Munnich A, Cormier-Daire V (2003) In frame fibrillin-1 gene deletion in autosomal dominant Weill-Marchesani syndrome. J Med Genet 40:34–36

    Article  PubMed  CAS  Google Scholar 

  224. Dagoneau N, Benoist-Lasselin C, Huber C, Faivre L, Megarbane A, Alswaid A, Dollfus H, Alembik Y, Munnich A, Legeai-Mallet L, Cormier-Daire V (2004) ADAMTS10 mutations in autosomal recessive Weill-Marchesani syndrome. Am J Hum Genet 75:801–806

    Article  PubMed  CAS  Google Scholar 

  225. Morales J, Al-Sharif L, Khalil DS, Shinwari JM, Bavi P, Al-Mahrouqi RA, Al-Rajhi A, Alkuraya FS, Meyer BF, Al Tassan N (2009) Homozygous mutations in ADAMTS10 and ADAMTS17 cause lenticular myopia, ectopia lentis, glaucoma, spherophakia, and short stature. Am J Hum Genet 85:558–568

    Article  PubMed  CAS  Google Scholar 

  226. Amiel J, Bougeard G, Francannet C, Raclin V, Munnich A, Lyonnet S, Frebourg T (2001) TP63 gene mutation in ADULT syndrome. Eur J Hum Genet 9:642–645

    Article  PubMed  CAS  Google Scholar 

  227. McGrath JA, Duijf PH, Doetsch V, Irvine AD, de Waal R, Vanmolkot KR, Wessagowit V, Kelly A, Atherton DJ, Griffiths WA, Orlow SJ, van Haeringen A, Ausems MG, Yang A, McKeon F, Bamshad MA, Brunner HG, Hamel BC, van Bokhoven H (2001) Hay-Wells syndrome is caused by heterozygous missense mutations in the SAM domain of p63. Hum Mol Genet 10:221–229

    Article  PubMed  CAS  Google Scholar 

  228. Suzuki K, Hu D, Bustos T, Zlotogora J, Richieri-Costa A, Helms JA, Spritz RA (2000) Mutations of PVRL1, encoding a cell-cell adhesion molecule/herpesvirus receptor, in cleft lip/palate-ectodermal dysplasia. Nat Genet 25:427–430

    Article  PubMed  CAS  Google Scholar 

  229. Kjaer KW, Hansen L, Schwabe GC, Marques-de-Faria AP, Eiberg H, Mundlos S, Tommerup N, Rosenberg T (2005) Distinct CDH3 mutations cause ectodermal dysplasia, ectrodactyly, macular dystrophy (EEM syndrome). J Med Genet 42:292–298

    Article  PubMed  CAS  Google Scholar 

  230. Lamartine J, Munhoz Essenfelder G, Kibar Z, Lanneluc I, Callouet E, Laoudj D, Lemaitre G, Hand C, Hayflick SJ, Zonana J, Antonarakis S, Radhakrishna U, Kelsell DP, Christianson AL, Pitaval A, Der Kaloustian V, Fraser C, Blanchet-Bardon C, Rouleau GA, Waksman G (2000) Mutations in GJB6 cause hidrotic ectodermal dysplasia. Nat Genet 26:142–144

    Article  PubMed  CAS  Google Scholar 

  231. Monreal AW, Ferguson BM, Headon DJ, Street SL, Overbeek PA, Zonana J (1999) Mutations in the human homologue of mouse dl cause autosomal recessive and dominant hypohidrotic ectodermal dysplasia. Nat Genet 22:366–369

    Article  PubMed  CAS  Google Scholar 

  232. Bal E, Baala L, Cluzeau C, El Kerch F, Ouldim K, Hadj-Rabia S, Bodemer C, Munnich A, Courtois G, Sefiani A, Smahi A (2007) Autosomal dominant anhidrotic ectodermal dysplasias at the EDARADD locus. Hum Mutat 28:703–709

    Article  PubMed  CAS  Google Scholar 

  233. Headon DJ, Emmal SA, Ferguson BM, Tucker AS, Justice MJ, Sharpe PT, Zonana J, Overbeek PA (2001) Gene defect in ectodermal dysplasia implicates a death domain adapter in development. Nature 414:913–916

    Article  PubMed  CAS  Google Scholar 

  234. Kere J, Srivastava AK, Montonen O, Zonana J, Thomas N, Ferguson B, Munoz F, Morgan D, Clarke A, Baybayan P, Chen EY, Ezer S, Saarialho-Kere U, de la Chapelle A, Schlessinger D (1996) X-linked anhidrotic (hypohidrotic) ectodermal dysplasia is caused by mutation in a novel transmembrane protein. Nat Genet 13:409–416

    Article  PubMed  CAS  Google Scholar 

  235. Zonana J, Elder ME, Schneider LC, Orlow SJ, Moss C, Golabi M, Shapira SK, Farndon PA, Wara DW, Emmal SA, Ferguson BM (2000) A novel X-linked disorder of immune deficiency and hypohidrotic ectodermal dysplasia is allelic to incontinentia pigmenti and due to mutations in IKK-gamma (NEMO). Am J Hum Genet 67:1555–1562

    Article  PubMed  CAS  Google Scholar 

  236. Courtois G, Smahi A, Reichenbach J, Doffinger R, Cancrini C, Bonnet M, Puel A, Chable-Bessia C, Yamaoka S, Feinberg J, Dupuis-Girod S, Bodemer C, Livadiotti S, Novelli F, Rossi P, Fischer A, Israel A, Munnich A, Le Deist F, Casanova JL (2003) A hypermorphic IkappaBalpha mutation is associated with autosomal dominant anhidrotic ectodermal dysplasia and T cell immunodeficiency. J Clin Invest 112:1108–1115

    PubMed  CAS  Google Scholar 

  237. Naeem M, Wajid M, Lee K, Leal SM, Ahmad W (2006) A mutation in the hair matrix and cuticle keratin KRTHB5 gene causes ectodermal dysplasia of hair and nail type. J Med Genet 43:274–279

    Article  PubMed  CAS  Google Scholar 

  238. Celli J, Duijf P, Hamel BC, Bamshad M, Kramer B, Smits AP, Newbury-Ecob R, Hennekam RC, Van Buggenhout G, van Haeringen A, Woods CG, van Essen AJ, de Waal R, Vriend G, Haber DA, Yang A, McKeon F, Brunner HG, van Bokhoven H (1999) Heterozygous germline mutations in the p53 homolog p63 are the cause of EEC syndrome. Cell 99:143–153

    Article  PubMed  CAS  Google Scholar 

  239. Ruiz-Perez VL, Ide SE, Strom TM, Lorenz B, Wilson D, Woods K, King L, Francomano C, Freisinger P, Spranger S, Marino B, Dallapiccola B, Wright M, Meitinger T, Polymeropoulos MH, Goodship J (2000) Mutations in a new gene in Ellis-van Creveld syndrome and Weyers acrodental dysostosis. Nat Genet 24:283–286

    Article  PubMed  CAS  Google Scholar 

  240. Ng SB, Bigham AW, Buckingham KJ, Hannibal MC, McMillin MJ, Gildersleeve HI, Beck AE, Tabor HK, Cooper GM, Mefford HC, Lee C, Turner EH, Smith JD, Rieder MJ, Yoshiura K, Matsumoto N, Ohta T, Niikawa N, Nickerson DA, Bamshad MJ, Shendure J (2010) Exome sequencing identifies MLL2 mutations as a cause of Kabuki syndrome. Nat Genet 42:790–793

    Article  PubMed  CAS  Google Scholar 

  241. van Bokhoven H, Hamel BC, Bamshad M, Sangiorgi E, Gurrieri F, Duijf PH, Vanmolkot KR, van Beusekom E, van Beersum SE, Celli J, Merkx GF, Tenconi R, Fryns JP, Verloes A, Newbury-Ecob RA, Raas-Rotschild A, Majewski F, Beemer FA, Janecke A, Chitayat D, Crisponi G, Kayserili H, Yates JR, Neri G, Brunner HG (2001) p63 Gene mutations in eec syndrome, limb-mammary syndrome, and isolated split hand-split foot malformation suggest a genotype-phenotype correlation. Am J Hum Genet 69:481–492

    Article  PubMed  Google Scholar 

  242. Fang J, Dagenais SL, Erickson RP, Arlt MF, Glynn MW, Gorski JL, Seaver LH, Glover TW (2000) Mutations in FOXC2 (MFH-1), a forkhead family transcription factor, are responsible for the hereditary lymphedema-distichiasis syndrome. Am J Hum Genet 67:1382–1388

    Article  PubMed  CAS  Google Scholar 

  243. Adaimy L, Chouery E, Megarbane H, Mroueh S, Delague V, Nicolas E, Belguith H, de Mazancourt P, Megarbane A (2007) Mutation in WNT10A is associated with an autosomal recessive ectodermal dysplasia: the odonto-onycho-dermal dysplasia. Am J Hum Genet 81:821–828

    Article  PubMed  CAS  Google Scholar 

  244. Ferrante MI, Giorgio G, Feather SA, Bulfone A, Wright V, Ghiani M, Selicorni A, Gammaro L, Scolari F, Woolf AS, Sylvie O, Bernard L, Malcolm S, Winter R, Ballabio A, Franco B (2001) Identification of the gene for oral-facial-digital type I syndrome. Am J Hum Genet 68:569–576

    Article  PubMed  CAS  Google Scholar 

  245. Kantaputra PN, Hamada T, Kumchai T, McGrath JA (2003) Heterozygous mutation in the SAM domain of p63 underlies Rapp-Hodgkin ectodermal dysplasia. J Dent Res 82:433–437

    Article  PubMed  CAS  Google Scholar 

  246. Ianakiev P, Kilpatrick MW, Toudjarska I, Basel D, Beighton P, Tsipouras P (2000) Split-hand/split-foot malformation is caused by mutations in the p63 gene on 3q27. Am J Hum Genet 67:59–66

    Article  PubMed  CAS  Google Scholar 

  247. Gul D, Oktenli C (2002) Evidence for autosomal recessive inheritance of split hand/split foot malformation: a report of nine cases. Clin Dysmorphol 11:183–186

    Article  PubMed  Google Scholar 

  248. Price JA, Bowden DW, Wright JT, Pettenati MJ, Hart TC (1998) Identification of a mutation in DLX3 associated with tricho-dento-osseous (TDO) syndrome. Hum Mol Genet 7:563–569

    Article  PubMed  CAS  Google Scholar 

  249. Momeni P, Glockner G, Schmidt O, von Holtum D, Albrecht B, Gillessen-Kaesbach G, Hennekam R, Meinecke P, Zabel B, Rosenthal A, Horsthemke B, Ludecke HJ (2000) Mutations in a new gene, encoding a zinc-finger protein, cause tricho-rhino-phalangeal syndrome type I. Nat Genet 24:71–74

    Article  PubMed  CAS  Google Scholar 

  250. Hou J, Parrish J, Ludecke HJ, Sapru M, Wang Y, Chen W, Hill A, Siegel-Bartelt J, Northrup H, Elder FF et al (1995) A 4-megabase YAC contig that spans the Langer-Giedion syndrome region on human chromosome 8q24.1: use in refining the location of the trichorhinophalangeal syndrome and multiple exostoses genes (TRPS1 and EXT1). Genomics 29:87–97

    Article  PubMed  CAS  Google Scholar 

  251. Ludecke HJ, Wagner MJ, Nardmann J, La Pillo B, Parrish JE, Willems PJ, Haan EA, Frydman M, Hamers GJ, Wells DE et al (1995) Molecular dissection of a contiguous gene syndrome: localization of the genes involved in the Langer-Giedion syndrome. Hum Mol Genet 4:31–36

    Article  PubMed  CAS  Google Scholar 

  252. Ludecke HJ, Schaper J, Meinecke P, Momeni P, Gross S, von Holtum D, Hirche H, Abramowicz MJ, Albrecht B, Apacik C, Christen HJ, Claussen U, Devriendt K, Fastnacht E, Forderer A, Friedrich U, Goodship TH, Greiwe M, Hamm H, Hennekam RC, Hinkel GK, Hoeltzenbein M, Kayserili H, Majewski F, Mathieu M, McLeod R, Midro AT, Moog U, Nagai T, Niikawa N, Orstavik KH, Plochl E, Seitz C, Schmidtke J, Tranebjaerg L, Tsukahara M, Wittwer B, Zabel B, Gillessen-Kaesbach G, Horsthemke B (2001) Genotypic and phenotypic spectrum in tricho-rhino-phalangeal syndrome types I and III. Am J Hum Genet 68:81–91

    Article  PubMed  CAS  Google Scholar 

  253. Jumlongras D, Bei M, Stimson JM, Wang WF, DePalma SR, Seidman CE, Felbor U, Maas R, Seidman JG, Olsen BR (2001) A nonsense mutation in MSX1 causes Witkop syndrome. Am J Hum Genet 69:67–74

    Article  PubMed  CAS  Google Scholar 

  254. Walker DC, McCloskey DA, Simard LR, McInnes RR (1990) Molecular analysis of human argininosuccinate lyase: mutant characterization and alternative splicing of the coding region. Proc Natl Acad Sci U S A 87:9625–9629

    Article  PubMed  CAS  Google Scholar 

  255. Ahmad W, Faiyaz ul Haque M, Tsou HC, ul Haque S, Lam H, Aita VM, Owen J, deBlaquiere M, Frank J, Cserhalmi-Friedman PB, Leask A, McGrath JA, Peacocke M, Ahmad M, Ott J, Christiano AM (1998) Alopecia universalis associated with a mutation in the human hairless gene. Science 279:720–724

    Article  PubMed  CAS  Google Scholar 

  256. Blaydon DC, Ishii Y, O’Toole EA, Unsworth HC, Teh MT, Ruschendorf F, Sinclair C, Hopsu-Havu VK, Tidman N, Moss C, Watson R, de Berker D, Wajid M, Christiano AM, Kelsell DP (2006) The gene encoding R-spondin 4 (RSPO4), a secreted protein implicated in Wnt signaling, is mutated in inherited anonychia. Nat Genet 38:1245–1247

    Article  PubMed  CAS  Google Scholar 

  257. Sprecher E, Bergman R, Szargel R, Friedman-Birnbaum R, Cohen N (1999) Identification of a genetic defect in the hairless gene in atrichia with papular lesions: evidence for phenotypic heterogeneity among inherited atrichias. Am J Hum Genet 64:1323–1329

    Article  PubMed  CAS  Google Scholar 

  258. Hinson JT, Fantin VR, Schonberger J, Breivik N, Siem G, McDonough B, Sharma P, Keogh I, Godinho R, Santos F, Esparza A, Nicolau Y, Selvaag E, Cohen BH, Hoppel CL, Tranebjaerg L, Eavey RD, Seidman JG, Seidman CE (2007) Missense mutations in the BCS1L gene as a cause of the Bjornstad syndrome. N Engl J Med 356:809–819

    Article  PubMed  CAS  Google Scholar 

  259. Ridanpaa M, van Eenennaam H, Pelin K, Chadwick R, Johnson C, Yuan B, vanVenrooij W, Pruijn G, Salmela R, Rockas S, Makitie O, Kaitila I, de la Chapelle A (2001) Mutations in the RNA component of RNase MRP cause a pleiotropic human disease, cartilage-hair hypoplasia. Cell 104:195–203

    Article  PubMed  CAS  Google Scholar 

  260. Bomont P, Koenig M (2003) Intermediate filament aggregation in fibroblasts of giant axonal neuropathy patients is aggravated in non dividing cells and by microtubule destabilization. Hum Mol Genet 12:813–822

    Article  PubMed  CAS  Google Scholar 

  261. Sprecher E, Bergman R, Richard G, Lurie R, Shalev S, Petronius D, Shalata A, Anbinder Y, Leibu R, Perlman I, Cohen N, Szargel R (2001) Hypotrichosis with juvenile macular dystrophy is caused by a mutation in CDH3, encoding P-cadherin. Nat Genet 29:134–136

    Article  PubMed  CAS  Google Scholar 

  262. Kljuic A, Bazzi H, Sundberg JP, Martinez-Mir A, O’Shaughnessy R, Mahoney MG, Levy M, Montagutelli X, Ahmad W, Aita VM, Gordon D, Uitto J, Whiting D, Ott J, Fischer S, Gilliam TC, Jahoda CA, Morris RJ, Panteleyev AA, Nguyen VT, Christiano AM (2003) Desmoglein 4 in hair follicle differentiation and epidermal adhesion: evidence from inherited hypotrichosis and acquired pemphigus vulgaris. Cell 113:249–260

    Article  PubMed  CAS  Google Scholar 

  263. Kazantseva A, Goltsov A, Zinchenko R, Grigorenko AP, Abrukova AV, Moliaka YK, Kirillov AG, Guo Z, Lyle S, Ginter EK, Rogaev EI (2006) Human hair growth deficiency is linked to a genetic defect in the phospholipase gene LIPH. Science 314:982–985

    Article  PubMed  CAS  Google Scholar 

  264. Pasternack SM, von Kugelgen I, Aboud KA, Lee YA, Ruschendorf F, Voss K, Hillmer AM, Molderings GJ, Franz T, Ramirez A, Nurnberg P, Nothen MM, Betz RC (2008) G protein-coupled receptor P2Y5 and its ligand LPA are involved in maintenance of human hair growth. Nat Genet 40:329–334

    Article  PubMed  CAS  Google Scholar 

  265. Irrthum A, Devriendt K, Chitayat D, Matthijs G, Glade C, Steijlen PM, Fryns JP, Van Steensel MA, Vikkula M (2003) Mutations in the transcription factor gene SOX18 underlie recessive and dominant forms of hypotrichosis-lymphedema-telangiectasia. Am J Hum Genet 72:1470–1478

    Article  PubMed  CAS  Google Scholar 

  266. Levy-Nissenbaum E, Betz RC, Frydman M, Simon M, Lahat H, Bakhan T, Goldman B, Bygum A, Pierick M, Hillmer AM, Jonca N, Toribio J, Kruse R, Dewald G, Cichon S, Kubisch C, Guerrin M, Serre G, Nothen MM, Pras E (2003) Hypotrichosis simplex of the scalp is associated with nonsense mutations in CDSN encoding corneodesmosin. Nat Genet 34:151–153

    Article  PubMed  CAS  Google Scholar 

  267. Shimomura Y, Agalliu D, Vonica A, Luria V, Wajid M, Baumer A, Belli S, Petukhova L, Schinzel A, Brivanlou AH, Barres BA, Christiano AM (2010) APCDD1 is a novel Wnt inhibitor mutated in hereditary hypotrichosis simplex. Nature 464:1043–1047

    Article  PubMed  CAS  Google Scholar 

  268. Amselem S, Duquesnoy P, Attree O, Novelli G, Bousnina S, Postel-Vinay MC, Goossens M (1989) Laron dwarfism and mutations of the growth hormone-receptor gene. N Engl J Med 321:989–995

    Article  PubMed  CAS  Google Scholar 

  269. Calvo SE, Tucker EJ, Compton AG, Kirby DM, Crawford G, Burtt NP, Rivas M, Guiducci C, Bruno DL, Goldberger OA, Redman MC, Wiltshire E, Wilson CJ, Altshuler D, Gabriel SB, Daly MJ, Thorburn DR, Mootha VK (2010) High-throughput, pooled sequencing identifies mutations in NUBPL and FOXRED1 in human complex I deficiency. Nat Genet 42:851–858

    Article  PubMed  CAS  Google Scholar 

  270. Ogilvie I, Kennaway NG, Shoubridge EA (2005) A molecular chaperone for mitochondrial complex I assembly is mutated in a progressive encephalopathy. J Clin Invest 115:2784–2792

    Article  PubMed  CAS  Google Scholar 

  271. Sugiana C, Pagliarini DJ, McKenzie M, Kirby DM, Salemi R, Abu-Amero KK, Dahl HH, Hutchison WM, Vascotto KA, Smith SM, Newbold RF, Christodoulou J, Calvo S, Mootha VK, Ryan MT, Thorburn DR (2008) Mutation of C20orf7 disrupts complex I assembly and causes lethal neonatal mitochondrial disease. Am J Hum Genet 83:468–478

    Article  PubMed  CAS  Google Scholar 

  272. Pagliarini DJ, Calvo SE, Chang B, Sheth SA, Vafai SB, Ong SE, Walford GA, Sugiana C, Boneh A, Chen WK, Hill DE, Vidal M, Evans JG, Thorburn DR, Carr SA, Mootha VK (2008) A mitochondrial protein compendium elucidates complex I disease biology. Cell 134:112–123

    Article  PubMed  CAS  Google Scholar 

  273. Schuelke M, Smeitink J, Mariman E, Loeffen J, Plecko B, Trijbels F, Stockler-Ipsiroglu S, van den Heuvel L (1999) Mutant NDUFV1 subunit of mitochondrial complex I causes leukodystrophy and myoclonic epilepsy. Nat Genet 21:260–261

    Article  PubMed  CAS  Google Scholar 

  274. Benit P, Chretien D, Kadhom N, de Lonlay-Debeney P, Cormier-Daire V, Cabral A, Peudenier S, Rustin P, Munnich A, Rotig A (2001) Large-scale deletion and point mutations of the nuclear NDUFV1 and NDUFS1 genes in mitochondrial complex I deficiency. Am J Hum Genet 68:1344–1352

    Article  PubMed  CAS  Google Scholar 

  275. Benit P, Slama A, Cartault F, Giurgea I, Chretien D, Lebon S, Marsac C, Munnich A, Rotig A, Rustin P (2004) Mutant NDUFS3 subunit of mitochondrial complex I causes Leigh syndrome. J Med Genet 41:14–17

    Article  PubMed  CAS  Google Scholar 

  276. van den Heuvel L, Ruitenbeek W, Smeets R, Gelman-Kohan Z, Elpeleg O, Loeffen J, Trijbels F, Mariman E, de Bruijn D, Smeitink J (1998) Demonstration of a new pathogenic mutation in human complex I deficiency: a 5-bp duplication in the nuclear gene encoding the 18-kD (AQDQ) subunit. Am J Hum Genet 62:262–268

    Article  PubMed  Google Scholar 

  277. Smeitink J, van den Heuvel L (1999) Human mitochondrial complex I in health and disease. Am J Hum Genet 64:1505–1510

    Article  PubMed  CAS  Google Scholar 

  278. Loeffen J, Smeitink J, Triepels R, Smeets R, Schuelke M, Sengers R, Trijbels F, Hamel B, Mullaart R, van den Heuvel L (1998) The first nuclear-encoded complex I mutation in a patient with Leigh syndrome. Am J Hum Genet 63:1598–1608

    Article  PubMed  CAS  Google Scholar 

  279. Hoefs SJ, Dieteren CE, Distelmaier F, Janssen RJ, Epplen A, Swarts HG, Forkink M, Rodenburg RJ, Nijtmans LG, Willems PH, Smeitink JA, van den Heuvel LP (2008) NDUFA2 complex I mutation leads to Leigh disease. Am J Hum Genet 82:1306–1315

    Article  PubMed  CAS  Google Scholar 

  280. Tanaka M, Gong JS, Zhang J, Yoneda M, Yagi K (1998) Mitochondrial genotype associated with longevity. Lancet 351:185–186

    Article  PubMed  CAS  Google Scholar 

  281. McFarland R, Kirby DM, Fowler KJ, Ohtake A, Ryan MT, Amor DJ, Fletcher JM, Dixon JW, Collins FA, Turnbull DM, Taylor RW, Thorburn DR (2004) De novo mutations in the mitochondrial ND3 gene as a cause of infantile mitochondrial encephalopathy and complex I deficiency. Ann Neurol 55:58–64

    Article  PubMed  CAS  Google Scholar 

  282. Taylor RW, Morris AA, Hutchinson M, Turnbull DM (2002) Leigh disease associated with a novel mitochondrial DNA ND5 mutation. Eur J Hum Genet 10:141–144

    Article  PubMed  CAS  Google Scholar 

  283. Ugalde C, Triepels RH, Coenen MJ, van den Heuvel LP, Smeets R, Uusimaa J, Briones P, Campistol J, Majamaa K, Smeitink JA, Nijtmans LG (2003) Impaired complex I assembly in a Leigh syndrome patient with a novel missense mutation in the ND6 gene. Ann Neurol 54:665–669

    Article  PubMed  CAS  Google Scholar 

  284. Bourgeron T, Rustin P, Chretien D, Birch-Machin M, Bourgeois M, Viegas-Pequignot E, Munnich A, Rotig A (1995) Mutation of a nuclear succinate dehydrogenase gene results in mitochondrial respiratory chain deficiency. Nat Genet 11:144–149

    Article  PubMed  CAS  Google Scholar 

  285. de Lonlay P, Valnot I, Barrientos A, Gorbatyuk M, Tzagoloff A, Taanman JW, Benayoun E, Chretien D, Kadhom N, Lombes A, de Baulny HO, Niaudet P, Munnich A, Rustin P, Rotig A (2001) A mutant mitochondrial respiratory chain assembly protein causes complex III deficiency in patients with tubulopathy, encephalopathy and liver failure. Nat Genet 29:57–60

    Article  PubMed  CAS  Google Scholar 

  286. Miranda AF, Ishii S, DiMauro S, Shay JW (1989) Cytochrome c oxidase deficiency in Leigh’s syndrome: genetic evidence for a nuclear DNA-encoded mutation. Neurology 39:697–702

    Article  PubMed  CAS  Google Scholar 

  287. Tatuch Y, Christodoulou J, Feigenbaum A, Clarke JT, Wherret J, Smith C, Rudd N, Petrova-Benedict R, Robinson BH (1992) Heteroplasmic mtDNA mutation (T––G) at 8993 can cause Leigh disease when the percentage of abnormal mtDNA is high. Am J Hum Genet 50:852–858

    PubMed  CAS  Google Scholar 

  288. McFarland R, Clark KM, Morris AA, Taylor RW, Macphail S, Lightowlers RN, Turnbull DM (2002) Multiple neonatal deaths due to a homoplasmic mitochondrial DNA mutation. Nat Genet 30:145–146

    Article  PubMed  CAS  Google Scholar 

  289. Shoffner JM, Lott MT, Lezza AM, Seibel P, Ballinger SW, Wallace DC (1990) Myoclonic epilepsy and ragged-red fiber disease (MERRF) is associated with a mitochondrial DNA tRNA(Lys) mutation. Cell 61:931–937

    Article  PubMed  CAS  Google Scholar 

  290. Yoneda M, Tanno Y, Horai S, Ozawa T, Miyatake T, Tsuji S (1990) A common mitochondrial DNA mutation in the t-RNA(Lys) of patients with myoclonus epilepsy associated with ragged-red fibers. Biochem Int 21:789–796

    PubMed  CAS  Google Scholar 

  291. Santorelli FM, Tanji K, Sano M, Shanske S, El-Shahawi M, Kranz-Eble P, DiMauro S, De Vivo DC (1997) Maternally inherited encephalopathy associated with a single-base insertion in the mitochondrial tRNATrp gene. Ann Neurol 42:256–260

    Article  PubMed  CAS  Google Scholar 

  292. Yasukawa T, Suzuki T, Ueda T, Ohta S, Watanabe K (2000) Modification defect at anticodon wobble nucleotide of mitochondrial tRNAs(Leu)(UUR) with pathogenic mutations of mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes. J Biol Chem 275:4251–4257

    Article  PubMed  CAS  Google Scholar 

  293. Grafakou O, Oexle K, van den Heuvel L, Smeets R, Trijbels F, Goebel HH, Bosshard N, Superti-Furga A, Steinmann B, Smeitink J (2003) Leigh syndrome due to compound heterozygosity of dihydrolipoamide dehydrogenase gene mutations. Description of the first E3 splice site mutation. Eur J Pediatr 162:714–718

    Article  PubMed  CAS  Google Scholar 

  294. Lee N, Daly MJ, Delmonte T, Lander ES, Xu F, Hudson TJ, Mitchell GA, Morin CC, Robinson BH, Rioux JD (2001) A genomewide linkage-disequilibrium scan localizes the Saguenay-Lac-Saint-Jean cytochrome oxidase deficiency to 2p16. Am J Hum Genet 68:397–409

    Article  PubMed  CAS  Google Scholar 

  295. Matthews PM, Marchington DR, Squier M, Land J, Brown RM, Brown GK (1993) Molecular genetic characterization of an X-linked form of Leigh’s syndrome. Ann Neurol 33:652–655

    Article  PubMed  CAS  Google Scholar 

  296. Quinzii C, Naini A, Salviati L, Trevisson E, Navas P, Dimauro S, Hirano M (2006) A mutation in para-hydroxybenzoate-polyprenyl transferase (COQ2) causes primary coenzyme Q10 deficiency. Am J Hum Genet 78:345–349

    Article  PubMed  CAS  Google Scholar 

  297. Quinzii CM, Kattah AG, Naini A, Akman HO, Mootha VK, DiMauro S, Hirano M (2005) Coenzyme Q deficiency and cerebellar ataxia associated with an aprataxin mutation. Neurology 64:539–541

    Article  PubMed  CAS  Google Scholar 

  298. Lopez LC, Schuelke M, Quinzii CM, Kanki T, Rodenburg RJ, Naini A, Dimauro S, Hirano M (2006) Leigh syndrome with nephropathy and CoQ10 deficiency due to decaprenyl diphosphate synthase subunit 2 (PDSS2) mutations. Am J Hum Genet 79:1125–1129

    Article  PubMed  CAS  Google Scholar 

  299. Mollet J, Delahodde A, Serre V, Chretien D, Schlemmer D, Lombes A, Boddaert N, Desguerre I, de Lonlay P, de Baulny HO, Munnich A, Rotig A (2008) CABC1 gene mutations cause ubiquinone deficiency with cerebellar ataxia and seizures. Am J Hum Genet 82:623–630

    Article  PubMed  CAS  Google Scholar 

  300. Mollet J, Giurgea I, Schlemmer D, Dallner G, Chretien D, Delahodde A, Bacq D, de Lonlay P, Munnich A, Rotig A (2007) Prenyldiphosphate synthase, subunit 1 (PDSS1) and OH-benzoate polyprenyltransferase (COQ2) mutations in ubiquinone deficiency and oxidative phosphorylation disorders. J Clin Invest 117:765–772

    Article  PubMed  CAS  Google Scholar 

  301. Duncan AJ, Bitner-Glindzicz M, Meunier B, Costello H, Hargreaves IP, Lopez LC, Hirano M, Quinzii CM, Sadowski MI, Hardy J, Singleton A, Clayton PT, Rahman S (2009) A nonsense mutation in COQ9 causes autosomal-recessive neonatal-onset primary coenzyme Q10 deficiency: a potentially treatable form of mitochondrial disease. Am J Hum Genet 84:558–566

    Article  PubMed  CAS  Google Scholar 

  302. Wen Y, Liu Y, Xu Y, Zhao Y, Hua R, Wang K, Sun M, Li Y, Yang S, Zhang XJ, Kruse R, Cichon S, Betz RC, Nothen MM, van Steensel MA, van Geel M, Steijlen PM, Hohl D, Huber M, Dunnill GS, Kennedy C, Messenger A, Munro CS, Terrinoni A, Hovnanian A, Bodemer C, de Prost Y, Paller AS, Irvine AD, Sinclair R, Green J, Shang D, Liu Q, Luo Y, Jiang L, Chen HD, Lo WH, McLean WH, He CD, Zhang X (2009) Loss-of-function mutations of an inhibitory upstream ORF in the human hairless transcript cause Marie Unna hereditary hypotrichosis. Nat Genet 41:228–233

    Article  PubMed  CAS  Google Scholar 

  303. Mercer JF, Livingston J, Hall B, Paynter JA, Begy C, Chandrasekharappa S, Lockhart P, Grimes A, Bhave M, Siemieniak D et al (1993) Isolation of a partial candidate gene for Menkes disease by positional cloning. Nat Genet 3:20–25

    Article  PubMed  CAS  Google Scholar 

  304. Chelly J, Tumer Z, Tonnesen T, Petterson A, Ishikawa-Brush Y, Tommerup N, Horn N, Monaco AP (1993) Isolation of a candidate gene for Menkes disease that encodes a potential heavy metal binding protein. Nat Genet 3:14–19

    Article  PubMed  CAS  Google Scholar 

  305. Vulpe C, Levinson B, Whitney S, Packman S, Gitschier J (1993) Isolation of a candidate gene for Menkes disease and evidence that it encodes a copper-transporting ATPase. Nat Genet 3:7–13

    Article  PubMed  CAS  Google Scholar 

  306. Celep F, Uzumcu A, Sonmez FM, Uyguner O, Balci YI, Bahadir S, Karaguzel A (2009) Pitfalls of mapping a large Turkish consanguineous family with vertical monilethrix inheritance. Genet Couns 20:1–8

    PubMed  CAS  Google Scholar 

  307. van Steensel MA, Steijlen PM, Bladergroen RS, Vermeer M, van Geel M (2005) A missense mutation in the type II hair keratin hHb3 is associated with monilethrix. J Med Genet 42:e19

    Article  PubMed  Google Scholar 

  308. Winter H, Rogers MA, Langbein L, Stevens HP, Leigh IM, Labreze C, Roul S, Taieb A, Krieg T, Schweizer J (1997) Mutations in the hair cortex keratin hHb6 cause the inherited hair disease monilethrix. Nat Genet 16:372–374

    Article  PubMed  CAS  Google Scholar 

  309. Dreyer SD, Zhou G, Baldini A, Winterpacht A, Zabel B, Cole W, Johnson RL, Lee B (1998) Mutations in LMX1B cause abnormal skeletal patterning and renal dysplasia in nail patella syndrome. Nat Genet 19:47–50

    Article  PubMed  CAS  Google Scholar 

  310. Frank J, Pignata C, Panteleyev AA, Prowse DM, Baden H, Weiner L, Gaetaniello L, Ahmad W, Pozzi N, Cserhalmi-Friedman PB, Aita VM, Uyttendaele H, Gordon D, Ott J, Brissette JL, Christiano AM (1999) Exposing the human nude phenotype. Nature 398:473–474

    Article  PubMed  CAS  Google Scholar 

  311. Nakabayashi K, Amann D, Ren Y, Saarialho-Kere U, Avidan N, Gentles S, MacDonald JR, Puffenberger EG, Christiano AM, Martinez-Mir A, Salas-Alanis JC, Rizzo R, Vamos E, Raams A, Les C, Seboun E, Jaspers NG, Beckmann JS, Jackson CE, Scherer SW (2005) Identification of C7orf11 (TTDN1) gene mutations and genetic heterogeneity in nonphotosensitive trichothiodystrophy. Am J Hum Genet 76:510–516

    Article  PubMed  CAS  Google Scholar 

  312. Broughton BC, Berneburg M, Fawcett H, Taylor EM, Arlett CF, Nardo T, Stefanini M, Menefee E, Price VH, Queille S, Sarasin A, Bohnert E, Krutmann J, Davidson R, Kraemer KH, Lehmann AR (2001) Two individuals with features of both xeroderma pigmentosum and trichothiodystrophy highlight the complexity of the clinical outcomes of mutations in the XPD gene. Hum Mol Genet 10:2539–2547

    Article  PubMed  CAS  Google Scholar 

  313. Vermeulen W, Bergmann E, Auriol J, Rademakers S, Frit P, Appeldoorn E, Hoeijmakers JH, Egly JM (2000) Sublimiting concentration of TFIIH transcription/DNA repair factor causes TTD-A trichothiodystrophy disorder. Nat Genet 26:307–313

    Article  PubMed  CAS  Google Scholar 

  314. Weeda G, Eveno E, Donker I, Vermeulen W, Chevallier-Lagente O, Taieb A, Stary A, Hoeijmakers JH, Mezzina M, Sarasin A (1997) A mutation in the XPB/ERCC3 DNA repair transcription gene, associated with trichothiodystrophy. Am J Hum Genet 60:320–329

    PubMed  CAS  Google Scholar 

  315. Hughes MR, Malloy PJ, Kieback DG, Kesterson RA, Pike JW, Feldman D, O’Malley BW (1988) Point mutations in the human vitamin D receptor gene associated with hypocalcemic rickets. Science 242:1702–1705

    Article  PubMed  CAS  Google Scholar 

  316. Shimomura Y, Wajid M, Petukhova L, Kurban M, Christiano AM (2010) Autosomal-dominant woolly hair resulting from disruption of keratin 74 (KRT74), a potential determinant of human hair texture. Am J Hum Genet 86:632–638

    Article  PubMed  CAS  Google Scholar 

  317. Shimomura Y, Wajid M, Ishii Y, Shapiro L, Petukhova L, Gordon D, Christiano AM (2008) Disruption of P2RY5, an orphan G protein-coupled receptor, underlies autosomal recessive woolly hair. Nat Genet 40:335–339

    Article  PubMed  CAS  Google Scholar 

  318. Morell R, Spritz RA, Ho L, Pierpont J, Guo W, Friedman TB, Asher JH Jr (1997) Apparent digenic inheritance of Waardenburg syndrome type 2 (WS2) and autosomal recessive ocular albinism (AROA). Hum Mol Genet 6:659–664

    Article  PubMed  CAS  Google Scholar 

  319. Tripathi RK, Strunk KM, Giebel LB, Weleber RG, Spritz RA (1992) Tyrosinase gene mutations in type I (tyrosinase-deficient) oculocutaneous albinism define two clusters of missense substitutions. Am J Med Genet 43:865–871

    Article  PubMed  CAS  Google Scholar 

  320. Giebel LB, Strunk KM, King RA, Hanifin JM, Spritz RA (1990) A frequent tyrosinase gene mutation in classic, tyrosinase-negative (type IA) oculocutaneous albinism. Proc Natl Acad Sci U S A 87:3255–3258

    Article  PubMed  CAS  Google Scholar 

  321. Durham-Pierre D, Gardner JM, Nakatsu Y, King RA, Francke U, Ching A, Aquaron R, del Marmol V, Brilliant MH (1994) African origin of an intragenic deletion of the human P gene in tyrosinase positive oculocutaneous albinism. Nat Genet 7:176–179

    Article  PubMed  CAS  Google Scholar 

  322. Boissy RE, Zhao H, Oetting WS, Austin LM, Wildenberg SC, Boissy YL, Zhao Y, Sturm RA, Hearing VJ, King RA, Nordlund JJ (1996) Mutation in and lack of expression of tyrosinase-related protein-1 (TRP-1) in melanocytes from an individual with brown oculocutaneous albinism: a new subtype of albinism classified as “OCA3”. Am J Hum Genet 58:1145–1156

    PubMed  CAS  Google Scholar 

  323. Newton JM, Cohen-Barak O, Hagiwara N, Gardner JM, Davisson MT, King RA, Brilliant MH (2001) Mutations in the human orthologue of the mouse underwhite gene (uw) underlie a new form of oculocutaneous albinism, OCA4. Am J Hum Genet 69:981–988

    Article  PubMed  CAS  Google Scholar 

  324. Barbosa MD, Barrat FJ, Tchernev VT, Nguyen QA, Mishra VS, Colman SD, Pastural E, Dufourcq-Lagelouse R, Fischer A, Holcombe RF, Wallace MR, Brandt SJ, de Saint Basile G, Kingsmore SF (1997) Identification of mutations in two major mRNA isoforms of the Chediak-Higashi syndrome gene in human and mouse. Hum Mol Genet 6:1091–1098

    Article  PubMed  CAS  Google Scholar 

  325. Miyamura Y, Suzuki T, Kono M, Inagaki K, Ito S, Suzuki N, Tomita Y (2003) Mutations of the RNA-specific adenosine deaminase gene (DSRAD) are involved in dyschromatosis symmetrica hereditaria. Am J Hum Genet 73:693–699

    Article  PubMed  CAS  Google Scholar 

  326. Pastural E, Barrat FJ, Dufourcq-Lagelouse R, Certain S, Sanal O, Jabado N, Seger R, Griscelli C, Fischer A, de Saint Basile G (1997) Griscelli disease maps to chromosome 15q21 and is associated with mutations in the myosin-Va gene. Nat Genet 16:289–292

    Article  PubMed  CAS  Google Scholar 

  327. Menasche G, Pastural E, Feldmann J, Certain S, Ersoy F, Dupuis S, Wulffraat N, Bianchi D, Fischer A, Le Deist F, de Saint Basile G (2000) Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome. Nat Genet 25:173–176

    Article  PubMed  CAS  Google Scholar 

  328. Menasche G, Ho CH, Sanal O, Feldmann J, Tezcan I, Ersoy F, Houdusse A, Fischer A, de Saint Basile G (2003) Griscelli syndrome restricted to hypopigmentation results from a melanophilin defect (GS3) or a MYO5A F-exon deletion (GS1). J Clin Invest 112:450–456

    PubMed  CAS  Google Scholar 

  329. Oh J, Bailin T, Fukai K, Feng GH, Ho L, Mao JI, Frenk E, Tamura N, Spritz RA (1996) Positional cloning of a gene for Hermansky-Pudlak syndrome, a disorder of cytoplasmic organelles. Nat Genet 14:300–306

    Article  PubMed  CAS  Google Scholar 

  330. Dell’Angelica EC, Shotelersuk V, Aguilar RC, Gahl WA, Bonifacino JS (1999) Altered trafficking of lysosomal proteins in Hermansky-Pudlak syndrome due to mutations in the beta 3A subunit of the AP-3 adaptor. Mol Cell 3:11–21

    Article  PubMed  Google Scholar 

  331. Anikster Y, Huizing M, White J, Shevchenko YO, Fitzpatrick DL, Touchman JW, Compton JG, Bale SJ, Swank RT, Gahl WA, Toro JR (2001) Mutation of a new gene causes a unique form of Hermansky-Pudlak syndrome in a genetic isolate of central Puerto Rico. Nat Genet 28:376–380

    Article  PubMed  CAS  Google Scholar 

  332. Suzuki T, Li W, Zhang Q, Karim A, Novak EK, Sviderskaya EV, Hill SP, Bennett DC, Levin AV, Nieuwenhuis HK, Fong CT, Castellan C, Miterski B, Swank RT, Spritz RA (2002) Hermansky-Pudlak syndrome is caused by mutations in HPS4, the human homolog of the mouse light-ear gene. Nat Genet 30:321–324

    PubMed  Google Scholar 

  333. Zhang Q, Zhao B, Li W, Oiso N, Novak EK, Rusiniak ME, Gautam R, Chintala S, O’Brien EP, Zhang Y, Roe BA, Elliott RW, Eicher EM, Liang P, Kratz C, Legius E, Spritz RA, O’Sullivan TN, Copeland NG, Jenkins NA, Swank RT (2003) Ru2 and Ru encode mouse orthologs of the genes mutated in human Hermansky-Pudlak syndrome types 5 and 6. Nat Genet 33:145–153

    Article  PubMed  CAS  Google Scholar 

  334. Li W, Zhang Q, Oiso N, Novak EK, Gautam R, O’Brien EP, Tinsley CL, Blake DJ, Spritz RA, Copeland NG, Jenkins NA, Amato D, Roe BA, Starcevic M, Dell’Angelica EC, Elliott RW, Mishra V, Kingsmore SF, Paylor RE, Swank RT (2003) Hermansky-Pudlak syndrome type 7 (HPS-7) results from mutant dysbindin, a member of the biogenesis of lysosome-related organelles complex 1 (BLOC-1). Nat Genet 35:84–89

    Article  PubMed  CAS  Google Scholar 

  335. Morgan NV, Pasha S, Johnson CA, Ainsworth JR, Eady RA, Dawood B, McKeown C, Trembath RC, Wilde J, Watson SP, Maher ER (2006) A germline mutation in BLOC1S3/reduced pigmentation causes a novel variant of Hermansky-Pudlak syndrome (HPS8). Am J Hum Genet 78:160–166

    Article  PubMed  CAS  Google Scholar 

  336. Smahi A, Courtois G, Vabres P, Yamaoka S, Heuertz S, Munnich A, Israel A, Heiss NS, Klauck SM, Kioschis P, Wiemann S, Poustka A, Esposito T, Bardaro T, Gianfrancesco F, Ciccodicola A, D’Urso M, Woffendin H, Jakins T, Donnai D, Stewart H, Kenwrick SJ, Aradhya S, Yamagata T, Levy M, Lewis RA, Nelson DL (2000) Genomic rearrangement in NEMO impairs NF-kappaB activation and is a cause of incontinentia pigmenti. The International Incontinentia Pigmenti (IP) Consortium. Nature 405:466–472

    Article  PubMed  CAS  Google Scholar 

  337. Brems H, Chmara M, Sahbatou M, Denayer E, Taniguchi K, Kato R, Somers R, Messiaen L, De Schepper S, Fryns JP, Cools J, Marynen P, Thomas G, Yoshimura A, Legius E (2007) Germline loss-of-function mutations in SPRED1 cause a neurofibromatosis 1-like phenotype. Nat Genet 39:1120–1126

    Article  PubMed  CAS  Google Scholar 

  338. Digilio MC, Conti E, Sarkozy A, Mingarelli R, Dottorini T, Marino B, Pizzuti A, Dallapiccola B (2002) Grouping of multiple-lentigines/LEOPARD and Noonan syndromes on the PTPN11 gene. Am J Hum Genet 71:389–394

    Article  PubMed  CAS  Google Scholar 

  339. Weinstein LS, Shenker A, Gejman PV, Merino MJ, Friedman E, Spiegel AM (1991) Activating mutations of the stimulatory G protein in the McCune-Albright syndrome. N Engl J Med 325:1688–1695

    Article  PubMed  CAS  Google Scholar 

  340. Touraine RL, Attie-Bitach T, Manceau E, Korsch E, Sarda P, Pingault V, Encha-Razavi F, Pelet A, Auge J, Nivelon-Chevallier A, Holschneider AM, Munnes M, Doerfler W, Goossens M, Munnich A, Vekemans M, Lyonnet S (2000) Neurological phenotype in Waardenburg syndrome type 4 correlates with novel SOX10 truncating mutations and expression in developing brain. Am J Hum Genet 66:1496–1503

    Article  PubMed  CAS  Google Scholar 

  341. Giebel LB, Spritz RA (1991) Mutation of the KIT (mast/stem cell growth factor receptor) protooncogene in human piebaldism. Proc Natl Acad Sci U S A 88:8696–8699

    Article  PubMed  CAS  Google Scholar 

  342. Volpi L, Roversi G, Colombo EA, Leijsten N, Concolino D, Calabria A, Mencarelli MA, Fimiani M, Macciardi F, Pfundt R, Schoenmakers EF, Larizza L (2010) Targeted next-generation sequencing appoints c16orf57 as clericuzio-type poikiloderma with neutropenia gene. Am J Hum Genet 86:72–76

    Article  PubMed  CAS  Google Scholar 

  343. Tassabehji M, Newton VE, Liu XZ, Brady A, Donnai D, Krajewska-Walasek M, Murday V, Norman A, Obersztyn E, Reardon W et al (1995) The mutational spectrum in Waardenburg syndrome. Hum Mol Genet 4:2131–2137

    Article  PubMed  CAS  Google Scholar 

  344. Tassabehji M, Read AP, Newton VE, Harris R, Balling R, Gruss P, Strachan T (1992) Waardenburg’s syndrome patients have mutations in the human homologue of the Pax-3 paired box gene. Nature 355:635–636

    Article  PubMed  CAS  Google Scholar 

  345. Tassabehji M, Newton VE, Read AP (1994) Waardenburg syndrome type 2 caused by mutations in the human microphthalmia (MITF) gene. Nat Genet 8:251–255

    Article  PubMed  CAS  Google Scholar 

  346. Sanchez-Martin M, Rodriguez-Garcia A, Perez-Losada J, Sagrera A, Read AP, Sanchez-Garcia I (2002) SLUG (SNAI2) deletions in patients with Waardenburg disease. Hum Mol Genet 11:3231–3236

    Article  PubMed  CAS  Google Scholar 

  347. Bondurand N, Kuhlbrodt K, Pingault V, Enderich J, Sajus M, Tommerup N, Warburg M, Hennekam RC, Read AP, Wegner M, Goossens M (1999) A molecular analysis of the yemenite deaf-blind hypopigmentation syndrome: SOX10 dysfunction causes different neurocristopathies. Hum Mol Genet 8:1785–1789

    Article  PubMed  CAS  Google Scholar 

  348. Hoth CF, Milunsky A, Lipsky N, Sheffer R, Clarren SK, Baldwin CT (1993) Mutations in the paired domain of the human PAX3 gene cause Klein-Waardenburg syndrome (WS-III) as well as Waardenburg syndrome type I (WS-I). Am J Hum Genet 52:455–462

    PubMed  CAS  Google Scholar 

  349. Puffenberger EG, Hosoda K, Washington SS, Nakao K, deWit D, Yanagisawa M, Chakravart A (1994) A missense mutation of the endothelin-B receptor gene in multigenic Hirschsprung’s disease. Cell 79:1257–1266

    Article  PubMed  CAS  Google Scholar 

  350. Edery P, Attie T, Amiel J, Pelet A, Eng C, Hofstra RM, Martelli H, Bidaud C, Munnich A, Lyonnet S (1996) Mutation of the endothelin-3 gene in the Waardenburg-Hirschsprung disease (Shah-Waardenburg syndrome). Nat Genet 12:442–444

    Article  PubMed  CAS  Google Scholar 

  351. Pingault V, Bondurand N, Kuhlbrodt K, Goerich DE, Prehu MO, Puliti A, Herbarth B, Hermans-Borgmeyer I, Legius E, Matthijs G, Amiel J, Lyonnet S, Ceccherini I, Romeo G, Smith JC, Read AP, Wegner M, Goossens M (1998) SOX10 mutations in patients with Waardenburg-Hirschsprung disease. Nat Genet 18:171–173

    Article  PubMed  CAS  Google Scholar 

  352. Kury S, Dreno B, Bezieau S, Giraudet S, Kharfi M, Kamoun R, Moisan JP (2002) Identification of SLC39A4, a gene involved in acrodermatitis enteropathica. Nat Genet 31:239–240

    Article  PubMed  CAS  Google Scholar 

  353. Fernandez-Canon JM, Granadino B, Beltran-Valero de Bernabe D, Renedo M, Fernandez-Ruiz E, Penalva MA, Rodriguez de Cordoba S (1996) The molecular basis of alkaptonuria. Nat Genet 14:19–24

    Article  PubMed  CAS  Google Scholar 

  354. Ariga T, Igarashi T, Ramesh N, Parad R, Cicardi M, Davis AE 3rd (1989) Type I C1 inhibitor deficiency with a small messenger RNA resulting from deletion of one exon. J Clin Invest 83:1888–1893

    Article  PubMed  CAS  Google Scholar 

  355. Levy NJ, Ramesh N, Cicardi M, Harrison RA, Davis AE 3rd (1990) Type II hereditary angioneurotic edema that may result from a single nucleotide change in the codon for alanine-436 in the C1 inhibitor gene. Proc Natl Acad Sci U S A 87:265–268

    Article  PubMed  CAS  Google Scholar 

  356. Dewald G, Bork K (2006) Missense mutations in the coagulation factor XII (Hageman factor) gene in hereditary angioedema with normal C1 inhibitor. Biochem Biophys Res Commun 343:1286–1289

    Article  PubMed  CAS  Google Scholar 

  357. Connelly PW, Maguire GF, Hofmann T, Little JA (1987) Structure of apolipoprotein C-IIToronto, a nonfunctional human apolipoprotein. Proc Natl Acad Sci U S A 84:270–273

    Article  PubMed  CAS  Google Scholar 

  358. Pomponio RJ, Reynolds TR, Cole H, Buck GA, Wolf B (1995) Mutational hotspot in the human biotinidase gene causes profound biotinidase deficiency. Nat Genet 11:96–98

    Article  PubMed  CAS  Google Scholar 

  359. Cali JJ, Hsieh CL, Francke U, Russell DW (1991) Mutations in the bile acid biosynthetic enzyme sterol 27-hydroxylase underlie cerebrotendinous xanthomatosis. J Biol Chem 266:7779–7783

    PubMed  CAS  Google Scholar 

  360. Martasek P, Nordmann Y, Grandchamp B (1994) Homozygous hereditary coproporphyria caused by an arginine to tryptophane substitution in coproporphyrinogen oxidase and common intragenic polymorphisms. Hum Mol Genet 3:477–480

    Article  PubMed  CAS  Google Scholar 

  361. Bernstein HS, Bishop DF, Astrin KH, Kornreich R, Eng CM, Sakuraba H, Desnick RJ (1989) Fabry disease: six gene rearrangements and an exonic point mutation in the alpha-galactosidase gene. J Clin Invest 83:1390–1399

    Article  PubMed  CAS  Google Scholar 

  362. Koch J, Gartner S, Li CM, Quintern LE, Bernardo K, Levran O, Schnabel D, Desnick RJ, Schuchman EH, Sandhoff K (1996) Molecular cloning and characterization of a full-length complementary DNA encoding human acid ceramidase. Identification of the first molecular lesion causing Farber disease. J Biol Chem 271:33110–33115

    Article  PubMed  CAS  Google Scholar 

  363. Darby JK, Willems PJ, Nakashima P, Johnsen J, Ferrell RE, Wijsman EM, Gerhard DS, Dracopoli NC, Housman D, Henke J et al (1988) Restriction analysis of the structural alpha-L-fucosidase gene and its linkage to fucosidosis. Am J Hum Genet 43:749–755

    PubMed  CAS  Google Scholar 

  364. Willems PJ, Darby JK, DiCioccio RA, Nakashima P, Eng C, Kretz KA, Cavalli-Sforza LL, Shooter EM, O’Brien JS (1988) Identification of a mutation in the structural alpha-L-fucosidase gene in fucosidosis. Am J Hum Genet 43:756–763

    PubMed  CAS  Google Scholar 

  365. Bourgeron T, Chretien D, Poggi-Bach J, Doonan S, Rabier D, Letouze P, Munnich A, Rotig A, Landrieu P, Rustin P (1994) Mutation of the fumarase gene in two siblings with progressive encephalopathy and fumarase deficiency. J Clin Invest 93:2514–2518

    Article  PubMed  CAS  Google Scholar 

  366. Tsuji S, Choudary PV, Martin BM, Stubblefield BK, Mayor JA, Barranger JA, Ginns EI (1987) A mutation in the human glucocerebrosidase gene in neuronopathic Gaucher’s disease. N Engl J Med 316:570–575

    Article  PubMed  CAS  Google Scholar 

  367. Kleta R, Romeo E, Ristic Z, Ohura T, Stuart C, Arcos-Burgos M, Dave MH, Wagner CA, Camargo SR, Inoue S, Matsuura N, Helip-Wooley A, Bockenhauer D, Warth R, Bernardini I, Visser G, Eggermann T, Lee P, Chairoungdua A, Jutabha P, Babu E, Nilwarangkoon S, Anzai N, Kanai Y, Verrey F, Gahl WA, Koizumi A (2004) Mutations in SLC6A19, encoding B0AT1, cause Hartnup disorder. Nat Genet 36:999–1002

    Article  PubMed  CAS  Google Scholar 

  368. Feder JN, Gnirke A, Thomas W, Tsuchihashi Z, Ruddy DA, Basava A, Dormishian F, Domingo R Jr, Ellis MC, Fullan A, Hinton LM, Jones NL, Kimmel BE, Kronmal GS, Lauer P, Lee VK, Loeb DB, Mapa FA, McClelland E, Meyer NC, Mintier GA, Moeller N, Moore T, Morikang E, Prass CE, Quintana L, Starnes SM, Schatzman RC, Brunke KJ, Drayna DT, Risch NJ, Bacon BR, Wolff RK (1996) A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet 13:399–408

    Article  PubMed  CAS  Google Scholar 

  369. Milet J, Dehais V, Bourgain C, Jouanolle AM, Mosser A, Perrin M, Morcet J, Brissot P, David V, Deugnier Y, Mosser J (2007) Common variants in the BMP2, BMP4, and HJV genes of the hepcidin regulation pathway modulate HFE hemochromatosis penetrance. Am J Hum Genet 81:799–807

    Article  PubMed  CAS  Google Scholar 

  370. Papanikolaou G, Samuels ME, Ludwig EH, MacDonald ML, Franchini PL, Dube MP, Andres L, MacFarlane J, Sakellaropoulos N, Politou M, Nemeth E, Thompson J, Risler JK, Zaborowska C, Babakaiff R, Radomski CC, Pape TD, Davidas O, Christakis J, Brissot P, Lockitch G, Ganz T, Hayden MR, Goldberg YP (2004) Mutations in HFE2 cause iron overload in chromosome 1q-linked juvenile hemochromatosis. Nat Genet 36:77–82

    Article  PubMed  CAS  Google Scholar 

  371. Roetto A, Papanikolaou G, Politou M, Alberti F, Girelli D, Christakis J, Loukopoulos D, Camaschella C (2003) Mutant antimicrobial peptide hepcidin is associated with severe juvenile hemochromatosis. Nat Genet 33:21–22

    Article  PubMed  CAS  Google Scholar 

  372. Camaschella C, Roetto A, Cali A, De Gobbi M, Garozzo G, Carella M, Majorano N, Totaro A, Gasparini P (2000) The gene TFR2 is mutated in a new type of haemochromatosis mapping to 7q22. Nat Genet 25:14–15

    Article  PubMed  CAS  Google Scholar 

  373. Njajou OT, Vaessen N, Joosse M, Berghuis B, van Dongen JW, Breuning MH, Snijders PJ, Rutten WP, Sandkuijl LA, Oostra BA, van Duijn CM, Heutink P (2001) A mutation in SLC11A3 is associated with autosomal dominant hemochromatosis. Nat Genet 28:213–214

    Article  PubMed  CAS  Google Scholar 

  374. Suzuki Y, Aoki Y, Ishida Y, Chiba Y, Iwamatsu A, Kishino T, Niikawa N, Matsubara Y, Narisawa K (1994) Isolation and characterization of mutations in the human holocarboxylase synthetase cDNA. Nat Genet 8:122–128

    Article  PubMed  CAS  Google Scholar 

  375. Skovby F, Kraus JP, Rosenberg LE (1984) Homocystinuria: biogenesis of cystathionine beta-synthase subunits in cultured fibroblasts and in an in vitro translation system programmed with fibroblast messenger RNA. Am J Hum Genet 36:452–459

    PubMed  CAS  Google Scholar 

  376. Lehrman MA, Goldstein JL, Brown MS, Russell DW, Schneider WJ (1985) Internalization-defective LDL receptors produced by genes with nonsense and frameshift mutations that truncate the cytoplasmic domain. Cell 41:735–743

    Article  PubMed  CAS  Google Scholar 

  377. Young SG, Bertics SJ, Curtiss LK, Witztum JL (1987) Characterization of an abnormal species of apolipoprotein B, apolipoprotein B-37, associated with familial hypobetalipoproteinemia. J Clin Invest 79:1831–1841

    Article  PubMed  CAS  Google Scholar 

  378. Varret M, Rabes JP, Saint-Jore B, Cenarro A, Marinoni JC, Civeira F, Devillers M, Krempf M, Coulon M, Thiart R, Kotze MJ, Schmidt H, Buzzi JC, Kostner GM, Bertolini S, Pocovi M, Rosa A, Farnier M, Martinez M, Junien C, Boileau C (1999) A third major locus for autosomal dominant hypercholesterolemia maps to 1p34.1-p32. Am J Hum Genet 64:1378–1387

    Article  PubMed  CAS  Google Scholar 

  379. Funke H, Klug J, Assmann G (1987) Hind III RFLP in the lipoprotein lipase gene, (LPL). Nucleic Acids Res 15:9102

    Article  PubMed  CAS  Google Scholar 

  380. Heinzmann C, Ladias J, Antonarakis S, Kirchgessner T, Schotz M, Lusis AJ (1987) RFLP for the human lipoprotein lipase (LPL) gene: HindIII. Nucleic Acids Res 15:6763

    Article  PubMed  CAS  Google Scholar 

  381. Weisgraber KH, Rall SC Jr, Mahley RW (1981) Human E apoprotein heterogeneity. Cysteine-arginine interchanges in the amino acid sequence of the apo-E isoforms. J Biol Chem 256:9077–9083

    PubMed  CAS  Google Scholar 

  382. Howells DW, Forrest SM, Dahl HH, Cotton RG (1990) Insertion of an extra codon for threonine is a cause of dihydropteridine reductase deficiency. Am J Hum Genet 47:279–285

    PubMed  CAS  Google Scholar 

  383. Thony B, Leimbacher W, Blau N, Harvie A, Heizmann CW (1994) Hyperphenylalaninemia due to defects in tetrahydrobiopterin metabolism: molecular characterization of mutations in 6-pyruvoyl-tetrahydropterin synthase. Am J Hum Genet 54:782–792

    PubMed  CAS  Google Scholar 

  384. Karathanasis SK, McPherson J, Zannis VI, Breslow JL (1983) Linkage of human apolipoproteins A-I and C-III genes. Nature 304:371–373

    Article  PubMed  CAS  Google Scholar 

  385. Wen XY, Hegele RA, Wang J, Wang DY, Cheung J, Wilson M, Yahyapour M, Bai Y, Zhuang L, Skaug J, Young TK, Connelly PW, Koop BF, Tsui LC, Stewart AK (2003) Identification of a novel lipase gene mutated in lpd mice with hypertriglyceridemia and associated with dyslipidemia in humans. Hum Mol Genet 12:1131–1143

    Article  PubMed  CAS  Google Scholar 

  386. Fujita Y, Ezura Y, Emi M, Ono S, Takada D, Takahashi K, Uemura K, Iino Y, Katayama Y, Bujo H, Saito Y (2003) Hypertriglyceridemia associated with amino acid variation Asn985Tyr of the RP1 gene. J Hum Genet 48:305–308

    PubMed  CAS  Google Scholar 

  387. Brooks-Wilson A, Marcil M, Clee SM, Zhang LH, Roomp K, van Dam M, Yu L, Brewer C, Collins JA, Molhuizen HO, Loubser O, Ouelette BF, Fichter K, Ashbourne-Excoffon KJ, Sensen CW, Scherer S, Mott S, Denis M, Martindale D, Frohlich J, Morgan K, Koop B, Pimstone S, Kastelein JJ, Genest J Jr, Hayden MR (1999) Mutations in ABC1 in Tangier disease and familial high-density lipoprotein deficiency. Nat Genet 22:336–345

    Article  PubMed  CAS  Google Scholar 

  388. Utermann G, Feussner G, Franceschini G, Haas J, Steinmetz A (1982) Genetic variants of group A apolipoproteins. Rapid methods for screening and characterization without ultracentrifugation. J Biol Chem 257:501–507

    PubMed  CAS  Google Scholar 

  389. Levran O, Desnick RJ, Schuchman EH (1991) Niemann-Pick disease: a frequent missense mutation in the acid sphingomyelinase gene of Ashkenazi Jewish type A and B patients. Proc Natl Acad Sci U S A 88:3748–3752

    Article  PubMed  CAS  Google Scholar 

  390. DiLella AG, Kwok SC, Ledley FD, Marvit J, Woo SL (1986) Molecular structure and polymorphic map of the human phenylalanine hydroxylase gene. Biochemistry 25:743–749

    Article  PubMed  CAS  Google Scholar 

  391. Deybach JC, de Verneuil H, Phung N, Nordmann Y, Puissant A, Boffety B (1981) Congenital erythropoietic porphyria (Gunther’s disease): enzymatic studies on two cases of late onset. J Lab Clin Med 97:551–558

    PubMed  CAS  Google Scholar 

  392. Garey JR, Hansen JL, Harrison LM, Kennedy JB, Kushner JP (1989) A point mutation in the coding region of uroporphyrinogen decarboxylase associated with familial porphyria cutanea tarda. Blood 73:892–895

    PubMed  CAS  Google Scholar 

  393. Moran-Jimenez MJ, Ged C, Romana M, Enriquez De Salamanca R, Taieb A, Topi G, D’Alessandro L, de Verneuil H (1996) Uroporphyrinogen decarboxylase: complete human gene sequence and molecular study of three families with hepatoerythropoietic porphyria. Am J Hum Genet 58:712–721

    PubMed  CAS  Google Scholar 

  394. Deybach JC, Puy H, Robreau AM, Lamoril J, Da Silva V, Grandchamp B, Nordmann Y (1996) Mutations in the protoporphyrinogen oxidase gene in patients with variegate porphyria. Hum Mol Genet 5:407–410

    Article  PubMed  CAS  Google Scholar 

  395. Tanoue A, Endo F, Matsuda I (1990) Structural organization of the gene for human prolidase (peptidase D) and demonstration of a partial gene deletion in a patient with prolidase deficiency. J Biol Chem 265:11306–11311

    PubMed  CAS  Google Scholar 

  396. Lamoril J, Boulechfar S, de Verneuil H, Grandchamp B, Nordmann Y, Deybach JC (1991) Human erythropoietic protoporphyria: two point mutations in the ferrochelatase gene. Biochem Biophys Res Commun 181:594–599

    Article  PubMed  CAS  Google Scholar 

  397. Lee MH, Lu K, Hazard S, Yu H, Shulenin S, Hidaka H, Kojima H, Allikmets R, Sakuma N, Pegoraro R, Srivastava AK, Salen G, Dean M, Patel SB (2001) Identification of a gene, ABCG5, important in the regulation of dietary cholesterol absorption. Nat Genet 27:79–83

    Article  PubMed  CAS  Google Scholar 

  398. Berge KE, Tian H, Graf GA, Yu L, Grishin NV, Schultz J, Kwiterovich P, Shan B, Barnes R, Hobbs HH (2000) Accumulation of dietary cholesterol in sitosterolemia caused by mutations in adjacent ABC transporters. Science 290:1771–1775

    Article  PubMed  CAS  Google Scholar 

  399. Bull PC, Thomas GR, Rommens JM, Forbes JR, Cox DW (1993) The Wilson disease gene is a putative copper transporting P-type ATPase similar to the Menkes gene. Nat Genet 5:327–337

    Article  PubMed  CAS  Google Scholar 

  400. Miceli-Richard C, Lesage S, Rybojad M, Prieur AM, Manouvrier-Hanu S, Hafner R, Chamaillard M, Zouali H, Thomas G, Hugot JP (2001) CARD15 mutations in Blau syndrome. Nat Genet 29:19–20

    Article  PubMed  CAS  Google Scholar 

  401. Feldmann J, Prieur AM, Quartier P, Berquin P, Certain S, Cortis E, Teillac-Hamel D, Fischer A, de Saint Basile G (2002) Chronic infantile neurological cutaneous and articular syndrome is caused by mutations in CIAS1, a gene highly expressed in polymorphonuclear cells and chondrocytes. Am J Hum Genet 71:198–203

    Article  PubMed  CAS  Google Scholar 

  402. Horwitz M, Benson KF, Person RE, Aprikyan AG, Dale DC (1999) Mutations in ELA2, encoding neutrophil elastase, define a 21-day biological clock in cyclic haematopoiesis. Nat Genet 23:433–436

    Article  PubMed  CAS  Google Scholar 

  403. Hoffman HM, Mueller JL, Broide DH, Wanderer AA, Kolodner RD (2001) Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome. Nat Genet 29:301–305

    Article  PubMed  CAS  Google Scholar 

  404. French FMF Consortium (1997) A candidate gene for familial Mediterranean fever. Nat Genet. 17:25–31

    Google Scholar 

  405. Booth DR, Gillmore JD, Lachmann HJ, Booth SE, Bybee A, Soyturk M, Akar S, Pepys MB, Tunca M, Hawkins PN (2000) The genetic basis of autosomal dominant familial Mediterranean fever. QJM 93:217–221

    Article  PubMed  CAS  Google Scholar 

  406. Houten SM, Kuis W, Duran M, de Koning TJ, van Royen-Kerkhof A, Romeijn GJ, Frenkel J, Dorland L, de Barse MM, Huijbers WA, Rijkers GT, Waterham HR, Wanders RJ, Poll-The BT (1999) Mutations in MVK, encoding mevalonate kinase, cause hyperimmunoglobulinaemia D and periodic fever syndrome. Nat Genet 22:175–177

    Article  PubMed  CAS  Google Scholar 

  407. Ferguson PJ, Chen S, Tayeh MK, Ochoa L, Leal SM, Pelet A, Munnich A, Lyonnet S, Majeed HA, El-Shanti H (2005) Homozygous mutations in LPIN2 are responsible for the syndrome of chronic recurrent multifocal osteomyelitis and congenital dyserythropoietic anaemia (Majeed syndrome). J Med Genet 42:551–557

    Article  PubMed  CAS  Google Scholar 

  408. McDermott MF, Aksentijevich I, Galon J, McDermott EM, Ogunkolade BW, Centola M, Mansfield E, Gadina M, Karenko L, Pettersson T, McCarthy J, Frucht DM, Aringer M, Torosyan Y, Teppo AM, Wilson M, Karaarslan HM, Wan Y, Todd I, Wood G, Schlimgen R, Kumarajeewa TR, Cooper SM, Vella JP, Amos CI, Mulley J, Quane KA, Molloy MG, Ranki A, Powell RJ, Hitman GA, O’Shea JJ, Kastner DL (1999) Germline mutations in the extracellular domains of the 55 kDa TNF receptor, TNFR1, define a family of dominantly inherited autoinflammatory syndromes. Cell 97:133–144

    Article  PubMed  CAS  Google Scholar 

  409. Wise CA, Gillum JD, Seidman CE, Lindor NM, Veile R, Bashiardes S, Lovett M (2002) Mutations in CD2BP1 disrupt binding to PTP PEST and are responsible for PAPA syndrome, an autoinflammatory disorder. Hum Mol Genet 11:961–969

    Article  PubMed  CAS  Google Scholar 

  410. Psiachou H, Mitton S, Alaghband-Zadeh J, Hone J, Taylor SI, Sinclair L (1993) Leprechaunism and homozygous nonsense mutation in the insulin receptor gene. Lancet 342:924

    Article  PubMed  CAS  Google Scholar 

  411. Agarwal AK, Arioglu E, De Almeida S, Akkoc N, Taylor SI, Bowcock AM, Barnes RI, Garg A (2002) AGPAT2 is mutated in congenital generalized lipodystrophy linked to chromosome 9q34. Nat Genet 31:21–23

    Article  PubMed  CAS  Google Scholar 

  412. Magre J, Delepine M, Khallouf E, Gedde-Dahl T Jr, Van Maldergem L, Sobel E, Papp J, Meier M, Megarbane A, Bachy A, Verloes A, d’Abronzo FH, Seemanova E, Assan R, Baudic N, Bourut C, Czernichow P, Huet F, Grigorescu F, de Kerdanet M, Lacombe D, Labrune P, Lanza M, Loret H, Matsuda F, Navarro J, Nivelon-Chevalier A, Polak M, Robert JJ, Tric P, Tubiana-Rufi N, Vigouroux C, Weissenbach J, Savasta S, Maassen JA, Trygstad O, Bogalho P, Freitas P, Medina JL, Bonnicci F, Joffe BI, Loyson G, Panz VR, Raal FJ, O’Rahilly S, Stephenson T, Kahn CR, Lathrop M, Capeau J (2001) Identification of the gene altered in Berardinelli-Seip congenital lipodystrophy on chromosome 11q13. Nat Genet 28:365–370

    Article  PubMed  CAS  Google Scholar 

  413. Cao H, Hegele RA (2000) Nuclear lamin A/C R482Q mutation in canadian kindreds with Dunnigan-type familial partial lipodystrophy. Hum Mol Genet 9:109–112

    Article  PubMed  CAS  Google Scholar 

  414. Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe JW, Soos MA, Maslen GL, Williams TD, Lewis H, Schafer AJ, Chatterjee VK, O’Rahilly S (1999) Dominant negative mutations in human PPARgamma associated with severe insulin resistance, diabetes ­mellitus and hypertension. Nature 402:880–883

    PubMed  CAS  Google Scholar 

  415. Hegele RA, Cao H, Liu DM, Costain GA, Charlton-Menys V, Rodger NW, Durrington PN (2006) Sequencing of the reannotated LMNB2 gene reveals novel mutations in patients with acquired partial lipodystrophy. Am J Hum Genet 79:383–389

    Article  PubMed  CAS  Google Scholar 

  416. Henning KA, Li L, Iyer N, McDaniel LD, Reagan MS, Legerski R, Schultz RA, Stefanini M, Lehmann AR, Mayne LV, Friedberg EC (1995) The Cockayne syndrome group A gene encodes a WD repeat protein that interacts with CSB protein and a subunit of RNA polymerase II TFIIH. Cell 82:555–564

    Article  PubMed  CAS  Google Scholar 

  417. Mallery DL, Tanganelli B, Colella S, Steingrimsdottir H, van Gool AJ, Troelstra C, Stefanini M, Lehmann AR (1998) Molecular analysis of mutations in the CSB (ERCC6) gene in patients with Cockayne syndrome. Am J Hum Genet 62:77–85

    Article  PubMed  CAS  Google Scholar 

  418. Eriksson M, Brown WT, Gordon LB, Glynn MW, Singer J, Scott L, Erdos MR, Robbins CM, Moses TY, Berglund P, Dutra A, Pak E, Durkin S, Csoka AB, Boehnke M, Glover TW, Collins FS (2003) Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature 423:293–298

    Article  PubMed  CAS  Google Scholar 

  419. Novelli G, Muchir A, Sangiuolo F, Helbling-Leclerc A, D’Apice MR, Massart C, Capon F, Sbraccia P, Federici M, Lauro R, Tudisco C, Pallotta R, Scarano G, Dallapiccola B, Merlini L, Bonne G (2002) Mandibuloacral dysplasia is caused by a mutation in LMNA-encoding lamin A/C. Am J Hum Genet 71:426–431

    Article  PubMed  CAS  Google Scholar 

  420. Simha V, Agarwal AK, Oral EA, Fryns JP, Garg A (2003) Genetic and phenotypic heterogeneity in patients with mandibuloacral dysplasia-associated lipodystrophy. J Clin Endocrinol Metab 88:2821–2824

    Article  PubMed  CAS  Google Scholar 

  421. Yu CE, Oshima J, Fu YH, Wijsman EM, Hisama F, Alisch R, Matthews S, Nakura J, Miki T, Ouais S, Martin GM, Mulligan J, Schellenberg GD (1996) Positional cloning of the Werner’s syndrome gene. Science 272:258–262

    Article  PubMed  CAS  Google Scholar 

  422. Eerola I, Boon LM, Mulliken JB, Burrows PE, Dompmartin A, Watanabe S, Vanwijck R, Vikkula M (2003) Capillary malformation-arteriovenous malformation, a new clinical and genetic disorder caused by RASA1 mutations. Am J Hum Genet 73:1240–1249

    Article  PubMed  CAS  Google Scholar 

  423. Laberge-le Couteulx S, Jung HH, Labauge P, Houtteville JP, Lescoat C, Cecillon M, Marechal E, Joutel A, Bach JF, Tournier-Lasserve E (1999) Truncating mutations in CCM1, encoding KRIT1, cause hereditary cavernous angiomas. Nat Genet 23:189–193

    Article  PubMed  CAS  Google Scholar 

  424. Liquori CL, Berg MJ, Siegel AM, Huang E, Zawistowski JS, Stoffer T, Verlaan D, Balogun F, Hughes L, Leedom TP, Plummer NW, Cannella M, Maglione V, Squitieri F, Johnson EW, Rouleau GA, Ptacek L, Marchuk DA (2003) Mutations in a gene encoding a novel protein containing a phosphotyrosine-binding domain cause type 2 cerebral cavernous malformations. Am J Hum Genet 73:1459–1464

    Article  PubMed  CAS  Google Scholar 

  425. Bergametti F, Denier C, Labauge P, Arnoult M, Boetto S, Clanet M, Coubes P, Echenne B, Ibrahim R, Irthum B, Jacquet G, Lonjon M, Moreau JJ, Neau JP, Parker F, Tremoulet M, Tournier-Lasserve E (2005) Mutations within the programmed cell death 10 gene cause cerebral cavernous malformations. Am J Hum Genet 76:42–51

    Article  PubMed  CAS  Google Scholar 

  426. Yang Y, Wang Y, Li S, Xu Z, Li H, Ma L, Fan J, Bu D, Liu B, Fan Z, Wu G, Jin J, Ding B, Zhu X, Shen Y (2004) Mutations in SCN9A, encoding a sodium channel alpha subunit, in patients with primary erythermalgia. J Med Genet 41:171–174

    Article  PubMed  CAS  Google Scholar 

  427. Brouillard P, Boon LM, Mulliken JB, Enjolras O, Ghassibe M, Warman ML, Tan OT, Olsen BR, Vikkula M (2002) Mutations in a novel factor, glomulin, are responsible for glomuvenous malformations (“glomangiomas”). Am J Hum Genet 70:866–874

    Article  PubMed  CAS  Google Scholar 

  428. Jinnin M, Medici D, Park L, Limaye N, Liu Y, Boscolo E, Bischoff J, Vikkula M, Boye E, Olsen BR (2008) Suppressed NFAT-dependent VEGFR1 expression and constitutive VEGFR2 signaling in infantile hemangioma. Nat Med 14:1236–1246

    Article  PubMed  CAS  Google Scholar 

  429. Gallione CJ, Repetto GM, Legius E, Rustgi AK, Schelley SL, Tejpar S, Mitchell G, Drouin E, Westermann CJ, Marchuk DA (2004) A combined syndrome of juvenile polyposis and hereditary haemorrhagic telangiectasia associated with mutations in MADH4 (SMAD4). Lancet 363:852–859

    Article  PubMed  CAS  Google Scholar 

  430. Ferrell RE, Levinson KL, Esman JH, Kimak MA, Lawrence EC, Barmada MM, Finegold DN (1998) Hereditary lymphedema: evidence for linkage and genetic heterogeneity. Hum Mol Genet 7:2073–2078

    Article  PubMed  CAS  Google Scholar 

  431. Ferrell RE, Baty CJ, Kimak MA, Karlsson JM, Lawrence EC, Franke-Snyder M, Meriney SD, Feingold E, Finegold DN (2010) GJC2 missense mutations cause human lymphedema. Am J Hum Genet 86:943–948

    Article  PubMed  CAS  Google Scholar 

  432. McAllister KA, Grogg KM, Johnson DW, Gallione CJ, Baldwin MA, Jackson CE, Helmbold EA, Markel DS, McKinnon WC, Murrell J et al (1994) Endoglin, a TGF-beta binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat Genet 8:345–351

    Article  PubMed  CAS  Google Scholar 

  433. Johnson DW, Berg JN, Baldwin MA, Gallione CJ, Marondel I, Yoon SJ, Stenzel TT, Speer M, Pericak-Vance MA, Diamond A, Guttmacher AE, Jackson CE, Attisano L, Kucherlapati R, Porteous ME, Marchuk DA (1996) Mutations in the activin receptor-like kinase 1 gene in hereditary haemorrhagic telangiectasia type 2. Nat Genet 13:189–195

    Article  PubMed  CAS  Google Scholar 

  434. Nagamine K, Peterson P, Scott HS, Kudoh J, Minoshima S, Heino M, Krohn KJ, Lalioti MD, Mullis PE, Antonarakis SE, Kawasaki K, Asakawa S, Ito F, Shimizu N (1997) Positional cloning of the APECED gene. Nat Genet 17:393–398

    Article  PubMed  CAS  Google Scholar 

  435. Glocker EO, Hennigs A, Nabavi M, Schaffer AA, Woellner C, Salzer U, Pfeifer D, Veelken H, Warnatz K, Tahami F, Jamal S, Manguiat A, Rezaei N, Amirzargar AA, Plebani A, Hannesschlager N, Gross O, Ruland J, Grimbacher B (2009) A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N Engl J Med 361:1727–1735

    Article  PubMed  CAS  Google Scholar 

  436. Ferwerda B, Ferwerda G, Plantinga TS, Willment JA, van Spriel AB, Venselaar H, Elbers CC, Johnson MD, Cambi A, Huysamen C, Jacobs L, Jansen T, Verheijen K, Masthoff L, Morre SA, Vriend G, Williams DL, Perfect JR, Joosten LA, Wijmenga C, van der Meer JW, Adema GJ, Kullberg BJ, Brown GD, Netea MG (2009) Human dectin-1 deficiency and mucocutaneous fungal infections. N Engl J Med 361:1760–1767

    Article  PubMed  CAS  Google Scholar 

  437. Matthijs G, Schollen E, Pardon E, Veiga-Da-Cunha M, Jaeken J, Cassiman JJ, Van Schaftingen E (1997) Mutations in PMM2, a phosphomannomutase gene on chromosome 16p13, in carbohydrate-deficient glycoprotein type I syndrome (Jaeken syndrome). Nat Genet 16:88–92

    Article  PubMed  CAS  Google Scholar 

  438. Korner C, Knauer R, Stephani U, Marquardt T, Lehle L, von Figura K (1999) Carbohydrate deficient glycoprotein syndrome type IV: deficiency of dolichyl-P-Man:Man(5)GlcNAc(2)-PP-dolichyl mannosyltransferase. EMBO J 18:6816–6822

    Article  PubMed  CAS  Google Scholar 

  439. Kim S, Westphal V, Srikrishna G, Mehta DP, Peterson S, Filiano J, Karnes PS, Patterson MC, Freeze HH (2000) Dolichol phosphate mannose synthase (DPM1) mutations define congenital disorder of glycosylation Ie (CDG-Ie). J Clin Invest 105:191–198

    Article  PubMed  CAS  Google Scholar 

  440. Schenk B, Imbach T, Frank CG, Grubenmann CE, Raymond GV, Hurvitz H, Korn-Lubetzki I, Revel-Vik S, Raas-Rotschild A, Luder AS, Jaeken J, Berger EG, Matthijs G, Hennet T, Aebi M (2001) MPDU1 mutations underlie a novel human congenital disorder of glycosylation, designated type If. J Clin Invest 108:1687–1695

    PubMed  CAS  Google Scholar 

  441. Wu X, Rush JS, Karaoglu D, Krasnewich D, Lubinsky MS, Waechter CJ, Gilmore R, Freeze HH (2003) Deficiency of UDP-GlcNAc:Dolichol Phosphate N-Acetylglucosamine-1 Phosphate Transferase (DPAGT1) causes a novel congenital disorder of Glycosylation Type Ij. Hum Mutat 22:144–150

    Article  PubMed  CAS  Google Scholar 

  442. Tan J, Dunn J, Jaeken J, Schachter H (1996) Mutations in the MGAT2 gene controlling complex N-glycan synthesis cause carbohydrate-deficient glycoprotein syndrome type II, an autosomal recessive disease with defective brain development. Am J Hum Genet 59:810–817

    PubMed  CAS  Google Scholar 

  443. De Praeter CM, Gerwig GJ, Bause E, Nuytinck LK, Vliegenthart JF, Breuer W, Kamerling JP, Espeel MF, Martin JJ, De Paepe AM, Chan NW, Dacremont GA, Van Coster RN (2000) A novel disorder caused by defective biosynthesis of N-linked oligosaccharides due to glucosidase I deficiency. Am J Hum Genet 66:1744–1756

    Article  PubMed  Google Scholar 

  444. Luhn K, Wild MK, Eckhardt M, Gerardy-Schahn R, Vestweber D (2001) The gene defective in leukocyte adhesion deficiency II encodes a putative GDP-fucose transporter. Nat Genet 28:69–72

    PubMed  CAS  Google Scholar 

  445. Lubke T, Marquardt T, Etzioni A, Hartmann E, von Figura K, Korner C (2001) Complementation cloning identifies CDG-IIc, a new type of congenital disorders of glycosylation, as a GDP-fucose transporter deficiency. Nat Genet 28:73–76

    PubMed  CAS  Google Scholar 

  446. Wu X, Steet RA, Bohorov O, Bakker J, Newell J, Krieger M, Spaapen L, Kornfeld S, Freeze HH (2004) Mutation of the COG complex subunit gene COG7 causes a lethal congenital disorder. Nat Med 10:518–523

    Article  PubMed  CAS  Google Scholar 

  447. Foulquier F, Vasile E, Schollen E, Callewaert N, Raemaekers T, Quelhas D, Jaeken J, Mills P, Winchester B, Krieger M, Annaert W, Matthijs G (2006) Conserved oligomeric Golgi complex subunit 1 deficiency reveals a previously uncharacterized congenital disorder of glycosylation type II. Proc Natl Acad Sci U S A 103:3764–3769

    Article  PubMed  CAS  Google Scholar 

  448. Reynders E, Foulquier F, Leao Teles E, Quelhas D, Morelle W, Rabouille C, Annaert W, Matthijs G (2009) Golgi function and dysfunction in the first COG4-deficient CDG type II patient. Hum Mol Genet 18:3244–3256

    Article  PubMed  CAS  Google Scholar 

  449. Schwarz M, Thiel C, Lubbehusen J, Dorland B, de Koning T, von Figura K, Lehle L, Korner C (2004) Deficiency of GDP-Man:GlcNAc2-PP-dolichol mannosyltransferase causes congenital disorder of glycosylation type Ik. Am J Hum Genet 74:472–481

    Article  PubMed  CAS  Google Scholar 

  450. Clark RA, Malech HL, Gallin JI, Nunoi H, Volpp BD, Pearson DW, Nauseef WM, Curnutte JT (1989) Genetic variants of chronic granulomatous disease: prevalence of deficiencies of two cytosolic components of the NADPH oxidase system. N Engl J Med 321:647–652

    Article  PubMed  CAS  Google Scholar 

  451. Nunoi H, Iwata M, Tatsuzawa S, Onoe Y, Shimizu S, Kanegasaki S, Matsuda I (1995) AG dinucleotide insertion in a patient with chronic granulomatous disease lacking cytosolic 67-kD protein. Blood 86:329–333

    PubMed  CAS  Google Scholar 

  452. Dinauer MC, Curnutte JT, Rosen H, Orkin SH (1989) A missense mutation in the neutrophil cytochrome b heavy chain in cytochrome-positive X-linked chronic granulomatous disease. J Clin Invest 84:2012–2016

    Article  PubMed  CAS  Google Scholar 

  453. Minegishi Y, Saito M, Tsuchiya S, Tsuge I, Takada H, Hara T, Kawamura N, Ariga T, Pasic S, Stojkovic O, Metin A, Karasuyama H (2007) Dominant-negative mutations in the DNA-binding domain of STAT3 cause hyper-IgE syndrome. Nature 448:1058–1062

    Article  PubMed  CAS  Google Scholar 

  454. Zhang Q, Davis JC, Lamborn IT, Freeman AF, Jing H, Favreau AJ, Matthews HF, Davis J, Turner ML, Uzel G, Holland SM, Su HC (2009) Combined immunodeficiency associated with DOCK8 mutations. N Engl J Med 361:2046–2055

    Article  PubMed  CAS  Google Scholar 

  455. Chatila TA, Blaeser F, Ho N, Lederman HM, Voulgaropoulos C, Helms C, Bowcock AM (2000) JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. J Clin Invest 106:R75–R81

    Article  PubMed  CAS  Google Scholar 

  456. Onouchi Y, Onoue S, Tamari M, Wakui K, Fukushima Y, Yashiro M, Nakamura Y, Yanagawa H, Kishi F, Ouchi K, Terai M, Hamamoto K, Kudo F, Aotsuka H, Sato Y, Nariai A, Kaburagi Y, Miura M, Saji T, Kawasaki T, Hata A (2004) CD40 ligand gene and Kawasaki disease. Eur J Hum Genet 12:1062–1068

    Article  PubMed  CAS  Google Scholar 

  457. Dana N, Clayton LK, Tennen DG, Pierce MW, Lachmann PJ, Law SA, Arnaout MA (1987) Leukocytes from four patients with complete or partial Leu-CAM deficiency contain the common beta-subunit precursor and beta-subunit messenger RNA. J Clin Invest 79:1010–1015

    Article  PubMed  CAS  Google Scholar 

  458. Svensson L, Howarth K, McDowall A, Patzak I, Evans R, Ussar S, Moser M, Metin A, Fried M, Tomlinson I, Hogg N (2009) Leukocyte adhesion deficiency-III is caused by mutations in KINDLIN3 affecting integrin activation. Nat Med 15:306–312

    Article  PubMed  CAS  Google Scholar 

  459. Villa A, Santagata S, Bozzi F, Giliani S, Frattini A, Imberti L, Gatta LB, Ochs HD, Schwarz K, Notarangelo LD, Vezzoni P, Spanopoulou E (1998) Partial V(D)J recombination activity leads to Omenn syndrome. Cell 93:885–896

    Article  PubMed  CAS  Google Scholar 

  460. Ege M, Ma Y, Manfras B, Kalwak K, Lu H, Lieber MR, Schwarz K, Pannicke U (2005) Omenn syndrome due to ARTEMIS mutations. Blood 105:4179–4186

    Article  PubMed  CAS  Google Scholar 

  461. Valerio D, Dekker BM, Duyvesteyn MG, van der Voorn L, Berkvens TM, van Ormondt H, van der Eb AJ (1986) One adenosine deaminase allele in a patient with severe combined immunodeficiency contains a point mutation abolishing enzyme activity. EMBO J 5:113–119

    PubMed  CAS  Google Scholar 

  462. Minegishi Y, Saito M, Morio T, Watanabe K, Agematsu K, Tsuchiya S, Takada H, Hara T, Kawamura N, Ariga T, Kaneko H, Kondo N, Tsuge I, Yachie A, Sakiyama Y, Iwata T, Bessho F, Ohishi T, Joh K, Imai K, Kogawa K, Shinohara M, Fujieda M, Wakiguchi H, Pasic S, Abinun M, Ochs HD, Renner ED, Jansson A, Belohradsky BH, Metin A, Shimizu N, Mizutani S, Miyawaki T, Nonoyama S, Karasuyama H (2006) Human tyrosine kinase 2 deficiency reveals its requisite roles in multiple cytokine signals involved in innate and acquired immunity. Immunity 25:745–755

    Article  PubMed  CAS  Google Scholar 

  463. Hernandez PA, Gorlin RJ, Lukens JN, Taniuchi S, Bohinjec J, Francois F, Klotman ME, Diaz GA (2003) Mutations in the chemokine receptor gene CXCR4 are associated with WHIM syndrome, a combined immunodeficiency disease. Nat Genet 34:70–74

    Article  PubMed  CAS  Google Scholar 

  464. Derry JM, Ochs HD, Francke U (1994) Isolation of a novel gene mutated in Wiskott-Aldrich syndrome. Cell 78:635–644

    Article  PubMed  CAS  Google Scholar 

  465. Hultin MB, McKay J, Abildgaard U (1988) Antithrombin Oslo: type Ib classification of the first reported antithrombin-deficient family, with a review of hereditary antithrombin variants. Thromb Haemost 59:468–473

    PubMed  CAS  Google Scholar 

  466. Warwicker P, Goodship TH, Donne RL, Pirson Y, Nicholls A, Ward RM, Turnpenny P, Goodship JA (1998) Genetic studies into inherited and sporadic hemolytic uremic syndrome. Kidney Int 53:836–844

    Article  PubMed  CAS  Google Scholar 

  467. Zipfel PF, Edey M, Heinen S, Jozsi M, Richter H, Misselwitz J, Hoppe B, Routledge D, Strain L, Hughes AE, Goodship JA, Licht C, Goodship TH, Skerka C (2007) Deletion of complement factor H-related genes CFHR1 and CFHR3 is associated with atypical hemolytic uremic syndrome. PLoS Genet 3:e41

    Article  PubMed  CAS  Google Scholar 

  468. Bertina RM, Koeleman BP, Koster T, Rosendaal FR, Dirven RJ, de Ronde H, van der Velden PA, Reitsma PH (1994) Mutation in blood coagulation factor V associated with resistance to activated protein C. Nature 369:64–67

    Article  PubMed  CAS  Google Scholar 

  469. Romeo G, Hassan HJ, Staempfli S, Roncuzzi L, Cianetti L, Leonardi A, Vicente V, Mannucci PM, Bertina R, Peschle C et al (1987) Hereditary thrombophilia: identification of nonsense and missense mutations in the protein C gene. Proc Natl Acad Sci U S A 84:2829–2832

    Article  PubMed  CAS  Google Scholar 

  470. Millar DS, Johansen B, Berntorp E, Minford A, Bolton-Maggs P, Wensley R, Kakkar V, Schulman S, Torres A, Bosch N, Cooper DN (2000) Molecular genetic analysis of severe protein C deficiency. Hum Genet 106:646–653

    Article  PubMed  CAS  Google Scholar 

  471. Formstone CJ, Wacey AI, Berg LP, Rahman S, Bevan D, Rowley M, Voke J, Bernardi F, Legnani C, Simioni P, Girolami A, Tuddenham EG, Kakkar VV, Cooper DN (1995) Detection and characterization of seven novel protein S (PROS) gene lesions: evaluation of reverse transcript-polymerase chain reaction as a mutation screening strategy. Blood 86:2632–2641

    PubMed  CAS  Google Scholar 

  472. Schafer AI (1999) Venous thrombosis as a chronic disease. N Engl J Med 340:955–956

    Article  PubMed  CAS  Google Scholar 

  473. Levy GG, Nichols WC, Lian EC, Foroud T, McClintick JN, McGee BM, Yang AY, Siemieniak DR, Stark KR, Gruppo R, Sarode R, Shurin SB, Chandrasekaran V, Stabler SP, Sabio H, Bouhassira EE, Upshaw JD Jr, Ginsburg D, Tsai HM (2001) Mutations in a member of the ADAMTS gene family cause thrombotic thrombocytopenic purpura. Nature 413:488–494

    Article  PubMed  CAS  Google Scholar 

  474. Groden J, Thliveris A, Samowitz W, Carlson M, Gelbert L, Albertsen H, Joslyn G, Stevens J, Spirio L, Robertson M et al (1991) Identification and characterization of the familial adenomatous polyposis coli gene. Cell 66:589–600

    Article  PubMed  CAS  Google Scholar 

  475. Savitsky K, Bar-Shira A, Gilad S, Rotman G, Ziv Y, Vanagaite L, Tagle DA, Smith S, Uziel T, Sfez S, Ashkenazi M, Pecker I, Frydman M, Harnik R, Patanjali SR, Simmons A, Clines GA, Sartiel A, Gatti RA, Chessa L, Sanal O, Lavin MF, Jaspers NG, Taylor AM, Arlett CF, Miki T, Weissman SM, Lovett M, Collins FS, Shiloh Y (1995) A single ataxia telangiectasia gene with a product similar to PI-3 kinase. Science 268:1749–1753

    Article  PubMed  CAS  Google Scholar 

  476. Marsh DJ, Dahia PL, Zheng Z, Liaw D, Parsons R, Gorlin RJ, Eng C (1997) Germline mutations in PTEN are present in Bannayan-Zonana syndrome. Nat Genet 16:333–334

    Article  PubMed  CAS  Google Scholar 

  477. Johnson RL, Rothman AL, Xie J, Goodrich LV, Bare JW, Bonifas JM, Quinn AG, Myers RM, Cox DR, Epstein EH Jr, Scott MP (1996) Human homolog of patched, a candidate gene for the basal cell nevus syndrome. Science 272:1668–1671

    Article  PubMed  CAS  Google Scholar 

  478. Hahn H, Wicking C, Zaphiropoulous PG, Gailani MR, Shanley S, Chidambaram A, Vorechovsky I, Holmberg E, Unden AB, Gillies S, Negus K, Smyth I, Pressman C, Leffell DJ, Gerrard B, Goldstein AM, Dean M, Toftgard R, Chenevix-Trench G, Wainwright B, Bale AE (1996) Mutations of the human homolog of Drosophila patched in the nevoid basal cell carcinoma syndrome. Cell 85:841–851

    Article  PubMed  CAS  Google Scholar 

  479. Smyth I, Narang MA, Evans T, Heimann C, Nakamura Y, Chenevix-Trench G, Pietsch T, Wicking C, Wainwright BJ (1999) Isolation and characterization of human patched 2 (PTCH2), a putative tumour suppressor gene inbasal cell carcinoma and medulloblastoma on chromosome 1p32. Hum Mol Genet 8:291–297

    Article  PubMed  CAS  Google Scholar 

  480. Pastorino L, Ghiorzo P, Nasti S, Battistuzzi L, Cusano R, Marzocchi C, Garre ML, Clementi M, Scarra GB (2009) Identification of a SUFU germline mutation in a family with Gorlin syndrome. Am J Med Genet A 149A:1539–1543

    Article  PubMed  CAS  Google Scholar 

  481. Nickerson ML, Warren MB, Toro JR, Matrosova V, Glenn G, Turner ML, Duray P, Merino M, Choyke P, Pavlovich CP, Sharma N, Walther M, Munroe D, Hill R, Maher E, Greenberg C, Lerman MI, Linehan WM, Zbar B, Schmidt LS (2002) Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the Birt-Hogg-Dube syndrome. Cancer Cell 2:157–164

    Article  PubMed  CAS  Google Scholar 

  482. Willis AE, Lindahl T (1987) DNA ligase I deficiency in Bloom’s syndrome. Nature 325:355–357

    Article  PubMed  CAS  Google Scholar 

  483. Chan JY, Becker FF, German J, Ray JH (1987) Altered DNA ligase I activity in Bloom’s syndrome cells. Nature 325:357–359

    Article  PubMed  CAS  Google Scholar 

  484. Poblete Gutierrez P, Eggermann T, Holler D, Jugert FK, Beermann T, Grussendorf-Conen EI, Zerres K, Merk HF, Frank J (2002) Phenotype diversity in familial cylindromatosis: a frameshift mutation in the tumor suppressor gene CYLD underlies different tumors of skin appendages. J Invest Dermatol 119:527–531

    Article  PubMed  Google Scholar 

  485. Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, Cho-Chung YS, Stratakis CA (2000) Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit in patients with the Carney complex. Nat Genet 26:89–92

    Article  PubMed  CAS  Google Scholar 

  486. Liaw D, Marsh DJ, Li J, Dahia PL, Wang SI, Zheng Z, Bose S, Call KM, Tsou HC, Peacocke M, Eng C, Parsons R (1997) Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat Genet 16:64–67

    Article  PubMed  CAS  Google Scholar 

  487. Ni Y, Zbuk KM, Sadler T, Patocs A, Lobo G, Edelman E, Platzer P, Orloff MS, Waite KA, Eng C (2008) Germline mutations and variants in the succinate dehydrogenase genes in Cowden and Cowden-like syndromes. Am J Hum Genet 83:261–268

    Article  PubMed  CAS  Google Scholar 

  488. Bignell GR, Warren W, Seal S, Takahashi M, Rapley E, Barfoot R, Green H, Brown C, Biggs PJ, Lakhani SR, Jones C, Hansen J, Blair E, Hofmann B, Siebert R, Turner G, Evans DG, Schrander-Stumpel C, Beemer FA, van Den Ouweland A, Halley D, Delpech B, Cleveland MG, Leigh I, Leisti J, Rasmussen S (2000) Identification of the familial cylindromatosis tumour-suppressor gene. Nat Genet 25:160–165

    Article  PubMed  CAS  Google Scholar 

  489. Heiss NS, Knight SW, Vulliamy TJ, Klauck SM, Wiemann S, Mason PJ, Poustka A, Dokal I (1998) X-linked dyskeratosis congenita is caused by mutations in a highly conserved gene with putative nucleolar functions. Nat Genet 19:32–38

    Article  PubMed  CAS  Google Scholar 

  490. Vulliamy T, Marrone A, Goldman F, Dearlove A, Bessler M, Mason PJ, Dokal I (2001) The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita. Nature 413:432–435

    Article  PubMed  CAS  Google Scholar 

  491. Armanios M, Chen JL, Chang YP, Brodsky RA, Hawkins A, Griffin CA, Eshleman JR, Cohen AR, Chakravarti A, Hamosh A, Greider CW (2005) Haploinsufficiency of telomerase reverse transcriptase leads to anticipation in autosomal dominant dyskeratosis congenita. Proc Natl Acad Sci U S A 102:15960–15964

    Article  PubMed  CAS  Google Scholar 

  492. Walne AJ, Vulliamy T, Beswick R, Kirwan M, Dokal I (2008) TINF2 mutations result in very short telomeres: analysis of a large cohort of patients with dyskeratosis congenita and related bone marrow failure syndromes. Blood 112:3594–3600

    Article  PubMed  CAS  Google Scholar 

  493. Walne AJ, Vulliamy T, Marrone A, Beswick R, Kirwan M, Masunari Y, Al-Qurashi FH, Aljurf M, Dokal I (2007) Genetic heterogeneity in autosomal recessive dyskeratosis congenita with one subtype due to mutations in the telomerase-associated protein NOP10. Hum Mol Genet 16:1619–1629

    Article  PubMed  CAS  Google Scholar 

  494. Vulliamy T, Beswick R, Kirwan M, Marrone A, Digweed M, Walne A, Dokal I (2008) Mutations in the telomerase component NHP2 cause the premature ageing syndrome dyskeratosis congenita. Proc Natl Acad Sci U S A 105:8073–8078

    Article  PubMed  CAS  Google Scholar 

  495. Ramoz N, Rueda LA, Bouadjar B, Montoya LS, Orth G, Favre M (2002) Mutations in two adjacent novel genes are associated with epidermodysplasia verruciformis. Nat Genet 32:579–581

    Article  PubMed  CAS  Google Scholar 

  496. Tomlinson IP, Alam NA, Rowan AJ, Barclay E, Jaeger EE, Kelsell D, Leigh I, Gorman P, Lamlum H, Rahman S, Roylance RR, Olpin S, Bevan S, Barker K, Hearle N, Houlston RS, Kiuru M, Lehtonen R, Karhu A, Vilkki S, Laiho P, Eklund C, Vierimaa O, Aittomaki K, Hietala M, Sistonen P, Paetau A, Salovaara R, Herva R, Launonen V, Aaltonen LA (2002) Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat Genet 30:406–410

    Article  PubMed  CAS  Google Scholar 

  497. Klopstock T, Naumann M, Schalke B, Bischof F, Seibel P, Kottlors M, Eckert P, Reiners K, Toyka KV, Reichmann H (1994) Multiple symmetric lipomatosis: abnormalities in complex IV and multiple deletions in mitochondrial DNA. Neurology 44:862–866

    Article  PubMed  CAS  Google Scholar 

  498. Longley BJ, Tyrrell L, Lu SZ, Ma YS, Langley K, Ding TG, Duffy T, Jacobs P, Tang LH, Modlin I (1996) Somatic c-KIT activating mutation in urticaria pigmentosa and aggressive mastocytosis: establishment of clonality in a human mast cell neoplasm. Nat Genet 12:312–314

    Article  PubMed  CAS  Google Scholar 

  499. Kamb A, Shattuck-Eidens D, Eeles R, Liu Q, Gruis NA, Ding W, Hussey C, Tran T, Miki Y, Weaver-Feldhaus J et al (1994) Analysis of the p16 gene (CDKN2) as a candidate for the chromosome 9p melanoma susceptibility locus. Nat Genet 8:23–26

    Article  PubMed  CAS  Google Scholar 

  500. Wolfel T, Hauer M, Schneider J, Serrano M, Wolfel C, Klehmann-Hieb E, De Plaen E, Hankeln T, Meyer zum Buschenfelde KH, Beach D (1995) A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. Science 269:1281–1284

    Article  PubMed  CAS  Google Scholar 

  501. Valverde P, Healy E, Sikkink S, Haldane F, Thody AJ, Carothers A, Jackson IJ, Rees JL (1996) The Asp84Glu variant of the melanocortin 1 receptor (MC1R) is associated with melanoma. Hum Mol Genet 5:1663–1666

    Article  PubMed  CAS  Google Scholar 

  502. Whelan AJ, Bartsch D, Goodfellow PJ (1995) Brief report: a familial syndrome of pancreatic cancer and melanoma with a mutation in the CDKN2 tumor-suppressor gene. N Engl J Med 333:975–977

    Article  PubMed  CAS  Google Scholar 

  503. Sobreira NL, Cirulli ET, Avramopoulos D, Wohler E, Oswald GL, Stevens EL, Ge D, Shianna KV, Smith JP, Maia JM, Gumbs CE, Pevsner J, Thomas G, Valle D, Hoover-Fong JE, Goldstein DB (2010) Whole-genome sequencing of a single proband together with linkage analysis identifies a Mendelian disease gene. PLoS Genet 6:e1000991

    Article  PubMed  CAS  Google Scholar 

  504. Hamilton SR, Liu B, Parsons RE, Papadopoulos N, Jen J, Powell SM, Krush AJ, Berk T, Cohen Z, Tetu B et al (1995) The molecular basis of Turcot’s syndrome. N Engl J Med 332:839–847

    Article  PubMed  CAS  Google Scholar 

  505. Ricciardone MD, Ozcelik T, Cevher B, Ozdag H, Tuncer M, Gurgey A, Uzunalimoglu O, Cetinkaya H, Tanyeli A, Erken E, Ozturk M (1999) Human MLH1 deficiency predisposes to hematological malignancy and neurofibromatosis type 1. Cancer Res 59:290–293

    PubMed  CAS  Google Scholar 

  506. Whiteside D, McLeod R, Graham G, Steckley JL, Booth K, Somerville MJ, Andrew SE (2002) A homozygous germ-line mutation in the human MSH2 gene predisposes to hematological malignancy and multiple cafe-au-lait spots. Cancer Res 62:359–362

    PubMed  CAS  Google Scholar 

  507. Menko FH, Kaspers GL, Meijer GA, Claes K, van Hagen JM, Gille JJ (2004) A homozygous MSH6 mutation in a child with cafe-au-lait spots, oligodendroglioma and rectal cancer. Fam Cancer 3:123–127

    Article  PubMed  CAS  Google Scholar 

  508. Kruse R, Lamberti C, Wang Y, Ruelfs C, Bruns A, Esche C, Lehmann P, Ruzicka T, Rutten A, Friedl W, Propping P (1996) Is the mismatch repair deficient type of Muir-Torre syndrome confined to mutations in the hMSH2 gene? Hum Genet 98:747–750

    Article  PubMed  CAS  Google Scholar 

  509. Chandrasekharappa SC, Guru SC, Manickam P, Olufemi SE, Collins FS, Emmert-Buck MR, Debelenko LV, Zhuang Z, Lubensky IA, Liotta LA, Crabtree JS, Wang Y, Roe BA, Weisemann J, Boguski MS, Agarwal SK, Kester MB, Kim YS, Heppner C, Dong Q, Spiegel AM, Burns AL, Marx SJ (1997) Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science 276:404–407

    Article  PubMed  CAS  Google Scholar 

  510. Mulligan LM, Kwok JB, Healey CS, Elsdon MJ, Eng C, Gardner E, Love DR, Mole SE, Moore JK, Papi L et al (1993) Germ-line mutations of the RET proto-oncogene in multiple endocrine neoplasia type 2A. Nature 363:458–460

    Article  PubMed  CAS  Google Scholar 

  511. Hofstra RM, Landsvater RM, Ceccherini I, Stulp RP, Stelwagen T, Luo Y, Pasini B, Hoppener JW, van Amstel HK, Romeo G et al (1994) A mutation in the RET proto-oncogene associated with multiple endocrine neoplasia type 2B and sporadic medullary thyroid carcinoma. Nature 367:375–376

    Article  PubMed  CAS  Google Scholar 

  512. Pellegata NS, Quintanilla-Martinez L, Siggelkow H, Samson E, Bink K, Hofler H, Fend F, Graw J, Atkinson MJ (2006) Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proc Natl Acad Sci U S A 103:15558–15563

    Article  PubMed  CAS  Google Scholar 

  513. Goudie DR, D’Alessandro M, Merriman B, Lee H, Szeverenyi I, Avery S, O’Connor BD, Nelson SF, Coats SE, Stewart A, Christie L, Pichert G, Friedel J, Hayes I, Burrows N, Whittaker S, Gerdes AM, Broesby-Olsen S, Ferguson-Smith MA, Verma C, Lunny DP, Reversade B, Lane EB (2011) Multiple self-healing squamous epithelioma is caused by a disease-specific spectrum of mutations in TGFBR1. Nat Genet 43:365–369

    Article  PubMed  CAS  Google Scholar 

  514. Rouleau GA, Merel P, Lutchman M, Sanson M, Zucman J, Marineau C, Hoang-Xuan K, Demczuk S, Desmaze C, Plougastel B et al (1993) Alteration in a new gene encoding a putative membrane-organizing protein causes neuro-fibromatosis type 2. Nature 363:515–521

    Article  PubMed  CAS  Google Scholar 

  515. Baralle D, Mattocks C, Kalidas K, Elmslie F, Whittaker J, Lees M, Ragge N, Patton MA, Winter RM, ffrench-Constant C (2003) Different mutations in the NF1 gene are associated with Neurofibromatosis-Noonan syndrome (NFNS). Am J Med Genet A 119A:1–8

    Article  PubMed  Google Scholar 

  516. Jenne DE, Reimann H, Nezu J, Friedel W, Loff S, Jeschke R, Muller O, Back W, Zimmer M (1998) Peutz-Jeghers syndrome is caused by mutations in a novel serine threonine kinase. Nat Genet 18:38–43

    Article  PubMed  CAS  Google Scholar 

  517. Zhou XP, Marsh DJ, Hampel H, Mulliken JB, Gimm O, Eng C (2000) Germline and germline mosaic PTEN mutations associated with a Proteus-like syndrome of hemihypertrophy, lower limb asymmetry, arteriovenous malformations and lipomatosis. Hum Mol Genet 9:765–768

    Article  PubMed  CAS  Google Scholar 

  518. Kitao S, Shimamoto A, Goto M, Miller RW, Smithson WA, Lindor NM, Furuichi Y (1999) Mutations in RECQL4 cause a subset of cases of Rothmund-Thomson syndrome. Nat Genet 22:82–84

    Article  PubMed  CAS  Google Scholar 

  519. Hu G, Onder M, Gill M, Aksakal B, Oztas M, Gurer MA, Celebi JT (2003) A novel missense mutation in CYLD in a family with Brooke-Spiegler syndrome. J Invest Dermatol 121:732–734

    Article  PubMed  CAS  Google Scholar 

  520. van Slegtenhorst M, de Hoogt R, Hermans C, Nellist M, Janssen B, Verhoef S, Lindhout D, van den Ouweland A, Halley D, Young J, Burley M, Jeremiah S, Woodward K, Nahmias J, Fox M, Ekong R, Osborne J, Wolfe J, Povey S, Snell RG, Cheadle JP, Jones AC, Tachataki M, Ravine D, Sampson JR, Reeve MP, Richardson P, Wilmer F, Munro C, Hawkins TL, Sepp T, Ali JB, Ward S, Green AJ, Yates JR, Kwiatkowska J, Henske EP, Short MP, Haines JH, Jozwiak S, Kwiatkowski DJ (1997) Identification of the tuberous sclerosis gene TSC1 on chromosome 9q34. Science 277:805–808

    Article  PubMed  Google Scholar 

  521. Kumar A, Wolpert C, Kandt RS, Segal J, Pufky J, Roses AD, Pericak-Vance MA, Gilbert JR (1995) A de novo frame-shift mutation in the tuberin gene. Hum Mol Genet 4:1471–1472

    Article  PubMed  CAS  Google Scholar 

  522. Vikkula M, Boon LM, Carraway KL 3rd, Calvert JT, Diamonti AJ, Goumnerov B, Pasyk KA, Marchuk DA, Warman ML, Cantley LC, Mulliken JB, Olsen BR (1996) Vascular dysmorphogenesis caused by an activating mutation in the receptor tyrosine kinase TIE2. Cell 87:1181–1190

    Article  PubMed  CAS  Google Scholar 

  523. Upadhyaya M, Shen M, Cherryson A, Farnham J, Maynard J, Huson SM, Harper PS (1992) Analysis of mutations at the neurofibromatosis 1 (NF1) locus. Hum Mol Genet 1:735–740

    PubMed  CAS  Google Scholar 

  524. Tanaka K, Miura N, Satokata I, Miyamoto I, Yoshida MC, Satoh Y, Kondo S, Yasui A, Okayama H, Okada Y (1990) Analysis of a human DNA excision repair gene involved in group A xeroderma pigmentosum and containing a zinc-finger domain. Nature 348:73–76

    Article  PubMed  CAS  Google Scholar 

  525. Weeda G, van Ham RC, Vermeulen W, Bootsma D, van der Eb AJ, Hoeijmakers JH (1990) A presumed DNA helicase encoded by ERCC-3 is involved in the human repair disorders xeroderma pigmentosum and Cockayne’s syndrome. Cell 62:777–791

    Article  PubMed  CAS  Google Scholar 

  526. Li L, Bales ES, Peterson CA, Legerski RJ (1993) Characterization of molecular defects in xeroderma pigmentosum group C. Nat Genet 5:413–417

    Article  PubMed  CAS  Google Scholar 

  527. Flejter WL, McDaniel LD, Johns D, Friedberg EC, Schultz RA (1992) Correction of xeroderma pigmentosum complementation group D mutant cell phenotypes by chromosome and gene transfer: involvement of the human ERCC2 DNA repair gene. Proc Natl Acad Sci U S A 89:261–265

    Article  PubMed  CAS  Google Scholar 

  528. Nichols AF, Ong P, Linn S (1996) Mutations specific to the xeroderma pigmentosum group E Ddb- phenotype. J Biol Chem 271:24317–24320

    Article  PubMed  CAS  Google Scholar 

  529. Sijbers AM, de Laat WL, Ariza RR, Biggerstaff M, Wei YF, Moggs JG, Carter KC, Shell BK, Evans E, de Jong MC, Rademakers S, de Rooij J, Jaspers NG, Hoeijmakers JH, Wood RD (1996) Xeroderma pigmentosum group F caused by a defect in a structure-specific DNA repair endonuclease. Cell 86:811–822

    Article  PubMed  CAS  Google Scholar 

  530. Lalle P, Nouspikel T, Constantinou A, Thorel F, Clarkson SG (2002) The founding members of xeroderma pigmentosum group G produce XPG protein with severely impaired endonuclease activity. J Invest Dermatol 118:344–351

    Article  PubMed  CAS  Google Scholar 

  531. Masutani C, Kusumoto R, Yamada A, Dohmae N, Yokoi M, Yuasa M, Araki M, Iwai S, Takio K, Hanaoka F (1999) The XPV (xeroderma pigmentosum variant) gene encodes human DNA polymerase eta. Nature 399:700–704

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Vesarat Wessagowit .

Editor information

Editors and Affiliations

Appendix: Genodermatoses with Currently Known Gene Defect

Appendix: Genodermatoses with Currently Known Gene Defect

  1. 1.

    Genodermatoses with keratin abnormalities and keratinisation disorders

    Diseases

    Gene

    Mode of inheritance

    MIM

    Adermatoglyphia

    SMARCAD1

    AD

    136000

    Arthrogryposis, renal dysfunction and cholestasis 1 (54)

    VPS33B

    AR

    208085

    Benign chronic pemphigus (55)

    ATP2C1

    AD

    169600

    Cardio-facio-cutaneous syndrome (56, 57)

    KRAS, BRAF, MEK1, MEK2

    AD

    115150

    Cardiomyopathy, dilated, with woolly hair and keratoderma (58)

    DSP

    AR

    605676

    Cerebral dysgenesis, neuropathy, ichthyosis and palmoplantar keratoderma syndrome (59)

    SNAP29

    AR

    609528

    Chanarin–Dorfman syndrome (60)

    CGI58

    AR

    275630

    Chondrodysplasia punctata 2, X-linked dominant (61)

    EBP

    XD

    302960

    Congenital disorder of glycosylation, type Im (62)

    TMEM15

    AR

    610768

    Congenital hemidysplasia with ichthyosiform erythroderma and limb defects (63)

    NSDHL

    XD

    308050

    Darier–White disease (64)

    ATP2A2

    AD

    124200

    Deafness, congenital, with keratopachydermia and constriction of fingers and toes (65)

    GJB2

    AD

    124500

    Deafness, non-syndromic sensorineural, mitochondrial (17)

    MTTS1

    mt

    500008

    Dermatopathia pigmentosa reticularis (66)

    KRT14

    AD

    125595

    Dowling–Degos disease (67)

    KRT5

    AD

    179850

    Epidermolytic hyperkeratosis (68)

    KRT1

    AD

    113800

    KRT10

    AD>>AR

    Erythrokeratodermia variabilis et progressiva (69, 70)

    GJB3, GJB4

    AD

    133200

    Haim–Munk syndrome (71)

    CTSC

    AR

    245010

    Ichthyosis hystrix, Curth–Macklin type (10)

    KRT1

    AD

    146590

    Ichthyosis with confetti (72)

    KRT10, LOH in chr17q

    AD

    609165

    Ichthyosis, cyclic, with epidermolytic hyperkeratosis (73)

    KRT1, KRT10

    AD

    607602

    Ichthyosis, congenital, autosomal recessive, NIPAL4-related (74)

    ARCI

    AR

    612281

    Ichthyosis congenital, Harlequin foetus type (75)

    ABCA12

    AR

    242500

    Ichthyosis, bullous type (76)

    KRT2

    AD

    146800

    Ichthyosis vulgaris (77)

    FLG

    AD

    146700

    Ichthyosis prematurity syndrome (78)

    FATP4

    AR

    608649

    Ichthyosis, hystrix-like, with deafness (79)

    GJB2

    AD

    602540

    Ichthyosis, lamellar, 1 (80)

    TGM1

    AR

    242300

    Ichthyosis, lamellar, 2 (81)

    ABCA12

    AR

    601277

    Ichthyosis, lamellar, 3 (82)

    CYP4F22

    AR

    604777

    Ichthyosis, leucocyte vacuoles, alopecia and sclerosing cholangitis (83)

    CLDN1

    AR

    607626

    Ichthyosiform erythroderma, congenital, non-bullous 1 (84, 85)

    TGM1, ALOXE3, ALOX12B

    AR

    242100

    Ichthyosis with hypotrichosis, autosomal recessive (86)

    ST14

    AR

    610765

    Keratitis–Ichthyosis–Deafness syndrome, autosomal dominant (87)

    GJB2

    AD

    148210

    Keratosis follicularis spinulosa decalvans, X-linked (88)

    MBTPS2

    XR

    308800

    Keratosis palmoplantaris striata 1 (89)

    DSG1

    AD

    148700

    Keratosis palmoplantaris striata 2 (90)

    DSP

    AD

    612908

    Keratosis palmoplantaris striata 3 (91)

    KRT1

    AD

    607654

    Knuckle pads, leuconychia and sensorineural deafness (92)

    GJB2

    AD

    149200

    Mal de Meleda (93)

    SLURP1

    AR

    248300

    Multiple sulphatase deficiency (94, 95)

    SUMF1

    AR

    272200

    Naegeli syndrome (66)

    KRT14

    AD

    161000

    Noonan syndrome 1 (96)

    PTPN11

    AD

    163950

    Noonan syndrome 3 (97)

    KRAS

    AD

    609942

    Noonan syndrome 4 (98, 99)

    SOS1

    AD

    610733

    Noonan syndrome 5 (100)

    RAF1

    AD

    611553

    Noonan syndrome 6 (101)

    NRAS

    AD

    613224

    Noonan syndrome 7 (102)

    BRAF

    AD

    613706

    Noonan syndrome-like disorder with or without juvenile myelomonocytic leukaemia (103)

    CBL

    AD

    613563

    Noonan syndrome-like disorder with loose anagen hair (104)

    SHOC2

    AD

    607721

    Naxos disease (105)

    JUP

    AR

    601214

    Netherton syndrome (106)

    SPINK5

    AR

    256500

    Oculo-dento-digital dysplasia (107)

    GJA1

    AD

    164200

    Pachyonychia congenita, type 1 (108, 109)

    KRT6A, KRT16

    AD

    167200

    Pachyonychia congenita, type 2 (110, 111)

    KRT6B, KRT17

    AD

    167210

    Palmoplantar keratoderma, epidermolytic (112, 113)

    KRT1, KRT9

    AD

    144200

    Palmoplantar keratoderma, non-epidermolytic (114)

    KRT1

    AD

    600962

    Palmoplantar keratoderma, non-epidermolytic, focal (115)

    KRT16

    AD

    613000

    Palmoplantar hyperkeratosis with squamous cell carcinoma of skin and 46,XX sex reversal (116)

    RSPO1

    AR

    610644

    Papillon–Lefèvre syndrome (117)

    CTSC

    AR

    245000

    Porokeratosis, disseminated superficial actinic, 1 (118)

    SART3

    AD

    175900

    Peeling skin syndrome (119)

    CDSN

    AR

    270300

    Peeling skin syndrome, acral type (120)

    TGM5

    AR

    609796

    Refsum disease, adult, 1 (121)

    PHYH

    AR

    266500

    Pseudofolliculitis barbae

    KRT75

    AD

    612318

    Refsum disease, adult, 2 (122)

    PEX7

    AR

    266500

    Rhizomelic chondrodysplasia punctata, type 1 (123125)

    PEX7

    AR

    215100

    Seborrhoea-like dermatitis with psoriasiform elements (126)

    ZNF750

    AD

    610227

    Steatocystoma multiplex (127)

    KRT17

    AD

    184500

    Tyrosinaemia, type II (128)

    TAT

    AR

    276600

    Vohwinkel syndrome, variant form (129)

    LCR

    AD

    604117

    White sponge naevus (130)

    KRT4, KRT13

    AD

    193900

  2. 2.

    Genodermatoses with skin fragility

    Diseases

    Gene

    Mode of inheritance

    MIM

    Ectodermal dysplasia, skin fragility syndrome (131)

    PKP1

    AR

    604536

    Epidermolysis bullosa simplex, Dowling–Meara type (132, 133)

    KRT5, KRT14

    AD

    131760

    Epidermolysis bullosa simplex, generalised (134, 135)

    KRT5, KRT14

    AD

    131900

    Epidermolysis bullosa simplex, localised (136, 137)

    KRT5, KRT14

    AD

    131800

    Epidermolysis bullosa simplex with mottled pigmentation (138)

    KRT5

    AD

    131960

    Epidermolysis bullosa simplex with migratory circinate erythema (139)

    KRT5

    AD

    609352

    Epidermolysis bullosa simplex, autosomal recessive (140)

    KRT5, KRT14

    AR

    601001

    Epidermolysis bullosa simplex, Ogna type (141)

    PLEC1

    AD

    131950

    Epidermolysis bullosa simplex with muscular dystrophy (142)

    PLEC1

    AR

    226670

    Epidermolysis bullosa simplex with pyloric atresia (143)

    PLEC1

    AR

    612138

    Epidermolysis bullosa, junctional, Herlitz type (144146)

    LAMA3, LAMB3, LAMC2

    AR

    226700

    Epidermolysis bullosa, junctional, non-Herlitz type (147150)

    COL17A1, LAMA3, LAMB3, LAMC2, ITGB4

    AR

    226650

    Epidermolysis bullosa junctionalis with pyloric atresia (151, 152)

    ITGA6, ITGB4

    AR

    226730

    Epidermolysis bullosa dystrophica, autosomal dominant (153)

    COL7A1

    AD

    131750

    Epidermolysis bullosa dystrophica, pretibial (154)

    COL7A1

    AD

    131850

    Epidermolysis bullosa dystrophica, pruriginosa (11)

    COL7A1

    AD

    604129

    Epidermolysis bullosa simplex superficialis (155)

    COL7A1

    AD

    607600

    Epidermolysis bullosa with congenital localised absence of skin and deformity of nails (156)

    COL7A1

    AD

    132000

    Epidermolysis bullosa dystrophica, autosomal recessive (157)

    COL7A1

    AR

    226600

    Epidermolysis bullosa, lethal acantholytic (8)

    DSP

    AR

    609638

    Hypotrichosis and recurrent skin vesicles (158)

    DSC3

    AR

    613102

    Kindler syndrome (159)

    FERMT1

    AR

    173650

    Laryngo-oculo-cutaneous syndrome (160)

    LAMA3

    AR

    245660

    Nephropathy with pretibial epidermolysis bullosa and deafness (161)

    CD151

    AR

    609057

    Skin fragility-woolly hair syndrome (162)

    DSP

    AR

    607655

    Transient bullous dermolysis of the newborn (163)

    COL7A1

    AD>>AR

    131705

    Epidermolysis bullosa, lethal congenital

    JUP

    AR

     
  3. 3.

    Connective tissue genodermatoses

    Diseases

    Gene

    Mode of inheritance

    MIM

    Amyloidosis, familial visceral (13, 164, 165)

    APOA1, FGA, LYZ

    AD

    105200

    Amyloidosis, Finnish type (166)

    GSN

    AD

    105120

    Amyloidosis, hereditary, transthyretin-related (167)

    TTR

    AD

    105210

    Amyloidosis, primary cutaneous (168)

    OSMR

    AD

    105250

    Arterial tortuosity syndrome (169)

    GLUT10

    AR

    208050

    Buschke–Ollendorff syndrome (170)

    LEMD3

    AD

    166700

    Costello syndrome (171)

    HRAS

    AD

    218040

    Cutis laxa, autosomal dominant (172, 173)

    ELN, FBLN5

    AD

    123700

    Cutis laxa, autosomal recessive, type I (174, 175)

    FBLN4, FBLN5

    AR

    219100

    Cutis laxa, autosomal recessive, type IIA (176)

    ATP6V0A2

    AR

    219200

    Cutis laxa, autosomal recessive, type IIB (177)

    PYCR1

    AR

    612940

    Ehlers–Danlos syndrome, type I (178180)

    COL1A1, COL5A1, COL5A2

    AD

    130000

    Ehlers–Danlos syndrome, type II (181, 182)

    COL5A1, COL5A2

    AD

    130010

    Ehlers–Danlos syndrome, type III (183, 184)

    TNXB, COL3A1

    AD

    130020

    Ehlers–Danlos syndrome, type IV, autosomal dominant (185)

    COL3A1

    AD

    130050

    Ehlers–Danlos syndrome, type VI (186)

    PLOD1

    AR

    225400

    Ehlers–Danlos syndrome, type VII, autosomal dominant (187, 188)

    COL1A1, COL1A2

    AD

    130060

    Ehlers–Danlos syndrome, type VII, autosomal recessive (189)

    ADAMTS2

    AR

    225410

    Ehlers–Danlos syndrome, autosomal recessive, cardiac valvular form (190)

    COL1A2

    AR

    225320

    Ehlers–Danlos syndrome, musculocontractural type (191)

    CHST14

    AR

    601776

    Ehlers–Danlos syndrome, progeroid form (192, 193)

    B4GALT7

    AR

    130070

    Ehlers–Danlos-like syndrome due to tenascin-X deficiency (194, 195)

    TNXB

    AR

    606408

    Fibromatosis, juvenile hyaline (196, 197)

    CMG2

    AR

    228600

    Focal dermal hypoplasia (198)

    PORCN

    XD

    305600

    Heterotopia, periventricular, Ehlers–Danlos variant (199)

    FLNA

    XD

    300537

    Hyalinosis, infantile systemic (196, 197)

    CMG2

    AR

    236490

    Lipoid proteinosis of Urbach and Wiethe (200)

    ECM1

    AR

    247100

    Loeys–Dietz syndrome, type 1A (201)

    TGFBR1

    AD

    609192

    Loeys–Dietz syndrome, type 1B (201)

    TGFBR2

    AD

    610168

    Loeys–Dietz syndrome, type 2A (202)

    TGFBR1

    AD

    608967

    Loeys–Dietz syndrome, type 2B (203)

    TGFBR2

    AD

    610380

    Marfan syndrome (204)

    FBN1

    AD

    154700

    Microphthalmia, syndromic 7 (205)

    HCCS

    XD

    309801

    Multiple pterygium syndrome, lethal type (206208)

    CHRNG, CHRNA1, CHRND

    AR

    253290

    Multiple pterygium syndrome, Escobar variant (206, 208)

    CHRNG

    AD

    265000

    Neonatal cutis laxa with marfanoid phenotype (209)

    LAMB1

    AD

    150240

    Nevo syndrome (210)

    PLOD1

    AR

    601451

    Occipital horn syndrome (211)

    ATP7A

    XR

    304150

    Popliteal pterygium syndrome (212)

    IRF6

    AR

    119500

    Pseudoxanthoma elasticum (213215)

    ABCC6

    AR

    264800

    Pseudoxanthoma elasticum-like disorder with multiple coagulation factor deficiency (216)

    GGCX

    AR

    610842

    Restrictive dermopathy, lethal (217)

    LMNA

    AD

    275210

    ZMPSTE24

    AR

    Shprintzen–Goldberg craniostosis syndrome (218)

    FBN1

    AD

    182212

    Spondylocheirodysplasia, Ehlers–Danlos-like (219)

    SLC39A13

    AR

    612350

    Stiff skin syndrome (220)

    FBN1

    AD

    184900

    Torg–Winchester syndrome (221)

    MMP2

    AR

    259600

    Ullrich congenital muscular dystrophy (222)

    COL6A1, COL6A2, COL6A3

    AD, AR

    254090

    van der Woude syndrome (212)

    IRF6

    AD

    119300

    Weill–Marchesani syndrome, autosomal dominant (223)

    FBN1

    AD

    608328

    Weill–Marchesani syndrome, autosomal recessive (224)

    ADAMTS10

    AR

    277600

    Weill–Marchesani-like syndrome (225)

    FBN1, ADAMTS10

    AR

    613195

    Wrinkly skin syndrome (176)

    ATP6V0A2

    AR

    278250

  4. 4.

    Ectodermal dysplasias

    Diseases

    Gene

    Mode of inheritance

    MIM

    ADULT syndrome (226)

    TP63

    AD

    103285

    Ankyloblepharon-ectodermal defects-cleft lip/palate (227)

    TP63

    AD

    106260

    Cleft lip/palate-ectodermal dysplasia syndrome (228)

    PVRL1

    AR

    225060

    EEM syndrome (229)

    CDH3

    AR

    225280

    Ectodermal dysplasia, hidrotic, autosomal dominant (230)

    GJB6

    AD

    129500

    Ectodermal dysplasia, hypohidrotic, autosomal dominant (231, 232)

    EDAR, EDARADD

    AD

    129490

    Ectodermal dysplasia, hypohidrotic, autosomal recessive (231, 233)

    EDAR, EDARADD

    AR

    224900

    Ectodermal dysplasia, hypohidrotic, X-linked (234)

    EDA

    XR

    305100

    Ectodermal dysplasia, hypohidrotic, with immune deficiency (235)

    IKBKG

    XR

    300291

    Ectodermal dysplasia, anhidrotic, with T-cell immunodeficiency, autosomal dominant (236)

    NFKBIA

    AD

    612132

    Ectodermal dysplasia, “pure” hair-nail type (237)

    KRTHB5

    AR

    602032

    Ectrodactyly, ectodermal dysplasias, and cleft lip/palate syndrome 3 (238)

    TP63

    AD

    604292

    Ellis–van Creveld syndrome (239)

    EVC, EVC2

    AR

    225500

    Kabuki syndrome (240)

    MLL2

    Sporadic>>AD

    147920

    Limb-mammary syndrome (241)

    TP63

    AD

    603543

    Lymphoedema-distichiasis syndrome (with yellow nails)(242)

    FOXC2

    AD

    153400

    Odonto-onycho-dermal dysplasia (243)

    WNT10A

    AR

    257980

    Orofaciodigital syndrome I (244)

    CXORF5

    XD

    311200

    Rapp–Hodgkin syndrome (245)

    TP63

    AD

    129400

    Split-hand/foot malformation 4 (246)

    TP63

    AD

    605289

    Split-hand/foot malformation 6 (247)

    WNT10B

    AR

    225300

    Tricho-dento-osseous syndrome (248)

    DLX3

    AD

    190320

    Tricho-rhino-phalangeal syndrome, type I (249)

    TRPS1

    AD

    190350

    Tricho-rhino-phalangeal syndrome, type II (250, 251)

    TRPS1 & EXT1

    AD, contiguous gene syndrome

    150230

    Tricho-rhino-phalangeal syndrome, type III (252)

    TRPS1

    AD

    190351

    Witkop syndrome (253)

    MSX1

    AD

    189500

  5. 5.

    Hereditary disorders of hair and nails

    Diseases

    Gene

    Mode of inheritance

    MIM

    Arginosuccinic aciduria (254)

    ASL

    AR

    207900

    Alopecia universalis congenital (255)

    HR

    AR

    203655

    Anonychia congenita (256)

    RSPO4

    AR

    206800

    Atrichia with popular lesions (257)

    HR

    AR

    209500

    Björnstad syndrome (258)

    BCS1L

    AR

    262000

    Cartilage-hair hypoplasia (259)

    RMRP

    AR

    250250

    Giant axonal neuropathy 1 (260)

    GAN

    AR

    256850

    Hypotrichosis, congenital, with juvenile macular dystrophy (261)

    CDH3

    AR

    601553

    Hypotrichosis, localised, autosomal recessive 1 (262)

    DSG1

    AR

    607903

    Hypotrichosis, localised, autosomal recessive 2 (263)

    LIPH

    AR

    604379

    Hypotrichosis, localised, autosomal recessive 3 (264)

    P2RY5

    AR

    611452

    Hypotrichosis-lymphoedema-telangiectasia syndrome (265)

    SOX18

    AD, AR

    607823

    Hypotrichosis simplex of scalp (266)

    CDSN

    AD

    146520

    Hypotrichosis simplex, hereditary (267)

    APCDD1

    AD, AR

    605389

    Laron syndrome (268)

    GHR

    AR

    262500

    Leigh syndrome, with complex I deficiency (269283)

    MTND2, MTND3, MTND5, MTND6

    mt

    256000

    NDUFV1, NDUFS1, NDUFS3, NDUFS4, NDUFS7, NDUFS8, NDUFA2, C8ORF38, C20ORF7, NDUFAF2, FOXRED1

    AR

    Leigh syndrome, with complex II deficiency (284)

    SDHA

    AR

    Leigh syndrome, with complex III deficiency (285)

    BCS1L

    AR

    Leigh syndrome, with complex IV deficiency (286)

    MTCO3

    mt

    COX10, COX15, SCO2, SURF1, TACO1

    AR

    Leigh syndrome, with complex V deficiency (287)

    MTATP6

    mt

    Leigh syndrome, with mutations in genes encoding mitochondrial tRNA proteins (288292)

    MTTV, MTTK, MTTW, MTTL1

    mt

    Leigh syndrome, with mutations in components of pyruvate dehydrogenase complex (293)

    DLD, PDHA1

    AR

    Leigh syndrome, French-Canadian type (294)

    LRPPRC

    AR

    220111

    Leigh syndrome, X-linked (295)

    PDHA1

    XR

    308930

    Leigh syndrome with growth retardation, co-enzyme Q10 deficiency (296301)

    COQ2, APTX, PDSS1, PDSS2, CABC1, COQ9

    AR

    607426

    Marie Unna hereditary hypotrichosis 1 (302)

    HR

    AD

    146550

    Menkes disease (303305)

    ATP7A

    XR

    309400

    Monilethrix (306308)

    KRTHB1, KRTHB3, KRTHB6

    AD

    158000

    Nail-patella syndrome (309)

    LMX1B

    AD

    161200

    T-cell immunodeficiency, congenital alopecia and nail dystrophy (310)

    FOXN1

    AR

    601705

    Toenail dystrophy, isolated (9)

    COL7A1

    AD

    607523

    Trichothiodystrophy, non-photosensitive 1 (311)

    C7ORF11

    AR

    234050

    Trichothiodystrophy, photosensitive (312314)

    ERCC2, ERCC3, GTF2H5

    AR

    601675

    Vitamin D-dependent rickets, type 2A (315)

    VDR

    AR

    277440

    Woolly hair, autosomal dominant (316)

    KRT74

    AD

    194300

    Woolly hair, autosomal recessive 1 (317)

    P2RY5

    AR

    278150

  6. 6.

    Genodermatoses with abnormal pigmentation

    Diseases

    Gene

    Mode of inheritance

    MIM

    Albinism, ocular, with sensorineural deafness (318)

    MITF and TYR (digenic)

    AD

    103470

    Albinism, oculocutaneous, type IA (319)

    TYR

    AR

    203100

    Albinism, oculocutaneous, type IB (320)

    TYR

    AR

    606952

    Albinism, oculocutaneous, type II (321)

    OCA2

    AR

    203200

    Albinism, oculocutaneous, type III (322)

    TYRP1

    AR

    203290

    Albinism, oculocutaneous, type IV (323)

    SLC45A2

    AR

    606574

    Chédiak–Higashi syndrome (324)

    LYST

    AR

    214500

    Dyschromatosis symmetrica hereditaria 1 (325)

    ADAR

    AD

    127400

    Griscelli syndrome, type 1 (326)

    MYO5A

    AR

    214450

    Griscelli syndrome, type 2 (327)

    RAB27A

    AR

    607624

    Griscelli syndrome, type 3 (328)

    MLPH, MYO5A

    AR

    609227

    Hermansky–Pudlak syndrome (329)

    HPS1

    AR

    203300

    Hermansky–Pudlak syndrome, type 2 (330)

    AP3B1

    AR

    608233

    Hermansky–Pudlak syndrome, type 3 (331)

    HPS3

    AR

    614072

    Hermansky–Pudlak syndrome, type 4 (332)

    HPS4

    AR

    606682

    Hermansky–Pudlak syndrome, type 5 (333)

    HPS5

    AR

    614074

    Hermansky–Pudlak syndrome, type 6 (333)

    HPS6

    AR

    614075

    Hermansky–Pudlak syndrome, type 7 (334)

    DTNBP1

    AR

    614076

    Hermansky–Pudlak syndrome, type 8 (335)

    BLOC1S3

    AR

    614077

    Incontinentia pigmenti (336)

    IKBKG

    XD

    308300

    Legius syndrome (337)

    SPRED1

    AD

    611431

    LEOPARD syndrome 1 (338)

    PTPN11

    AD

    151100

    LEOPARD syndrome 2 (100)

    RAF1

    AD

    611554

    LEOPARD syndrome 3 (102)

    BRAF

    AD

    613707

    McCune–Albright syndrome (339)

    GNAS1

    AD lethal

    174800

    Peripheral demyelinating neuropathy, central dysmyelination, Waardenburg syndrome and Hirschsprung disease (340)

    SOX10

    AD

    609136

    Piebald trait (341)

    KIT, SNAI2

    AD

    172800

    Poikiloderma with neutropenia (342)

    C16ORF57

    AR

    604173

    Tietz syndrome (343)

    MITF

    AD

    103500

    Waardenburg syndrome, type 1 (344)

    PAX3

    AD

    193500

    Waardenburg syndrome, type 2A (345)

    MITF

    AD

    193510

    Waardenburg syndrome, type 2D (346)

    SNAI2

    AR

    608890

    Waardenburg syndrome, type 2E (347)

    SOX10

    AD

    611584

    Waardenburg syndrome, type 3 (348)

    PAX3

    AD

    148820

    Waardenburg syndrome, type 4A (349)

    EDNRB

    AD, AR

    277580

    Waardenburg syndrome, type 4B (350)

    EDN3

    AD, AR

    613265

    Waardenburg syndrome, type 4C (351)

    SOX10

    AD

    613266

  7. 7.

    Metabolic genodermatoses

    Diseases

    Gene

    Mode of inheritance

    MIM

    Acrodermatitis enteropathica, zinc-deficiency type (352)

    SLC39A4

    AR

    201100

    Alkaptonuria (353)

    HGD

    AR

    203500

    Amyloidosis, familial visceral (13, 164, 165)

    APOA1, FGA, LYZ

    AD

    105200

    Angiooedema, hereditary, type I and type II (354, 355)

    C1NH

    AD

    106100

    Angiooedema, hereditary, type III (356)

    F12

    AD (female only)

    610618

    Apolipoprotein C-II deficiency (357)

    APOC2

    AR

    207750

    Biotinidase deficiency (358)

    BTD

    AR

    253260

    Cerebrotendinous xanthomatosis (359)

    CYP27A1

    AR

    213700

    Coproporphyria, hereditary (360)

    CPOX

    AD

    121300

    Fabry disease (361)

    GLA

    XR

    301500

    Farber lipogranulomatosis (362)

    ASAH

    AR

    228000

    Fucosidosis (363, 364)

    FUCA1

    AR

    230000

    Fumarase deficiency (365)

    FH

    AR

    606812

    Gaucher disease type I, II and III (366)

    GBA

    AR

    230800, 230900, 231000

    Hartnup disorder (367)

    SLC6A19

    AR

    234500

    Haemochromatosis (368, 369)

    HFE

    AR

    235200

    Haemochromatosis type 2A (370)

    HJV

    AR

    602390

    Haemochromatosis type 2B (371)

    HAMP

    AR

    613313

    Haemochromatosis type 3 (372)

    TFR2

    AR

    604250

    Haemochromatosis type 4 (373)

    SLC40A1

    AD

    606069

    Holocarboxydase synthetase deficiency (374)

    HLCS

    AR

    253270

    Homocystinuria due to cystathionine beta-synthase deficiency (375)

    CBS

    AR

    236200

    Hypercholesterolaemia, autosomal dominant (376)

    LDLR

    AD

    143890

    Hypercholesterolaemia, autosomal dominant, type B (377)

    APOB

    AD

    144010

    Hypercholesterolaemia, autosomal dominant, 3 (378)

    PCSK9

    AD

    603776

    Hyperlipoproteinaemia, type I (379, 380)

    LPL

    AR

    238600

    Hyperlipoproteinaemia, type III (381)

    APOE

    AD

    107741

    Hyperphenylalaninemia, BH4-deficient (382)

    QDPR

    AR

    261630

    Hyperphenylalaninemia, BH4-deficient, A (383)

    PTS

    AR

    261640

    Hypertriglyceridaemia, familial (384386)

    APOA5, LIPI, RP1

    AD

    145750

    Hypoalphalipoproteinaemia, familial (387, 388)

    ABCA1, APOA1

     

    604091

    Niemann–Pick disease, type A (389)

    SMPD1

    AR

    257200

    Phenylketonuria (390)

    PAH

    AR

    261600

    Porphyria, congenital erythropoietic (391)

    UROS

    AR

    263700

    Porphyria cutanea tarda (392)

    UROD

    AD

    176100

    Porphyria, hepatoerythropoietic (393)

    AR

    Porphyria, variegate (394)

    PPOX

    AD

    176200

    Prolidase deficiency (395)

    PEPD

    AR

    170100

    Protoporphyria, erythropoietic (396)

    FECH

    AD

    177000

    Sitosterolaemia (397, 398)

    ABCG5, ABCG8

    AR

    210250

    Tangier disease (387)

    ABCA1

    AR

    205400

    Wilson disease (399)

    ATP7B

    AR

    277900

  8. 8.

    Autoinflammatory genodermatoses

    Diseases

    Gene

    Mode of inheritance

    MIM

    Blau syndrome (400)

    NOD2

    AD

    186580

    CINCA syndrome (401)

    CIAS1

    AD

    607115

    Cold urticaria, immunodeficiency and autoimmunity related to PCLG2 deletions

    PMID: 22236196

    PCLG2

     

    600220

    Cyclic haematopoiesis (402)

    ELA2

    AD

    162800

    Familial cold autoinflammatory syndrome 1 (403)

    CIAS1

    AD

    120100

    Familial cold autoinflammatory syndrome 2 (157)

    NLRP12

    AD

    611762

    Familial Mediterranean fever (404)

    MEFV

    AR

    249100

    Familial Mediterranean fever, autosomal dominant (405)

    MEFV

    AD

    134610

    Hyper-IgD syndrome (406)

    MVK

    AR

    260920

    Majeed syndrome (407)

    LPIN2

    AR

    609628

    Muckle–Wells syndrome (403)

    CIAS1

    AD

    191900

    Periodic fever, familial, autosomal dominant (408)

    TNFRSF1A

    AD

    142680

    Pyogenic sterile arthritis, pyoderma gangrenosum and acne (409)

    PSTPIP1

    AD

    604416

  9. 9.

    Genodermatoses with lipodystrophy

    Diseases

    Gene

    Mode of inheritance

    MIM

    Donohue syndrome (410)

    INSR

    AR

    246200

    Lipodystrophy, congenital generalised, type 1 (411)

    AGPAT2

    AR

    608594

    Lipodystrophy, congenital generalised, type 2 (412)

    BSCL2

    AR

    269700

    Lipodystrophy, familial partial, type 2 (413)

    LMNA

    AD

    151660

    Lipodystrophy, familial partial, type 3 (414)

    PPARG

    AD

    604367

    Lipodystrophy, partial, acquired (415)

    LMNB2

    AD

    608709

  10. 10.

    Genodermatoses with premature ageing

    Diseases

    Gene

    Mode of inheritance

    MIM

    Cockayne syndrome, type A (416)

    ERCC8

    AR

    216400

    Cockayne syndrome, type B (417)

    ERCC6

    AR

    133540

    Ehlers–Danlos syndrome, progeroid form (193)

    B4GALT7

    AR

    130070

    Hutchinson–Gilford progeria syndrome (418)

    LMNA

    AD

    176670

    Mandibuloacral dysplasia with type A lipodystrophy (419)

    LMNA

    AD

    248370

    Mandibuloacral dysplasia with type B lipodystrophy (420)

    ZMPSTE24

    AR

    608612

    Werner syndrome (421)

    LMNA, HGPS

    AR

    277700

  11. 11.

    Genodermatoses with abnormal vascular system

    Diseases

    Gene

    Mode of inheritance

    MIM

    Capillary malformation, arteriovenous malformation (422)

    RASA1

    AD

    608354

    Cerebral cavernous malformations (423)

    KRIT1

    AD

    116860

    Cerebral cavernous malformations 2 (424)

    CCM2

    AD

    603284

    Cerebral cavernous malformations 3 (425)

    PDCD10

    AD

    603285

    Erythermalgia, primary (426)

    SCN9A

    AD

    133020

    Glomuvenous malformations (427)

    GLMN

    AD

    138000

    Haemangioma, capillary infantile (428)

    TEM8, VEGFR2

    AD

    602089

    Juvenile polyposis/hereditary haemorrhagic telangiectasia syndrome (429)

    SMAD4

    AD

    175050

    Lymphoedema, hereditary, IA (430)

    FLT4

    AD

    153100

    Lymphoedema, hereditary, IC (431)

    GJC2

    AD

    613480

    Lymphoedema-distichiasis syndrome (242)

    FOXC2

    AD

    153400

    Parkes–Weber syndrome (422)

    RASA1

    AD

    608355

    Telangiectasia, hereditary haemorrhagic, of Rendu, Osler and Weber (432)

    ENG

    AD

    187300

    Telangiectasia, hereditary haemorrhagic, type 2 (433)

    ACVRL1

    AD

    600376

  12. 12.

    Genodermatoses with abnormal immune system

    Diseases

    Gene

    Mode of inheritance

    MIM

    Autoimmune polyendocrine syndrome, type I (434)

    AIRE

    AR

    240300

    Candidiasis, familial, 2 (435)

    CARD9

    AR

    212050

    Candidiasis, familial, 4 (436)

    DEC1

    AR

    613108

    Congenital disorder of glycosylation, type Ia (437)

    PMM2

    AR

    212065

    Congenital disorder of glycosylation, type Id (438)

    ALG3

    AR

    601110

    Congenital disorder of glycosylation, type Ie (439)

    DPM1

    AR

    608799

    Congenital disorder of glycosylation, type If (440)

    MPDU1

    AR

    609180

    Congenital disorder of glycosylation, type Ij (441)

    DPAGT1

    AR

    608093

    Congenital disorder of glycosylation, type IIa (442)

    MGAT2

    AR

    212066

    Congenital disorder of glycosylation, type IIb (443)

    GCS1

    AR

    606056

    Congenital disorder of glycosylation, type IIc (444, 445)

    SLC35C1

    AR

    266265

    Congenital disorder of glycosylation, type IIe (446)

    COG7

    AR

    608779

    Congenital disorder of glycosylation, type IIg (447)

    COG1

    AR

    611209

    Congenital disorder of glycosylation, type IIj (448)

    COG4

    AR

    613489

    Congenital disorder of glycosylation, type IIk (449)

    ALG1

    AR

    608540

    Congenital disorder of glycosylation, type IIm (62)

    TMEM15

    AR

    610768

    Granulomatous disease, chronic, autosomal recessive, cytochrome b-positive, type I (450)

    NCF1

    AR

    233700

    Granulomatous disease, chronic, autosomal recessive, cytochrome b-positive, type II (450, 451)

    NCF2

    AR

    233710

    Granulomatous disease, chronic, autosomal recessive, cytochrome b-negative (450)

    CYBA

    AR

    233690

    Granulomatous disease, chronic, X-linked (452)

    CYBB

    XR

    306400

    Hyper-IgE recurrent infection syndrome, autosomal dominant (453)

    STAT3

    AD

    147060

    Hyper-IgE recurrent infection syndrome, autosomal recessive (454)

    DOCK8

    AR

    243700

    Immunodysregulation, polyendocrinopathy and enteropathy, X-linked (455)

    FOXP3

    XR

    304790

    Kawasaki disease (456)

    ITPKC

    AD

    611775

    Leucocyte adhesion deficiency, type I (457)

    ITGB2

    AR

    116920

    Leucocyte adhesion deficiency, type III (458)

    FERMT3

    AR

    612840

    Omenn syndrome (459, 460)

    RAG1, RAG2, DCLRE1C

    AR

    603554

    Severe combined immunodeficiency, autosomal recessive, T cell-negative, B cell-negative, NK cell-negative, due to adenosine deaminase deficiency (461)

    ADA

    AR

    102700

    Tyrosine kinase 2 deficiency (462)

    TYK2

    AR

    611521

    WHIM syndrome (463)

    CXCR4

    AR

    193670

    Wiskott–Aldrich syndrome (464)

    WAS

    XR

    301000

  13. 13.

    Genodermatoses with abnormal coagulation

    Diseases

    Gene

    Mode of inheritance

    MIM

    Antithrombin III deficiency (465)

    SERPINC1

    AD

    613118

    Haemolytic uraemic syndrome, atypical, susceptibility to, 1 (466, 467)

    CFH, CFHR1, CFHR3

    AD > AR

    235400

    Thrombophilia due to activated protein C resistance (468)

    F5

    AD

    188055

    Thrombophilia, hereditary, due to protein C deficiency, autosomal dominant (469)

    PROC

    AD

    176860

    Thrombophilia, hereditary, due to protein C deficiency, autosomal recessive (470)

    PROC

    AR

    612304

    Thrombophilia, hereditary, due to protein S deficiency, autosomal dominant (471)

    PROS1

    AD, AR more severe

    612336

    Thrombosis, susceptibility to (472)

    AT3, PROC, PROS, F5, HRG, PLG, PAI1, FGA, FGB, FGG, HCF2, THBD, MTHFR, HABP2

    AD

    188050

    Thrombotic, thrombocytopenic purpura, congenital (473)

    ADAMTS13

    AR

    274150

  14. 14.

    Genodermatoses with tumours/risk for tumour formation

    Diseases

    Gene

    Mode of inheritance

    MIM

    Adenomatous polyposis of the colon (474)

    APC

    AD

    175100

    Ataxia-telangiectasia (475)

    ATM

    AR

    208900

    Bannayan–Riley–Ruvalcaba syndrome (476)

    PTEN

    AD

    153480

    Basal cell naevus syndrome (477480)

    PTCH1, PTCH2, SUFU

    AD

    109400

    Birt–Hogg–Dubé syndrome (481)

    FLCN

    AD

    135150

    Bloom syndrome (482, 483)

    RECQL

    AR

    210900

    Brooke–Spiegler syndrome (484)

    CYLD

    AD

    605041

    Carney complex, type 1 (485)

    PRKAR1A

    AD

    160980

    Cowden disease (486)

    PTEN

    AD

    158350

    Cowden-like syndrome (487)

    SDHB, SDHD

    AD

    612359

    Cylindromatosis, familial (488)

    CYLD

    AD

    132700

    Dyskeratosis congenita, X-linked (489)

    DKC1

    XR

    305000

    Dyskeratosis congenita, autosomal dominant (490492)

    TERC, TERT, TINF2

    AD

    127550

    Dyskeratosis congenita, autosomal recessive (493, 494)

    NOLA2, NOLA3

    AR

    224230

    Epidermodysplasia verruciformis (495)

    TMC6, TMC8

    AR

    226400

    Leiomyomatosis, hereditary multiple, of skin (496)

    FH

    AD

    150800

    Leiomyomatosis and renal cell cancer (496)

    FH

    AD

    605839

    Lipomatosis, multiple symmetric (497)

    Multiple deletions

    mt

    151800

    Mast cell disease (498)

    KIT, TET2

    AD

    154800

    Melanoma, cutaneous malignant, susceptibility to, 2 (499)

    CDKN2A

    AD

    155601

    Melanoma, cutaneous malignant, susceptibility to, 3 (500)

    CDK4

    AD

    609048

    Melanoma, cutaneous malignant, susceptibility to, 5 (501)

    MC1R

    AD

    613099

    Melanoma-pancreatic cancer syndrome (502)

    CDKN2A

    AD

    606719

    Metachondromatosis (503)

    PTPN11

    AD

    156250

    Mismatch-repair cancer syndrome (504507)

    MLH1, MSH2, MSH6, PMS2

    AR

    276300

    Muir–Torre syndrome (508)

    MSH2, MLH1

    AD

    158320

    Multiple endocrine neoplasia, type I (509)

    MEN1

    AD

    131100

    Multiple endocrine neoplasia, type IIA (510)

    RET

    AD

    171400

    Multiple endocrine neoplasia, type IIB (511)

    RET

    AD

    162300

    Multiple endocrine neoplasia, type IV (512)

    CDKN1B

    AD

    610755

    Multiple self-healing squamous epithelioma (513)

    TGFBR1

    AD

    132800

    Neurofibromatosis type 1 (39)

    NF1

    AD

    162200

    Neurofibromatosis type 2 (514)

    NF2

    AD

    101000

    Neurofibromatosis-Noonan syndrome (515)

    NF1

    AD

    601321

    Peutz–Jeghers syndrome (516)

    STK11

    AD

    175200

    Proteus syndrome

    Pubmed ID: 21793738

    AKT1

    Somatic mosaicism

    176920

    Proteus-like syndrome (517)

    PTEN

    AR

    601728

    Rothmund–Thomson syndrome (518)

    RECQL4

    AR

    268400

    Trichoepithelioma, multiple familial, 1 (519)

    CYLD

    AD

    601606

    Tuberous sclerosis 1 (520)

    TSC1

    AD

    191100

    Tuberous sclerosis 2 (521)

    TSC2

    AD

    613254

    Venous malformations, multiple cutaneous and mucosal (522)

    TEK

    AD

    600195

    Watson syndrome (523)

    NF1

    AD

    193520

    Xeroderma pigmentosum, complementation group A (524)

    XPA

    AR

    278700

    Xeroderma pigmentosum, complementation group B (525)

    ERCC3

    AR

    610651

    Xeroderma pigmentosum, complementation group C (526)

    XPC

    AR

    278720

    Xeroderma pigmentosum, complementation group D (527)

    ERCC2

    AR

    278730

    Xeroderma pigmentosum, complementation group E (528)

    DDB2

    AR

    278740

    Xeroderma pigmentosum, complementation group F (529)

    ERCC4

    AR

    278760

    Xeroderma pigmentosum, complementation group G (530)

    ERCC5

    AR

    278780

    Xeroderma pigmentosum, variant type (531)

    POLH

    AR

    278750

Rights and permissions

Reprints and permissions

Copyright information

© 2013 Springer Science+Business Media, LLC

About this protocol

Cite this protocol

Wessagowit, V. (2013). Molecular Diagnosis of Genodermatoses. In: Has, C., Sitaru, C. (eds) Molecular Dermatology. Methods in Molecular Biology, vol 961. Humana Press, Totowa, NJ. https://doi.org/10.1007/978-1-62703-227-8_3

Download citation

  • DOI: https://doi.org/10.1007/978-1-62703-227-8_3

  • Published:

  • Publisher Name: Humana Press, Totowa, NJ

  • Print ISBN: 978-1-62703-226-1

  • Online ISBN: 978-1-62703-227-8

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics