Você está na página 1de 85

Reimpressão oficial do UpToDate ®

www.uptodate.com © 2021 UpToDate, Inc. e / ou suas afiliadas. Todos os direitos reservados.

As genodermatoses: uma visão geral


Autor: Teresa S Wright, MD, FAAD, FAAP
Editores de seção: Helen V Firth, DM, FRCP, FMedSci, Jennifer L Hand, MD
Editor-adjunto: Rosamaria Corona, MD, DSc

Todos os tópicos são atualizados conforme novas evidências se tornam disponíveis e nosso processo de
revisão por pares é concluído.

Revisão da literatura atualizada até: novembro de 2021. | Última atualização deste tópico: 21 de julho de
2020.

INTRODUÇÃO

As genodermatoses são um grande grupo de doenças hereditárias com manifestações


cutâneas. Muitos desses distúrbios são raros. No entanto, o reconhecimento de seus
achados cutâneos é importante não apenas para o início da terapia apropriada, mas
também para a detecção de outras anormalidades associadas, incluindo malignidade,
nessas doenças frequentemente multissistêmicas [ 1-3 ].

Uma visão geral das genodermatoses é apresentada aqui. Os recursos online que
fornecem informações gerais sobre esses distúrbios incluem Orphanet e Herança
Mendeliana Online no Homem (OMIM) .

TRANSTORNOS COM POTENCIAL MALIGNOS

Este grupo de genodermatoses é de particular importância devido à associação de


achados cutâneos com o desenvolvimento de neoplasias, tanto cutâneas como não
cutâneas ( tabela 1) Os exemplos incluem a síndrome do nevo basocelular, a síndrome
de Gardner, a síndrome de Peutz-Jeghers (PJS) e o xeroderma pigmentoso (XP).

Síndrome do nevo basocelular - A síndrome do nevo basocelular (síndrome do


carcinoma basocelular nevóide, síndrome de Gorlin, MIM # 109400) é um distúrbio raro
de herança autossômica dominante que resulta de mutações germinativas do gene
patched humano ( PTCH ). (Consulte "Síndrome do carcinoma de células basais nevóides
(síndrome de Gorlin)" .)

Os pacientes afetados apresentam anomalias de desenvolvimento e tumores pós-natais,


especialmente carcinomas basocelulares múltiplos (CBCs), geralmente por volta dos 35
anos. A maioria tem as seguintes características clínicas:

● Macrocefalia, protuberância frontal e hipertelorismo.

● Costelas bífidas.

● Palmar e corrosão plantar ( Imagem 1)

● Ceratocistos odontogênicos, especialmente na mandíbula, que geralmente se


desenvolvem na adolescência e normalmente são o sinal de apresentação da
doença.

● Meduloblastoma em 3 a 5 por cento; meningioma ocorre raramente. (Consulte


"Epidemiologia, patogênese e características clínicas do carcinoma basocelular" .)

A aparência histológica dos CBCs na síndrome do nevo basocelular não difere daquela
observada em casos esporádicos. O diagnóstico deve ser suspeitado em pacientes que
apresentam múltiplos CBCs, especialmente em idade precoce.

These patients require careful sun protection from infancy and regular skin surveillance by
a dermatologist. Radiotherapy should be avoided due to the risk of inducing BCCs in the
treatment fields.

Gardner syndrome — Gardner syndrome consists of familial adenomatous polyposis


(FAP, MIM #175100) with associated extraintestinal manifestations [4]. It is inherited as an
autosomal dominant disorder caused by mutations in the tumor suppressor gene,
adenomatous polyposis coli (APC). (See "Gardner syndrome".)

The most characteristic skin feature is multiple epidermoid cysts. Other findings include
desmoid tumors, lipomas, osteomas (especially of the mandible), supernumerary teeth,
gastric polyps, and juvenile nasopharyngeal angiofibromas. Congenital hypertrophy of the
retinal pigmented epithelium is a reliable and early marker of the disease when it is
present. Prophylactic colectomy is recommended because of the nearly universal
development of colorectal cancer in affected patients.
In addition to colorectal adenocarcinoma, patients with FAP are at risk for several
extracolonic malignancies, including:

● Duodenal ampullary carcinoma


● Follicular or papillary thyroid cancer
● Childhood hepatoblastoma
● Gastric carcinoma
● Central nervous system (CNS) tumors (mostly medulloblastomas)

Peutz-Jeghers syndrome — Peutz-Jeghers syndrome (PJS) is a rare, autosomal dominant


condition characterized by distinctive mucocutaneous pigmentations and multiple
hamartomatous polyps in the gastrointestinal tract [5]. Most cases are associated with
mutations in the tumor suppressor gene STK11 (serine/threonine kinase 11)/LKB1 [6]. (See
"Peutz-Jeghers syndrome: Clinical manifestations, diagnosis, and management".)

The characteristic mucocutaneous pigmentations (lentigines) of PJS are present in more


than 95 percent of patients and are caused by pigment-laden macrophages in the dermis.
They are typically flat, blue-gray to brown spots 1 to 5 mm in size that look like freckles.
However, the onset and location of PJS spots are different from those of freckles.

Lentigines occur most commonly on the lips and perioral region (94 percent), hands (74
percent), buccal mucosa (66 percent), and feet (62 percent) ( picture 2) [7]. They also
occur on the nose, perianal area, and genitals and are rarely found in the intestines. They
usually occur during the first one to two years of life, increase in size and number over the
ensuing years, and finally fade after puberty, with the exception of those on the buccal
mucosa.

Gastrointestinal hamartomatous polyps are present in most patients with PJS, and
patients may develop gastrointestinal malignancy. The risk of nongastrointestinal cancers,
including adenocarcinomas of the breast, cervix, pancreas, uterus, and ovaries, is also
increased.

Hereditary leiomyomatosis and renal cell cancer — Hereditary leiomyomatosis and


renal cell cancer (HLRCC; MIM #150800) is caused by autosomal dominant, heterozygous
mutations in the fumarate hydratase gene (FH) [8]. HLRCC presents in early adulthood
with multiple cutaneous leiomyomas, most frequently on the trunk and extremities (
picture 3A-D). Affected women frequently develop uterine leiomyomas at an early age.
HLRCC is associated with an increased risk of early-onset, aggressive renal cancer [9]. (See
"Hereditary leiomyomatosis and renal cell cancer (HLRCC)".)

Xeroderma pigmentosum — Xeroderma pigmentosum (XP) is a rare, autosomal


recessive disorder caused by mutations in any of eight genes involved in the recognition
and repair of ultraviolet radiation (UVR)-induced DNA damage [10]. XP is characterized by
increased sensitivity to UVR, early development of pigmentary changes and UVR-induced
skin and mucous membrane cancers (beginning in early childhood), and, in some patients,
progressive neurodegeneration.

The pathogenesis, clinical manifestations, diagnosis, and management of XP are


discussed in detail separately. (See "Xeroderma pigmentosum".)

Epidermolysis bullosa — Patients with particular subtypes of epidermolysis bullosa are at


increased risk for cutaneous malignancy. (See "Epidermolysis bullosa: Epidemiology,
pathogenesis, classification, and clinical features", section on 'Skin cancer'.)

DISORDERS OF KERATINIZATION

Keratins are intermediate filament proteins that form the cytoskeleton in all epithelial
cells, including the stratified epithelium of the epidermis [11]. Keratins represent the
major proteins produced by the keratinocyte, which is the primary cell type of the
epidermis. The maturation of basal epidermal cells to the flattened cells that constitute
the superficial stratum corneum is known as keratinization [12].

Over 50 genes that encode keratins have been identified in humans [13]. The phenotype
resulting from a particular mutation depends upon the tissue-specific expression pattern
of that keratin.

Ichthyoses — The ichthyoses are a diverse group of hereditary skin disorders


characterized by the accumulation of "fish-like" scales resulting from abnormal epidermal
cell kinetics or differentiation ( table 2) [14]. The severity of the individual disorders
ranges from asymptomatic to life threatening.

The cornerstone of therapy for all types is aggressive hydration of the skin with
emollients. When tolerated, keratolytics also may be used. Severe or extensive
involvement may require systemic retinoids.

Referral to a dermatologist is indicated when basic treatment measures, such as


emollients, are not working, when there are complications related to the skin condition, or
if the diagnosis is not clear. Biopsy is useful for certain types of ichthyoses or disorders of
cornification.

The major types of inherited ichthyoses are reviewed separately. Information for patients
and families is available on the website of the Foundation for Ichthyosis and Related Skin
Types.

● (See "Overview and classification of the inherited ichthyoses".)


● (See "Autosomal recessive congenital ichthyosis".)
● (See "Recessive X-linked ichthyosis".)
● (See "Keratinopathic ichthyoses".)
● (See "Netherton syndrome".)
● (See "Sjögren-Larsson syndrome".)

Palmoplantar keratodermas — These disorders share the common feature of palmar


and plantar hyperkeratosis that manifests as thickening and scaling of the palms and
soles. The general underlying defect in the majority of the palmoplantar keratodermas is
overproduction of a normal or an abnormal keratin in the palms and soles. The majority of
cases are mild to moderate, without systemic problems and with autosomal dominant
inheritance.

The keratodermas differ in their mode of inheritance, severity, and extent of involvement
and associated features [15,16]. This is illustrated by the following examples of these
disorders:

● Howel-Evans syndrome is a rare, autosomal dominant diffuse form with onset


between 5 and 15 years of age [17]. It has been associated with the early
development of esophageal cancer.

● Vohwinkel syndrome is a rare, autosomal dominant disorder in which patients may


have autoamputation of the digits (pseudoainhum) and high-frequency hearing loss.

● Papillon-Lefèvre syndrome is an autosomal recessive condition that presents in the


first six months of life. Patients often have severe periodontitis, leading to early
dental loss.

The clinical presentation, diagnosis, and management of palmoplantar keratodermas are


reviewed in detail elsewhere. (See "Palmoplantar keratoderma".)
Pachyonychia congenita — Pachyonychia congenita (PC) is an autosomal dominant
disorder caused by mutations in the genes that encode keratins (KRT6A, KRT6B, KRT6C,
KRT16, and KRT17), the type I and II intermediate filament proteins that form a cytoskeletal
network in all epithelial cells [18]. Affected patients present with thickened, discolored
nails of the fingers and toes ( picture 4). These changes are present at birth in
approximately 50 percent of the affected children [19]. Palmar and plantar hyperkeratoses
and hyperhidrosis, follicular keratoses of the knees and elbows, and oral leukoplakia may
develop within the first decade of life. Thickened nails and plantar hyperkeratoses may be
extremely painful [20].

The pathogenesis, clinical manifestations, diagnosis, and management of PC are


discussed in detail separately. (See "Pachyonychia congenita".)

Darier disease — Darier disease, also known as Darier-White disease or keratosis


follicularis (MIM #124200), is an autosomal dominant disorder caused by mutations in the
gene encoding the sarco/endoplasmic reticulum Ca(+2)-ATPase [21]. This results in loss of
adhesion between epidermal cells and abnormal keratinization. The disorder is a relatively
common genodermatosis with a frequency of up to 1 in 36,000 individuals [22].

The disorder typically presents in the second decade of life with hyperkeratotic, yellow-
brown, greasy-appearing papules that coalesce into verrucous-like plaques (
picture 5A-B) [23,24]. The lesions are often pruritic and frequently become purulent and
malodorous, especially if infected. Typical sites of involvement are in a seborrheic
distribution: trunk, face, scalp, and groin. Nails may demonstrate red/white vertical
stripes, subungual hyperkeratosis, and notching of the distal nail margins ( picture 6).
Palmar keratosis and pits often are present.

The course of the illness is chronic and persistent, with characteristic worsening in
summer months. Darier disease is discussed in detail separately. (See "Darier disease".)

GENETIC BLISTERING DISORDERS

These disorders result from abnormalities in the cohesion of the layers of the epidermis.
They result in separation of the layers in response to minimal injury.

Epidermolysis bullosa — Epidermolysis bullosa (EB) constitutes a clinically and


genetically heterogeneous group of rare inherited disorders characterized by marked skin
and mucosal fragility caused by mutations in skin structural proteins. There are four major
types of EB, based upon the ultrastructural level of tissue cleavage in the skin: EB simplex,
junctional EB, dystrophic EB, and Kindler EB ( table 3) [25,26]. Many subtypes have been
identified based upon clinical, pathophysiologic, and molecular criteria ( table 4A-D).

The clinical features, diagnosis, and management of EB are discussed in detail separately.
(See "Epidermolysis bullosa: Epidemiology, pathogenesis, classification, and clinical
features" and "Diagnosis of epidermolysis bullosa" and "Overview of the management of
epidermolysis bullosa".)

PIGMENTATION DISORDERS

Melanin, a black or brown pigment formed from tyrosine, is responsible for the color of
skin and hair [27]. Melanin is synthesized in melanocytes, which are specialized, dendritic
secretory cells derived from the neural crest. These cells migrate to the basal layer of the
epidermis during embryogenesis. The presence of melanin in the epidermis helps provide
protection from ultraviolet radiation.

Disorders include decreased and excessive pigmentation. Diagnosis is based on the


clinical features in most cases, although some may be clarified with molecular testing.
(See "Congenital and inherited hyperpigmentation disorders".)

Oculocutaneous albinism — Oculocutaneous albinism (OCA) is a group of rare genetic


disorders of melanin biosynthesis inherited in an autosomal recessive pattern [28,29].
There are seven types of OCA caused by mutations in different genes ( table 5).
Although all types share absent or reduced pigmentation of the hair, skin, and eyes, the
clinical phenotypes vary along a broad spectrum of disease severity.

The pathogenesis, clinical manifestations, diagnosis, and management of OCA are


discussed in detail separately. (See "Oculocutaneous albinism".)

Ocular albinism — Ocular albinism is albinism in which the hypopigmentation is primarily


limited to the eyes [30,31]. It is less common than oculocutaneous albinism.

Ocular albinism type 1 (OA1, MIM #300500), also known as Nettleship-Falls ocular
albinism, is the most common form of ocular albinism and has X-linked recessive
inheritance. It has an estimated prevalence of 1 in 50,000 to 150,000 live births [32,33].

The clinical manifestations of OA1 are variable. In affected males, clinical features may
include mild cutaneous hypopigmentation, hypopigmentation of the iris and retina, foveal
hypoplasia, prominent choroidal vessels, nystagmus, strabismus, head nodding,
photophobia, impaired vision, and abnormal crossing of the optic fibers resulting in
deficient stereoscopic vision [31,34,35]. Female carriers may have a patchy distribution of
retinal pigmentation resulting from X-inactivation [31,36].

OA1 is diagnosed by careful analysis of the family pedigree for X-linked inheritance and/or
molecular analysis of the OA1 gene [36]. The severity of OA1 appears to be related to
ethnic background, with individuals from lightly pigmented racial groups more severely
affected than those from darkly pigmented groups [37-39]. Life expectancy is normal [30].

OA1 has been associated with late-onset sensorineural deafness. This form (OASD, MIM
300650) is probably a contiguous gene defect that includes the OA1 gene [40,41]. Another
form of ocular albinism with sensorineural deafness has been linked to chromosome 11
and has autosomal recessive inheritance; this form is also known as Waardenburg
syndrome type 2 (MIM 103470) [42]. (See 'Waardenburg syndrome' below.)

Ocular albinism type 2 (OA2, MIM #300600), also known as Forsius-Eriksson type ocular
albinism and Aland Island eye disease, is a rare, X-linked disorder with clinical
manifestations that include nystagmus, myopia, astigmatism, foveal hypoplasia, reduced
visual acuity, pigmentary changes in the retina, and changes in color vision; the optic
nerves are normal [30].

Piebaldism — Piebaldism (piebald trait) is a rare, autosomal dominant disorder in which


cell proliferation and migration of neural crest-derived melanoblasts are defective. This
leads to an abnormal distribution of melanocytes during embryogenesis and results in
patchy areas of depigmentation [43]. The disorder is caused by mutations in the cell-
surface receptor tyrosine kinase gene (KIT) [44].

Affected patients have patches of depigmented skin, with hyperpigmented borders


occurring principally on the midforehead, neck, anterior trunk, and midextremities (
picture 7A-C). Normal pigmentation occurs on the hands, feet, back, shoulders, and
hips. A white forelock is a common finding. The depigmentation is stable and permanent.
Patients with piebaldism are generally otherwise healthy and have normal life spans.

The pathogenesis, clinical manifestations, diagnosis, and management of piebaldism are


discussed in detail separately. (See "Piebaldism".)

Waardenburg syndrome — Waardenburg syndrome is an autosomal dominant inherited


pigmentary disorder in which abnormal distribution of melanocytes during
embryogenesis results in patchy areas of depigmentation [43,45,46]. Several forms of
Waardenburg syndrome are described. All have the clinical features of type 1, which is
characterized by a piebald-like distribution of patchy depigmentation of the hair and skin.
Other distinctive noncutaneous features include pigmentary abnormalities of the iris
(heterochromia irides) and a broad nasal root, secondary to lateral displacement of the
inner canthi of the eyes ( picture 8). Congenital deafness occurs in one in five patients
with Waardenburg syndrome, and, conversely, an estimated 2 to 7 percent of cases of
congenital deafness result from the disorder [47,48]. Occasional findings in Waardenburg
syndrome type 1 include cleft lip and palate and neural tube defects (eg, spina bifida,
myelomeningocele) [49].

Waardenburg syndrome type 1 (MIM #193500) and type 3 (MIM #148820) are caused by
mutations in the gene for one of three transcription factors (PAX3), whereas type 2 (MIM
#193510) is caused by mutations in the transcription factor MITF [50,51]. Waardenburg
syndrome type 4 (MIM #277580) also has features of Hirschsprung disease. This type is a
result of biallelic mutation in the genes for the endothelin-B receptor (EDNRB) or its ligand
endothelin-B (EDN3) [52,53] or heterozygous mutation in the SOX10 gene [54]. (See
"Congenital aganglionic megacolon (Hirschsprung disease)".)

NEUROCUTANEOUS SYNDROMES

Neurocutaneous genetic disorders, also called phakomatoses, may present with a variety
of neurologic and cutaneous findings. Examples include the neurofibromatoses and
tuberous sclerosis complex.

Neurofibromatosis type 1 — Neurofibromatosis type 1 (NF1), also known as von


Recklinghausen's disease, is an autosomal dominant neurocutaneous disorder with
nervous system, skeletal, and dermatologic manifestations [55]. It is caused by mutations
in the NF1 gene, encoding the protein neurofibromin. (See "Neurofibromatosis type 1
(NF1): Pathogenesis, clinical features, and diagnosis".)

The characteristic skin findings that contribute to establishing the diagnosis are:

● Six or more café-au-lait macules of greatest diameter >5 mm in prepubertal and >15
mm in postpubertal individuals ( picture 9A)

● Two or more neurofibromas of any type or one plexiform neurofibroma (


picture 9B)

● Freckling in the axillary or inguinal regions (Crowe sign) ( picture 9C)

Neurofibromatosis type 2 — Neurofibromatosis type 2 (NF2) is the central form of


neurofibromatosis and is characterized by bilateral vestibular schwannomas (acoustic
neuromas), meningiomas of the brain, and schwannomas/neurilemmomas of the dorsal
roots of the spinal cord. The disorder typically presents in the teens or soon after puberty
with unilateral hearing loss. In contrast to NF1, café-au-lait spots in NF2 are typically few,
large, and relatively light in color [56]. NF2 is caused by mutations in the gene encoding
the intracellular membrane-associated protein neurofibromin-2 (NF-2), a tumor
suppressor, which is also known as merlin [57]. (See "Neurofibromatosis type 2".)

Tuberous sclerosis complex — Tuberous sclerosis complex (TSC) is an autosomal


dominant neurocutaneous disorder that's skin findings are often the first clues to its
diagnosis [58,59]. TSC is caused by mutations in one of two genes: TSC1, which encodes
hamartin, and TSC2, which encodes tuberin. (See "Tuberous sclerosis complex: Genetics,
clinical features, and diagnosis".)

It is estimated that more than 95 percent of patients with TSC have one of the
characteristic skin lesions [59]. The most common lesions are:

● Hypopigmented macules, also known as ash-leaf spots, which are usually elliptical in
shape ( picture 10A). These are often present at birth, although a Wood's lamp
examination may be required to visualize them.

● Angiofibromas, previously called adenoma sebaceum, which typically involve the


malar regions of the face ( picture 10B) and usually become apparent by late
childhood or early adolescence.

● Shagreen patches (connective tissue nevi), seen most commonly over the lower
trunk.

● A distinctive brown, fibrous plaque on the forehead, which may be the first and most
readily recognized feature of TSC to appear on physical examination of affected
neonates and infants ( picture 11) [59].

Ataxia-telangiectasia — Ataxia-telangiectasia (AT, also known as Louis-Bar syndrome) is


an autosomal recessive disorder caused by mutations in the gene designated ATM
(AT mutated). The ATM gene, which is expressed in all tissues in the body, is involved in the
detection of DNA damage and plays an important role in cell cycle progression. The
pathogenesis of AT is thought to be a defect in DNA repair resulting in increased
sensitivity to ionizing radiation, immunodeficiency, and progressive cerebellar Purkinje cell
death.

Patients with AT suffer from progressive cerebellar ataxia and other neurologic
abnormalities, oculocutaneous telangiectasias, and immune deficiency. Associated
features are an increased incidence of malignancy, radiation sensitivity, and diabetes
mellitus caused by insulin resistance. (See "Ataxia-telangiectasia".)

VASCULAR DISORDERS

Inherited syndromes associated with cutaneous vascular abnormalities include ataxia-


telangiectasia (AT) and hereditary hemorrhagic telangiectasia (HHT), also known as Osler-
Weber-Rendu syndrome. These disorders are discussed separately. (See "Ataxia-
telangiectasia" and "Clinical manifestations and diagnosis of hereditary hemorrhagic
telangiectasia (Osler-Weber-Rendu syndrome)".)

DISORDERS OF CONNECTIVE TISSUE

Abnormalities of connective tissue are frequently expressed in the skin. Thus, multisystem
inherited connective tissue disorders, such as Ehlers-Danlos syndrome, Marfan syndrome,
and osteogenesis imperfecta, can be classified as genodermatoses. Pseudoxanthoma
elasticum (PXE) and focal dermal hypoplasia are less common connective tissue disorders
with prominent skin abnormalities.

● (See "Focal dermal hypoplasia (Goltz syndrome)".)


● (See "Clinical manifestations and diagnosis of Ehlers-Danlos syndromes".)
● (See "Overview of the management of Ehlers-Danlos syndromes".)
● (See "Genetics, clinical features, and diagnosis of Marfan syndrome and related
disorders".)
● (See "Management of Marfan syndrome and related disorders".)
● (See "Osteogenesis imperfecta: An overview".)

Pseudoxanthoma elasticum — Pseudoxanthoma elasticum (PXE, also called Grönblad-


Strandberg syndrome, MIM #264800 and #177850) is a genetic disorder of abnormal
elastic tissue (fragmentation of elastic fibers) and calcification. Inherited forms are
autosomal recessive, but sporadic forms have been observed [60]. The underlying defect
is a mutation in the ABCC6 gene on chromosome 16 that encodes an ATP-binding cassette
transporter [61-63].

The onset of the disorder varies from childhood to early adulthood, although the
characteristic skin findings may be subtle in children. The primary organ systems involved
include the skin, eyes, and cardiovascular systems; gastrointestinal bleeding also can
occur [64-66].

Progressive skin lesions develop in 80 percent of individuals before age 20 years. The
characteristic skin findings are 2 to 5 mm yellow to orange papules, which may coalesce
into irregularly shaped plaques surrounded by normal skin. Because such lesions have a
pebbly appearance and are yellow, they are named pseudoxanthomas. The texture of the
skin has been likened to plucked-chicken skin. Lesions occur most commonly in flexural
areas, such as the neck and axillary folds, periumbilical region, and on the inner lower lip (
picture 12A-D). The involved skin may eventually become lax and redundant.

The primary ocular finding is that of angioid streaks, representing tears in Bruch's
membrane, but this finding is seen in other disorders as well ( picture 12E). Macular
degeneration and retinal deformities contribute to loss of central vision. Severe vision loss
occurs in 3 to 8 percent of patients.

Common cardiovascular manifestations include accelerated atherosclerosis, which is


thought to result from calcification of the internal elastic laminae. These changes can
result in myocardial infarction, cerebrovascular disease, and renovascular hypertension at
a young age and result in a shortened life expectancy.

The diagnosis of PXE is based upon the clinical appearance and findings on histologic
examination of lesional skin. Treatment consists of close ophthalmologic management,
monitoring and treatment of any cardiovascular symptoms, and dietary consultation.
Because of the risk of ocular disease, patients should be educated to avoid contact sports
and straining (eg, weight lifting) [66]. They should be referred for genetic counseling.

X-LINKED DOMINANT DISORDERS

Incontinentia pigmenti (IP), focal dermal hypoplasia, congenital hemidysplasia with


ichthyosiform erythroderma and limb defects (CHILD syndrome, MIM #308050), and
chondrodysplasia punctata (Conradi-Hünermann-Happle syndrome, MIM #302960) are
examples of X-linked dominant disorders with cutaneous manifestations [67,68].

● (See "Overview and classification of the inherited ichthyoses", section on 'X-linked


dominant disorders'.)
● (See "Focal dermal hypoplasia (Goltz syndrome)".)
● (See "Incontinentia pigmenti".)

ECTODERMAL DYSPLASIAS

The ectodermal dysplasias are a large group of inherited disorders that manifest as
developmental anomalies in at least two of the structures derived from the embryonic
ectoderm, with at least one involving the skin appendages (hair, nails, sweat glands) or
teeth ( table 6) [69]. The classic ectodermal dysplasias, including hypohidrotic
ectodermal dysplasia, hypohidrotic ectodermal dysplasia with immune deficiency, and
hidrotic ectodermal dysplasia; tumor protein p63-related disorders; and focal dermal
hypoplasias are discussed separately.

● (See "Ectodermal dysplasias".)


● (See "Tumor protein p63-related disorders".)
● (See "Focal dermal hypoplasia (Goltz syndrome)".)

SUMMARY

● The genodermatoses are a heterogeneous group of rare inherited single-gene


disorders with skin manifestations. Recognition is important for the initiation of
appropriate dermatologic therapy and detection of other associated abnormalities,
including malignancy. (See 'Introduction' above.)

● Genodermatoses associated with the development of cutaneous and noncutaneous


malignancies include basal cell nevus syndrome ( picture 1), Gardner syndrome,
Peutz-Jeghers syndrome (PJS) ( picture 2), and xeroderma pigmentosum (XP) (
table 1). (See 'Disorders with malignant potential' above.)

● Keratins form the cytoskeleton in epithelial cells. Disorders of keratinization include


the ichthyosiform dermatoses ( table 2 and picture 13A-C), palmoplantar
keratodermas, pachyonychia congenita ( picture 4), and Darier disease (
picture 5A-B). (See 'Disorders of keratinization' above.)
● Abnormalities in the cohesion of the layers of the epidermis underlie the congenital
blistering disorders (ie, the epidermolysis bullosa syndromes), in which blister
formation occurs with little or no trauma ( picture 14A-B). (See 'Epidermolysis
bullosa' above.)

● Congenital defects in melanin synthesis lead to pigmentation disorders. These


include oculocutaneous albinism, a group of autosomal recessive disorders resulting
in hypopigmentation of the hair, skin, and eyes ( table 5); piebaldism; and
Waardenburg syndrome ( picture 8). (See 'Pigmentation disorders' above.)

● Cutaneous findings are often a key to the diagnosis of the most common
neurocutaneous syndromes: neurofibromatosis ( picture 9A-C), tuberous sclerosis
complex (TSC) ( picture 10A-B), and ataxia-telangiectasia (AT). (See
'Neurocutaneous syndromes' above.)

● Multisystem inherited connective tissue disorders include Ehlers-Danlos syndrome,


Marfan syndrome, osteogenesis imperfecta, and pseudoxanthoma elasticum (
picture 12A-B, 12E). (See 'Disorders of connective tissue' above.)

● X-linked disorders with cutaneous manifestations include incontinentia pigmenti (


picture 15) and focal dermal hypoplasia ( picture 16). (See 'X-linked dominant
disorders' above.)

● The ectodermal dysplasias are a large group of inherited disorders that manifest as
developmental anomalies in at least two of the structures derived from the
embryonic ectoderm, with at least one involving the skin appendages (hair, nails,
sweat glands) or teeth ( table 6). (See 'Ectodermal dysplasias' above.)

Use of UpToDate is subject to the Subscription and License Agreement.

REFERENCES

1. Spitz JL. Genodermatoses: A Full-Color Clinical Guide to Genetic Skin Disorders, Willia
ms & Wilkins, New York 1996.
2. Sybert VP. Genetic Skin Disorders, Oxford University Press, New York 1997.
3. Reyes MA, Eisen DB. Inherited syndromes. Dermatol Ther 2010; 23:606.
4. Perniciaro C. Gardner's syndrome. Dermatol Clin 1995; 13:51.
5. Griffith CD, Bisset WH. Peutz-Jeghers syndrome. Arch Dis Child 1980; 55:866.
6. Scott RJ, Crooks R, Meldrum CJ, et al. Mutation analysis of the STK11/LKB1 gene and
clinical characteristics of an Australian series of Peutz-Jeghers syndrome patients. Clin
Genet 2002; 62:282.
7. Utsunomiya J, Gocho H, Miyanaga T, et al. Peutz-Jeghers syndrome: its natural course
and management. Johns Hopkins Med J 1975; 136:71.

8. Toro JR, Nickerson ML, Wei MH, et al. Mutations in the fumarate hydratase gene cause
hereditary leiomyomatosis and renal cell cancer in families in North America. Am J
Hum Genet 2003; 73:95.

9. Menko FH, Maher ER, Schmidt LS, et al. Hereditary leiomyomatosis and renal cell
cancer (HLRCC): renal cancer risk, surveillance and treatment. Fam Cancer 2014;
13:637.

10. DiGiovanna JJ, Kraemer KH. Shining a light on xeroderma pigmentosum. J Invest
Dermatol 2012; 132:785.

11. Porter RM, Lane EB. Phenotypes, genotypes and their contribution to understanding
keratin function. Trends Genet 2003; 19:278.
12. Roop D. Defects in the barrier. Science 1995; 267:474.
13. McLean WH, Moore CB. Keratin disorders: from gene to therapy. Hum Mol Genet
2011; 20:R189.
14. Oji V, Tadini G, Akiyama M, et al. Revised nomenclature and classification of inherited
ichthyoses: results of the First Ichthyosis Consensus Conference in Sorèze 2009. J Am
Acad Dermatol 2010; 63:607.
15. Itin PH, Lautenschlager S. Palmoplantar keratoderma and associated syndromes.
Semin Dermatol 1995; 14:152.
16. Lucker GP, Van de Kerkhof PC, Steijlen PM. The hereditary palmoplantar keratoses: an
updated review and classification. Br J Dermatol 1994; 131:1.
17. Hereditary disorders of cornification. In: Hurwitz Clinical Pediatric Dermatology: A Tex
tbook of Skin Disorders of Childhood and Adolescence, 3rd ed, Paller AS, Mancini AJ (E
ds), W.B. Elsevier Saunders, Philadelphia 2006. p.107.
18. Wilson NJ, O'Toole EA, Milstone LM, et al. The molecular genetic analysis of the
expanding pachyonychia congenita case collection. Br J Dermatol 2014; 171:343.

19. Shah S, Boen M, Kenner-Bell B, et al. Pachyonychia congenita in pediatric patients:


natural history, features, and impact. JAMA Dermatol 2014; 150:146.
20. Eliason MJ, Leachman SA, Feng BJ, et al. A review of the clinical phenotype of 254
patients with genetically confirmed pachyonychia congenita. J Am Acad Dermatol
2012; 67:680.
21. Sakuntabhai A, Ruiz-Perez V, Carter S, et al. Mutations in ATP2A2, encoding a Ca2+
pump, cause Darier disease. Nat Genet 1999; 21:271.
22. Ringpfeil F, Raus A, DiGiovanna JJ, et al. Darier disease--novel mutations in ATP2A2
and genotype-phenotype correlation. Exp Dermatol 2001; 10:19.
23. Burge S. Darier's disease--the clinical features and pathogenesis. Clin Exp Dermatol
1994; 19:193.

24. Burge SM, Wilkinson JD. Darier-White disease: a review of the clinical features in 163
patients. J Am Acad Dermatol 1992; 27:40.

25. Fine JD, Eady RA, Bauer EA, et al. The classification of inherited epidermolysis bullosa
(EB): Report of the Third International Consensus Meeting on Diagnosis and
Classification of EB. J Am Acad Dermatol 2008; 58:931.

26. Intong LR, Murrell DF. Inherited epidermolysis bullosa: new diagnostic criteria and
classification. Clin Dermatol 2012; 30:70.
27. Disorders of pigmentation. In: Hurwitz Clinical Pediatric Dermatology: A Textbook of S
kin Disorders of Childhood and Adolescence, 3rd ed, Paller AS, Mancini AJ (Eds), W.B. E
lsevier Saunders, Philadelphia 2006. p.265.
28. Summers CG. Albinism: classification, clinical characteristics, and recent findings.
Optom Vis Sci 2009; 86:659.
29. Grønskov K, Ek J, Brondum-Nielsen K. Oculocutaneous albinism. Orphanet J Rare Dis
2007; 2:43.
30. King RA, Hearing VJ, Dreel DJ, Oetting WS. Albinism. In: Metabolic and Molecular Base
s of Inherited Disease, 8th ed, Scriver CR, Beaudet AL, Sly WS, Valle D (Eds), McGraw-H
ill, New York 2001. p.5587.
31. Spritz RA. Molecular genetics of oculocutaneous albinism. Hum Mol Genet 1994; 3
Spec No:1469.
32. Rosenberg T, Schwartz M. X-linked ocular albinism: prevalence and mutations--a
national study. Eur J Hum Genet 1998; 6:570.
33. van Dorp DB. Albinism, or the NOACH syndrome (the book of Enoch c.v. 1-20). Clin
Genet 1987; 31:228.
34. Albinism, ocular, type I;OA1. In: Online Mendelian Inheritance in Man. Johns Hopkins
University. www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=300500 (Accessed on Nove
mber 02, 2006).

35. Shen B, Samaraweera P, Rosenberg B, Orlow SJ. Ocular albinism type 1: more than
meets the eye. Pigment Cell Res 2001; 14:243.
36. Oetting WS. New insights into ocular albinism type 1 (OA1): Mutations and
polymorphisms of the OA1 gene. Hum Mutat 2002; 19:85.
37. O'Donnell FE Jr, Green WR, Fleischman JA, Hambrick GW. X-linked ocular albinism in
Blacks. Ocular albinism cum pigmento. Arch Ophthalmol 1978; 96:1189.
38. Shiono T, Tsunoda M, Chida Y, et al. X linked ocular albinism in Japanese patients. Br J
Ophthalmol 1995; 79:139.
39. Lyle WM, Sangster JO, Williams TD. Albinism: an update and review of the literature. J
Am Optom Assoc 1997; 68:623.
40. Winship I, Gericke G, Beighton P. X-linked inheritance of ocular albinism with late-
onset sensorineural deafness. Am J Med Genet 1984; 19:797.
41. Winship IM, Babaya M, Ramesar RS. X-linked ocular albinism and sensorineural
deafness: linkage to Xp22.3. Genomics 1993; 18:444.
42. Albinism, ocular, with sensorineural deafness. In: Online Mendelian Inheritance in Ma
n. Johns Hopkins University www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=103470 (A
ccessed on November 02, 2006).
43. Spritz RA. Piebaldism, Waardenburg syndrome, and related disorders of melanocyte
development. Semin Cutan Med Surg 1997; 16:15.

44. Syrris P, Heathcote K, Carrozzo R, et al. Human piebaldism: six novel mutations of the
proto-oncogene KIT. Hum Mutat 2002; 20:234.

45. Dourmishev AL, Dourmishev LA, Schwartz RA, Janniger CK. Waardenburg syndrome.
Int J Dermatol 1999; 38:656.
46. Read AP, Newton VE. Waardenburg syndrome. J Med Genet 1997; 34:656.
47. Nayak CS, Isaacson G. Worldwide distribution of Waardenburg syndrome. Ann Otol
Rhinol Laryngol 2003; 112:817.
48. Silan F, Zafer C, Onder I. Waardenburg syndrome in the Turkish deaf population.
Genet Couns 2006; 17:41.
49. Hart J, Miriyala K. Neural tube defects in Waardenburg syndrome: A case report and
review of the literature. Am J Med Genet A 2017; 173:2472.
50. Potterf SB, Furumura M, Dunn KJ, et al. Transcription factor hierarchy in Waardenburg
syndrome: regulation of MITF expression by SOX10 and PAX3. Hum Genet 2000;
107:1.
51. Pingault V, Ente D, Dastot-Le Moal F, et al. Review and update of mutations causing
Waardenburg syndrome. Hum Mutat 2010; 31:391.
52. Edery P, Attié T, Amiel J, et al. Mutation of the endothelin-3 gene in the Waardenburg-
Hirschsprung disease (Shah-Waardenburg syndrome). Nat Genet 1996; 12:442.
53. McCallion AS, Chakravarti A. EDNRB/EDN3 and Hirschsprung disease type II. Pigment
Cell Res 2001; 14:161.

54. Pingault V, Bondurand N, Kuhlbrodt K, et al. SOX10 mutations in patients with


Waardenburg-Hirschsprung disease. Nat Genet 1998; 18:171.

55. Gutmann DH, Aylsworth A, Carey JC, et al. The diagnostic evaluation and
multidisciplinary management of neurofibromatosis 1 and neurofibromatosis 2. JAMA
1997; 278:51.

56. Hurwitz S. Clinical Pediatric Dermatology, 2nd ed, WB Saunders, Philadelphia 1993.
57. Lim JY, Kim H, Kim YH, et al. Merlin suppresses the SRE-dependent transcription by
inhibiting the activation of Ras-ERK pathway. Biochem Biophys Res Commun 2003;
302:238.
58. Kwiatkowski DJ, Short MP. Tuberous sclerosis. Arch Dermatol 1994; 130:348.
59. Webb DW, Clarke A, Fryer A, Osborne JP. The cutaneous features of tuberous sclerosis:
a population study. Br J Dermatol 1996; 135:1.
60. Ringpfeil F, McGuigan K, Fuchsel L, et al. Pseudoxanthoma elasticum is a recessive
disease characterized by compound heterozygosity. J Invest Dermatol 2006; 126:782.
61. Le Saux O, Urban Z, Tschuch C, et al. Mutations in a gene encoding an ABC
transporter cause pseudoxanthoma elasticum. Nat Genet 2000; 25:223.
62. Ringpfeil F, Lebwohl MG, Christiano AM, Uitto J. Pseudoxanthoma elasticum:
mutations in the MRP6 gene encoding a transmembrane ATP-binding cassette (ABC)
transporter. Proc Natl Acad Sci U S A 2000; 97:6001.
63. Bergen AA, Plomp AS, Schuurman EJ, et al. Mutations in ABCC6 cause
pseudoxanthoma elasticum. Nat Genet 2000; 25:228.

64. Sherer DW, Sapadin AN, Lebwohl MG. Pseudoxanthoma elasticum: an update.
Dermatology 1999; 199:3.

65. Engelman MW, Fliegelman MT. Pseudoxanthoma elasticum. Cutis 1978; 21:837.
66. Laube S, Moss C. Pseudoxanthoma elasticum. Arch Dis Child 2005; 90:754.
67. Hardelin JP, Levilliers J, del Castillo I, et al. X chromosome-linked Kallmann syndrome:
stop mutations validate the candidate gene. Proc Natl Acad Sci U S A 1992; 89:8190.
68. Happle R. X-linked dominant chondrodysplasia punctata. Review of literature and
report of a case. Hum Genet 1979; 53:65.
69. Wright JT, Grange DK, Fete M. Hypohidrotic ectodermal dysplasia. In: GeneReviews, A
dam MP, Ardinger HH, Pagon RA, et al (Eds), University of Washington, Seattle, 1993.
Topic 3004 Version 34.0
GRAPHICS

Genodermatoses associated with malignancy

Inheritance/
Key clinical features
pathogenesis
Basal Cell Autosomal Multiple basal cell carcinomas. Other features include
Nevus dominant odontogenic cysts of the jaw, palmoplantar pits, bifid ribs and
Syndrome other skeletal abnormalities. Common CNS abnormalites
Mutation of the
(Nevoid Basal include calcification of the falx cerebri and bridging of the sella
tumor
Cell turcica. Extracutaneous tumors, including medulloblastomas,
suppressor
Carcinoma meningiomas, and ovarian fibrosarcomas, are also associated
gene patched
Syndrome, with the syndrome.
(PTC)
Gorlin
Syndrome)

Gardner Autosomal Triad of familial adenomatous polyposis, benign osteomas, and


Syndrome dominant skin and soft tissue tumors. Multiple epidermoid cysts are the
most characteristic skin finding. Desmoid tumors may also be
Mutation of
seen. Congenital hypertrophy of the retinal pigmented
tumor
epithelium is a reliable and early marker of disease. Colon
suppressor
cancer develops in all affected individuals requiring
gene APC
prophylactic colectomy.

Peutz-Jegher Autosomal Dark periorificial and acral freckling most markedly on the lips;
Syndrome dominant patients develop hamartomatous polyps of GI tract which may
give rise to cancer. Also associated with increased risk of early
Mutation of
adenocarcinomas of the breast, cervix, pancreas, uterus and
tumor
ovaries.
suppressor
gene STK11

Xeroderma Autosomal Increased photosensitivity, development of cutaneous


Pigmentosa recessive malignancies in early childhood. Associated ocular and
neurologic abnormalities.
Defective
excision repair
of UV-induced
DNA damage

Epidermolysis bullosa (EB) syndromes


Dowling Autosomal Presents at birth or in early infancy; bullae occur with mild
Meara EB dominant trauma, but lesions heal without scarring; mild or no mucosal
simplex involvement. Increased risk of basal cell carcinoma during mid-
Mutation of
adulthood.
keratin 5 or
keratin 14 gene

Herlitz Autosomal Onset at birth with widespread bullae and erosions with
junctional recessive nonhealing granulation tissue; absent nails, dysplastic teeth,
EB oral lesions.
Mutations in the
laminin alpha-3, Increased risk of squamous cell carcinoma beginning in
beta-3, or adolescence.
gamma-2 gene

Hallopeau- Autosomal Onset at birth; generalized bullae that heal with extensive
Siemens recessive scarring; repeated episodes may lead to contraction flexures at
recessive the knees and elbows and fusion of the digits; frequent
Mutation in the
dystrophic involvement of the mucous membranes.
collagen type VII
EB
alpha-1 gene Increased risk of malignant melanoma beginning in childhood
and squamous cell carcinoma beginning in adolescence.

Graphic 72060 Version 4.0


Palmar pits associated with nevoid basal cell
carcinoma syndrome

Palmar surface of hand showing 1 to 2 mm, sharply marginated,


depressed, red lesions (ie, palmar pits).

Reproduced with permission from: Fitzpatrick TB, Johnson RA, Wolff K, Suurmond D.
Color Atlas & Synopsis of Clinical Dermatology. Common & Serious Diseases. (4th Ed).
McGraw-Hill, New York 2001. pgs. 268 & 289. Copyright © 2001 McGraw-Hill.

Graphic 72687 Version 4.0


Oral lesions in Peutz-Jeghers syndrome

Photograph shows the characteristic circumoral pigmentation in a


patient with the Peutz-Jeghers syndrome. The pigmentation may not
be obvious as in this patient, and it should always be sought
carefully in young patients presenting with unexplained
gastrointestinal bleeding, particularly if there is a family history of
such bleeding.

Reprinted with permission from Pounder RE, Allison MC, Dhillon AP. A Colour Atlas of
the Digestive System, Wolfe, London 1989 p. 118.

Graphic 80815 Version 4.0


Cutaneous leiomyomatosis

(A) Clustered, pink cutaneous leiomyomatosis nodules on the central and upper back.

(B) Close-up image of nodules in panel A.

Courtesy of Edward W Cowen, MD, MHSc.

Graphic 100293 Version 2.0


Cutaneous leiomyomatosis

Generalized cutaneous leiomyomatosis. Hundreds of painful, small, red papules and nodules on
the leg.

Courtesy of Edward W Cowen, MD, MHSc.

Graphic 100296 Version 2.0


Cutaneous leiomyomatosis

Typical ovoid, pink, banal appearance of a single cutaneous leiomyomatosis lesion.

Courtesy of Edward W Cowen, MD, MHSc.

Graphic 100292 Version 2.0


Cutaneous leiomyomatosis

Pink/red, variably sized and shaped papules and nodules.

Courtesy of Edward W Cowen, MD, MHSc.

Graphic 100291 Version 2.0


The ichthyoses

Inheritance
and Key clinical features
pathogenesis
Icthyosis vulgaris Autosomal Relatively common (1:250 to 1:300). Fine,
dominant white translucent scales predominantly on
extensor surfaces of extremities with sparing
Fillagrin defect
of flexures, palms, soles, and face.
resulting in a
Hyperlinear palms. Often associated with
retention
keratosis pilaris and atopy including eczema.
hyperkeratoses
Onset during childhood. Typically improves
(Normal with age.
epithelial
turnover)

X-linked ichthyosis (steroid X-linked recessive 1:6000 men. Failure to progress in labor in
sulfatase deficiency) mother of affected fetus (placental sulfatase
Steroid sulfatase
deficiency). Large brown "dirty" adherent
deficiency results
scales with relative sparing of flexures,
in cholesterol
palms, soles, and face. Asymptomatic
sulfate
corneal opacities (50 percent of adult males).
accumulation
12 to 15 percent incidence of cryptorchidism
and a retention
and independently increased risk of
hyperkeratoses
testicular cancer. Onset at two to six weeks.
(Normal Gradually worsens with age.
epithelial
turnover)

Lamellar ichthyosis Autosomal 1:300,000 live births. "Collodion baby" at


recessive birth (translucent taut parchment-like
membrane encasing infant, ectropion,
Transglutaminase
eclabium, generalized erythroderma;
deficiency
complications include sepsis, protein loss,
(Increased and electrolyte abnormalities). Children and
epithelial adults develop large brown polygonal scales
turnover) involving entire body surface, with increased
involvement in flexural folds. Palmoplantar
keratoderma, scarring alopecia, nail
dystrophy, and hypohidrosis with heat
intolerance frequently seen. Ectropion is also
almost always present.

Congenital ichthyosiform Autosomal "Collodion baby" at birth (see above).


erythroderma recessive Children and adults develop generalized
erythroderma with fine white scaling.
(Increased
Palmoplantar keratoderma, scarring
epithelial
alopecia, and persistent ectropion may also
turnover)
be seen.

Epidermolytic hichtyosis Autosomal Neonatal period with widespread blistering


(epidermolytic hyperkeratosis dominant and erythema. Later infancy and adulthood
or bullous congenital characterized by generalized hyperkeratosis
50 percent due to
ichthyosiform erythroderma) with dark scales and spiny ridges especially
spontaneous
pronounced flexurally. Denuded areas of
mutations of
skin where scales shed with full thickness
keratins 1 and 10
stratum corneum also seen. Focal bullae due
(Increased to secondary infection may also develop.
epithelial
turnover)

Harlequin ichthyosis Autosomal Massive armor-like plates of scale with deep


recessive fissures encasing fetus. Severe ectropion,
eclabium, deformed or absent ears, nose,
(Increased
fingers, toes. Infants are born stillborn or
epithelial
generally die shortly after birth due to
turnover)
respiratory and feeding compromise due to
constriction.

Graphic 71157 Version 5.0


Pachyonychia congenita

Thickened, discolored nails are present in this patient with


pachyonychia congenita.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 52261 Version 5.0


Darier disease

Multiple yellow-brown, hyperkeratotic papules are present on the


chest.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 56168 Version 7.0


Darier disease

Multiple hyperpigmented, hyperkeratotic papules are on the chest.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 70000 Version 7.0


Darier disease

Distal notching and linear, red and white bands are present on the
nails.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 81865 Version 7.0


Epidermolysis bullosa

EB type Pathogenesis Key clinical features


EB simplex (most Autosomal dominant; most due to Typically presents at birth or early
common) mutations in genes for keratins 5 and infancy. Bullae with mild trauma, heal
14 expressed on epithelial cell without scarring. Most forms with
cytoskeleton mild or no mucosal involvement.
Chronic course but blistering tends to
Intraepidermal separation
decrease with age, usually normal
lifespan.

Junctional EB Autosomal recessive; mutation in More severe variant characterized by


genes encoding hemidesmosomal widespread bullae and erosions with
proteins such as laminin 5 resulting in nonhealing granulation tissue;
defective cell adhesion associated with absent nails,
dysplastic teeth, oral lesions, and
Intra-lamina lucida separation
pyloric stenosis. Death usually occurs
in early childhood due to malnutrition
or infection. Normal life expectancy in
patients with milder variant who
develop bullae, primarily on the
extremities, that may heal with
atrophic scarring.

Dystrophic EB Autosomal recessive and autosomal Onset at birth; generalized bullae,


dominant; type VII collagen mutation heal with extensive scarring, milia.
leading to defective anchoring fibrils Repeated episodes of severe
blistering and scarring lead to
Sub-basal lamina separation contraction flexures at knees and
elbows and fusion of digits ("mitten
deformities") of hands and feet. Oral,
GI, and ocular mucous membrane
involvement is also frequently seen
and can be severe. Risk of developing
squamous cell cancer within scars.
Patients often die in early adulthood
from complications such as infection
or severe malnutrition. Milder
phenotype seen with autosomal
dominant forms.

Kindler EB Autosomal recessive Blisters induced by mild trauma,


progressive poikiloderma, and skin
Separation at the level of the basal atrophy (particularly of the dorsal
keratinocytes, within the lamina hands and feet). Photosensitivity may
lucida, or sub-basal lamina separation or may not be present. Additional
features may include desquamative
gingivitis and strictures of the
esophagus, anus, vagina, and urethra.
Blistering and photosensitivity may
decrease with age.

EB: epidermolysis bullosa.

Graphic 76897 Version 8.0


Genotypic and phenotypic variants of epidermolysis bullosa simplex
(epidermolytic)

Site of Inheritance
cleavage Mutated gene
Subtypes Clinical features
within the Targeted gene
epidermis product

Suprabasal Acantholytic EBS Autosomal Five cases of lethal acantholytic


(EBS-acanth) recessive epidermolysis bullosa due to
(cytolysis of
DSP, JUP mutations in DSP and one due
suprabasal
Desmoplakin to JUP have been described [1-5]
keratinocytes;
intact blisters (absent), Generalized skin fragility with
usually not plakoglobin rapidly progressive
clinically evident; (absent) epidermolysis and large sheets
keratinocytes of detached skin with superficial
form numerous erosions at birth; skin
interdigitating detachment on hands and feet
cellular in a glove and stocking pattern;
protrusions; only complete disruption of the
few desmosomal epidermal barrier without intact
remnants blisters or vesicles; no excessive
present) granulation tissue
Universal alopecia (complete
absence of scalp hair, eyebrows,
eyelashes despite discrete
follicular openings on scalp),
nail loss, and neonatal teeth
Involvement of oropharynx,
gastrointestinal, genitourinary,
and respiratory tract with
extensive suprabasal
acantholytic separation
Demise in neonatal period [1]

Skin fragility Autosomal Generalized skin fragility with


syndromes recessive trauma-induced and
Desmoplakin DSP, JUP, PKP1 spontaneous superficial
deficiency Desmoplakin erosions from birth;
(EBS- (reduced), palmoplantar keratoderma with
desmoplakin; plakoglobin painful, often disabling fissures
skin fragility- (reduced), (walking, bearing weight); nail
woolly hair plakophilin-1 dystrophy; alopecia
syndrome) (absent or Chronic cheilitis, perioral scale,
Plakoglobin reduced) perioral and lingual fissures
deficiency Blepharitis, astigmatism
(EBS- Persistent abnormal hair
plakoglobin; (hypotrichosis, loss of
skin fragility- eyelashes, complete alopecia;
plakoglobin woolly hair)
deficiency) Esophageal strictures,
Plakophilin constipation
deficiency Growth retardation (usually
(EBS- below the third centile for
plakophilin; height and weight)
skin fragility-
Variable clinical features include
ectodermal
scattered scale-crust on the
dysplasia
trunk and limbs, follicular
syndrome)
hyperkeratosis, inflammatory
scaly plaques in flexures,
perianal erythema and erosions,
pruritus; reduced sweating;
dental caries; recurrent
systemic infections; chronic
diarrhea

EBS superficialis Unknown Blistering between stratum


corneum and granulosum
Superficial erosions with
scarring and milia formation
from birth or early infancy [6]

Acral peeling skin Autosomal Superficial painless skin peeling


syndrome [7] recessive (volar and dorsal aspects of
TGM5 hands, feet; elbows, knees)
Transglutaminase- Acral blisters, erosions at
5 birth/since infancy; pruritus
aggravated by heat, sweating,
humidity, mechanical trauma,
exposure to water
Spontaneous healing with
residual erythema, burning
sensation, pruritus, and
hyperpigmentation
No scarring or atrophy

Basal EBS, localized Autosomal Most common EBS subtype


(EBS-loc, former
(cytolysis of Weber-Cockayne) dominant Mainly limited, regional
basal KRT5, KRT14 involvement of palms and soles
keratinocytes) Keratin 5, keratin Clinical onset usually during
14 infancy or, rarely, early
adulthood
Extracutaneous manifestations
(other than clinically
insignificant blisters within oral
cavity during early childhood)
rare [8]

EBS, generalized Autosomal Generalized, peculiar


severe (former dominant herpetiform distribution
Dowling-Meara) KRT5, KRT14 (arcuate grouping) of blisters
Keratin 5, keratin from birth (typically and best
14 seen when patients have
relatively milder disease
activity [9] )
Atrophic scarring, milia
formation, confluent
palmoplantar hyperkeratosis,
nail manifestations
Often mucosal involvement

EBS, generalized Autosomal Widespread but less severe


intermediate dominant form that usually presents at
(includes former KRT5, KRT14, birth
Koebner) EXPH5, DYS, Rarely eye involvement
KLHL24
Keratin 5, keratin
14, exophilin 5,
dystonin, Kelch-
like family
member 24

EBS-MP (EBS with Autosomal Skin blistering with reticulate


mottled dominant brown skin pigmentation,
pigmentation) KRT5 keratoderma, nail dystrophy
Keratin 5 Pigmentary lesions usually
present in early childhood, but
may become less distinctive or
even imperceptible by adult
life [9]

EBS, migratory Autosomal Blistering lesions on hands,


circinate dominant legs, and feet from birth
KRT5 Annular migratory erythema
Keratin 5 with vesicles and crusts at the
advancing edge
Brown postinflammatory
hyperpigmentation
Improves in the first few years
of life

EBS with Autosomal Generalized blistering from


muscular recessive birth
dystrophy PLEC1 Atrophic scarring, milia
Plectin formation, nail dystrophy and
loss
Granulation tissue formation
and stenosis within respiratory
tract
Muscular dystrophy may either
be present on or shortly after
birth in a child with severe
generalized skin involvement
(usually appearing as "floppy"
babies who are unable to easily
lift their arms and legs), or, in
others, may arise insidiously in
later childhood or adulthood;
this variant harbors a higher
mortality rate from muscle
involvement [8]

EBS with pyloric Autosomal Gestational hydramnion


atresia recessive Generalized blistering of
PLEC1, ITGA6, variable severity from birth,
ITGB4 atrophic scarring [10-12]
Plectin, integrin- Association with aplasia cutis
alpha-6, integrin- congenita [13]
beta-4 Mucosal involvement (protein
losing enteropathy, diarrhea)
Enamel hypoplasia, caries
Anemia, growth retardation,
contractures, cryptorchidism,
high mortality

EBS, autosomal Autosomal Generalized blistering from


recessive K14 recessive birth (frequently anogenital),
KRT14 [14] ichthyosiform plaques; rarely
Keratin 14 milia formation or atrophic
scarring
Involvement of oral cavity
(caries) and genitourinary tract
Growth retardation,
constipation

EBS, Ogna type Autosomal Hemorrhagic blisters at


dominant predominantly acral sites from
PLEC1 birth
Plectin Onychogryphosis (markedly
curved and deformed nails
resembling ram's horns) [9]

EBS, autosomal Autosomal First described in three siblings


recessive- recessive born to first cousin parents [15]
exophilin 5 EXPH5 Disrupted keratinocyte
deficiency Exophilin-5 adhesion within the lower
(Rab27B GTPase epidermis with prominent skin
effector protein fragility at birth
Slac2-b) Generalized trauma-induced,
occasionally spontaneous scale,
partly hemorrhagic crusts and
intermittent skin
blistering/erosions; bruising
Healing with slightly atrophic
scars and mild pigmentary
mottling
No involvement of mucous
membranes, nails, and hair
Symptomatic improvement
within the first years of life with
occasional small blisters,
erosions, linear crusts at sites of
mechanical trauma as the main
feature

EBS, autosomal Autosomal Only one reported case [14]


recessive-BP230 recessive Generalized, lifelong but
deficiency DST/BPAG1-e relatively mild trauma-induced
Dystonin/coiled and spontaneous blistering
coil domain of (ankles, feet)
epithelial isoform Skin peeling; erosions,
of bullous hemorrhage
pemphigoid Nail dystrophy; normal hair
antigen-1 (BPAG1- growth
e) Healing with postinflammatory
hypo- and hyperpigmentation
No scarring, milia formation, or
mucosal blistering
Episodes of collapse; recurrent
bilateral headaches, transient
episodes of arm numbness and
weakness reported in index
patient may represent
manifestation of concomitant
neurologic diagnosis of
CADASIL syndrome

Recessive EBS Autosomal Twenty-six cases in three


due to stabilizing recessive reports from China, Israel, and
mutations in the KLHL-24 the United Kingdom
ubiquitin ligase Kelch-like family Quite marked birth traumas,
enzyme that member 24 especially the lower limbs
processes Early involvement of trunk and
KRT14 [16-18] arms
Heal with subtle atrophic
scarring
Nail defects
Oral ulceration
Not much dyspigmentation
Transient milia
Alopecia ±

EBS: epidermolysis bullosa simplex; CADASIL: cerebral autosomal dominant arteriopathy with
subcortical infarcts and leukoencephalopathy.

References: ​
1. Jonkman MF, Pasmooij AM, Pasmans SG, et al. Loss of desmoplakin tail causes lethal acantholytic Epidermolysis
bullosa. Am J HumGenet 2005; 77:653.
2. Hobbs RP, Han SY, van der Zwaag PA, et al. Insights from a desmoplakin mutation identified in lethal acantholytic
epidermolysis bullosa. J Invest Dermatol 2010; 130:2680.
3. Kim SJ, Ko JM, Shin SH, et al. Korean Monozygotic Twins with Lethal Acantholytic Epidermolysis Bullosa Caused by
Two Novel DSP Mutations. Ann Clin Lab Sci 2017; 47:213.
4. Bolling MC, Veenstra MJ, Jonkman MF, et al. 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 2010; 162:1388.
5. Pigors M, Kiritsi D, Krümpelmann S, et al. Lack of plakoglobin leads to lethal congenital epidermolysis bullosa: a
novel clinico-genetic entity. Hum Mol Genet 2011; 20:1811.
6. Fine JD, Johnson L, Wright T. Epidermolysis bullosa simplex superficialis. A new variant of epidermolysis bullosa
characterized by subcorneal skin cleavage mimicking peeling skin syndrome. Arch Dermatol 1989; 125:633.
7. Kiritsi D, Cosgarea I, Franzke CW, et al. Acral peeling skin syndrome with TGM5 gene mutations may resemble
epidermolysis bullosa simplex in young individuals. J Invest Dermatol 2010; 130:1741.
8. Lanschuetzer CM, Fine JD. Classification and molecular basis of hereditary epidermolysis. In: Life with Epidermolysis
Bullosa (EB): Etiology, diagnosis, multidisciplinary care and therapy, Fine JD, Hintner H (Eds), Springer, Vienna, New
York 2008. p.6.
9. Fine JD. General cutaneous manifestations. In: Life with Epidermolysis Bullosa (EB): Etiology, diagnosis,
multidisciplinary care and therapy, Fine JD, Hintner H (Eds), Springer, Vienna, New York 2008. p.99.
10. Mellerio JE, Pulkkinen L, McMillan JR, et al. Pyloric atresia-junctional epidermolysis bullosa syndrome: Mutations in
the integrin beta4 gene (ITGB4) in two unrelated patients with mild disease. Br J Dermatol 1998; 139:862.
11. Nakano A, Pulkkinen L, Murrell D, et al. Epidermolysis bullosa with congenital pyloric atresia: novel mutations in the
beta 4 integrin gene (ITGB4) and genotype/phenotype correlations. Pediatr Res 2001; 49:618.
12. Pulkkinen L, Kim DU, Uitto J. Epidermolysis bullosa with pyloric atresia: Novel mutations in the beta4 integrin gene
(ITGB4). Am J Pathol 1998; 152:157.
13. Maman E, Maor E, Kachko L, Carmi R. Epidermolysis bullosa, pyloric atresia, aplasia cutis congenita:
Histopathological delineation of an autosomal recessive disease. Am J Med Genet 1998; 78:127.
14. Groves RW, Liu L, Dopping-Hepenstal PJ, et al. A homozygous nonsense mutation within the dystonin gene coding
for the coiled-coil domain of the epithelial isoform of BPAG1 underlies a new subtype of autosomal recessive
epidermolysis bullosa simplex. J Invest Dermatol 2010; 130:1551.
15. McGrath JA, Stone KL, Begum R, et al. Germline Mutation in EXPH5 Implicates the Rab27B Effector Protein Slac2-b in
Inherited Skin Fragility. Am J Hum Genet 2012; 91:1115.
16. He Y, Maier K, Leppert J, et al. Monoallelic mutations in the translation initiation codon of KLHL24 cause skin
fragility. Am J Hum Genet 2016; 99:1395.
17. Lin Z, Li S, Feng C, et al. Stabilizing mutations of KLHL24 ubiquitin ligase cause loss of keratin 14 and human skin
fragility. Nat Genet 2016; 48:1508.
18. Lee JYW, Liu L, Hsu CK, et al. Mutations in KLHL24 Add to the Molecular Heterogeneity of Epidermolysis Bullosa
Simplex. J Invest Dermatol 2017; 137:1378.

Graphic 77709 Version 10.0


Genotypic and phenotypic variants of junctional epidermolysis bullosa
(lucidolytic)

Inheritance

Mutated
gene
Subtype Clinical features
Targeted
gene
product

JEB, generalized Autosomal Varying degrees of mechanical fragility at birth with


severe recessive generalized, recurrent, often persistent blistering, erosions,
(formerly Herlitz) and crusting covering not only particularly exposed skin
LAMA3, LAMB3,
areas (like palms and soles) but most or almost all of the
LAMC2
body surface
Laminin-
Periungual inflammation, onychodystrophy, and nail loss
alpha-3 chain
Exuberant granulation tissue, often periorificial
of laminin-332
Involvement of oral cavity (blisters, erosions); teeth (enamel
Laminin-beta- hypoplasia, excessive caries); gastrointestinal (protein-
3 chain of losing enteropathy, diarrhea), respiratory, and genitourinary
laminin-332 tract; conjunctivae
Laminin-
Anemia, growth retardation, high lethality (survival past one
gamma-2
year of age uncommon)
chain of
laminin-332

JEB, generalized Autosomal Generalized blistering from birth, atrophic scarring, milia;
intermediate recessive "male-pattern" alopecia; nail dystrophy and loss
(formerly non- Involvement of oral cavity and teeth (excessive caries, teeth
COL17A1,
Herlitz) loss, enamel hypoplasia); gastrointestinal, respiratory, and
LAMA3, LAMB3,
LAMC2 genitourinary tract; conjunctivae
Anemia, growth retardation, protein losing
Type XVII
enteropathy, diarrhea
collagen
Laminin-
alpha-3 chain
of laminin-332
Laminin-beta-
3 chain of
laminin-332
Laminin-
gamma-2
chain of
laminin-332

JEB, localized Autosomal Localized disease of lesser extent and lesser severity, onset
recessive at birth; milia, nail dystrophy, and loss

COL17A1, Involvement of oral cavity and teeth (excessive caries,


ITGB4, LAMA3, enamel hypoplasia)
LAMB3, LAMC2 Extensive atrophic scarring, hair loss, anemia, impairments
of growth and development, ocular abnormalities or
Type XVII
alterations affecting gastrointestinal, genitourinary,
collagen
and respiratory tract mostly absent [1]
Integrin-beta-
4

Laminin-
alpha-3 chain
of laminin-332

Laminin-beta-
3 chain of
laminin-332

Laminin-
gamma-2
chain of
laminin-332

JEB with pyloric Autosomal Gestational hydramnion


atresia recessive Generalized and profound blistering from birth, occasionally
ITGA6, ITGB4 age-associated amelioration; atrophic scarring; nail
dystrophy and loss
Integrin-
Association with aplasia cutis congenita
alpha-6
Enamel hypoplasia
Integrin-beta-
Pyloric atresia
4
Congenital genitourinary malformations
Sporadically rudimentary ear anlage

JEB, inversa Autosomal Blistering predominantly located in intertriginous


recessive areas/sites, typically displaying an overall extent that
exceeds that observed in patients with JEB, localized [1]
LAMA3, LAMB3,
LAMC2 Atrophic scarring, milia, hypo-/hyperpigmentation,
dystrophic (or absent) nails
Laminin-
Moderate oral mucosal lesions; dental enamel hypoplasia,
alpha-3 chain
caries and gastrointestinal abnormalities to a lesser extent
of laminin-332
as compared with other JEB variants
Laminin-beta-
3 chain of
laminin-332
Laminin-
gamma-2
chain of
laminin-332

JEB, late onset Autosomal Mild form of JEB developing in young adulthood or later
recessive Dystrophic (or absent) nails, dental enamel hypoplasia and
COL17A1 caries to a lesser severity than JEB, generalized intermediate
Hyperhidrosis
Type XVII
Absence of dermatoglyphs on fingers, palms, toes, and
collagen
soles

JEB-LOC Autosomal High incidence in Punjab


(laryngo-onycho- recessive Minimal skin blistering, erosions
cutaneous) Extensive aberrant production of granulation tissue with
LAMA3A
syndrome [2-11] delayed healing, atrophic scarring
Laminin-
Involvement of larynx (altered cry) at birth or later, skin
alpha-3a
(face, nuchal) and conjunctiva (granulomatous papules,
chain of
symblepharon) in infancy
laminin-332
Nail dystrophy and loss
Enamel hypoplasia, caries
High childhood mortality due to laryngeal, gastrointestinal,
and urethral strictures

JEB with Autosomal Congenital nephrotic syndrome (small kidneys, atrophic


respiratory and recessive glomeruli, focal segmental glomerulosclerosis, diffuse
renal interstitial fibrosis, tubular atrophy and loss)
ITGA3
involvement Interstitial lung disease (tachypnea, respiratory distress,
(JEB-RR) [10-12] Integrin- cyanosis in first days of life; secondary aspiration with
alpha-3 respiratory tract infections)
Mild skin fragility (small blisters and erosions beginning at
age of two to four months
Fine and sparse scalp hair, eyebrows, eyelashes
Nail dystrophy, distal onycholysis
Delayed reepithelialization; healing with residual erythema
No scarring; no mucosal involvement

JEB: junctional epidermolysis bullosa.

References:

1. Fine JD, Eady RA, Bauer EA, et al. The classification of inherited epidermolysis bullosa (EB): Report of the Third
International Consensus Meeting on Diagnosis and Classification of EB. J Am Acad Dermatol 2008; 58:931.
2. Shabbir G, Hassan M, Kazmi A. Laryngo-onycho-cutaneous syndrome: A study of 22 cases. Biomedica 1986; 2:15.
3. Ainsworth JR, Spencer AF, Dudgeon J, et al. Laryngeal and ocular granulation tissue formation in two Punjabi
children: LOGIC syndrome. Eye (Lond) 1991; 5:717.
4. Ainsworth JR, Shabbir G, Spencer AF, Cockburn F. Multisystem disorder of Punjabi children exhibiting spontaneous
dermal and submucosal granulation tissue formation: LOGIC syndrome. Clin Dysmorphol 1992; 1:3.
5. Phillips RJ, Atherton DJ, Gibbs ML, et al. Laryngo-onycho-cutaneous syndrome: An inherited epithelial defect. Arch
Dis Child 1994; 70:319.
6. Murrell DF, Hamil K, Pfendner E, et al. Is Laryngo-Onycho-Cutaneous Syndrome a form of junctional epidermolysis
bullosa? Oral paper. Australasian College of Dermatologists' Spring Meeting, Cairns, Australia, September 2005.
7. Figueira EC, Crotty A, Challinor CJ, et al. Granulation tissue in the eyelid margin and conjunctiva in junctional
epidermolysis bullosa with features of laryngo-onycho-cutaneous syndrome. Clin Exp Ophthalmol 2007; 25:163.
8. Hamil K, Uitto J, Figueira EC, et al. Novel N-terminal mutation in LAMA3a isoform causing late-onset Herlitz
junctional epidermolysis bullosa with features of laryngo-onycho-cutaneous syndrome. Poster. Society for
Investigative Dermatology, Los Angeles, CA, May 2007.
9. Cohn HI, Murrell DF. Laryngo-onycho-cutaneous syndrome. Dermatol Clin 2010; 28:89.
10. Nicolaou N, Margadant C, Lilen MR, et al. Gain of glycosylation in integrin α3 causes lung disease and nephrotic
syndrome. J Clin Invest 2012; 122:4375.
11. Yalcin EG, He Y, Orhan D, et al. Crucial role of posttranslational modifications of integrin α3 in interstitial lung
disease and nephrotic syndrome. Hum Mol Genet 2015; 24:3679.
12. Has C, Spartà G, Kiritsi D, et al. Integrin α3 mutations with kidney, lung, and skin disease. N Engl J Med 2012;
366:1508.

Graphic 67202 Version 8.0


Genotypic and phenotypic variants of dystrophic epidermolysis bullosa [1-3]

Inheritance

Mutated
gene
Subtype Clinical features
Targeted
gene
product

Dominant DEB

Dominant DEB, Autosomal Generalized blistering from birth, atrophic scarring,


generalized dominant dystrophic (or absent) nails; alopecia

COL7A1 "Albopapuloid" lesions (small, grouped, hypopigmented


papules usually on the lower back)
Type VII
Involvement of oral cavity, gastrointestinal and, rarely,
collagen
genitourinary tract

Dominant DEB, Autosomal Predilection of blistering for hands and feet from early
acral dominant, infancy, milia, atrophic scarring, dystrophic (or absent)
autosomal nails
recessive

COL7A1

Type VII
collagen

Dominant DEB, Autosomal Pretibial blistering from birth or early infancy,


pretibial dominant, involvement of hands and feet, milia, atrophic scarring;
autosomal dystrophic nails
recessive Lichen planus-like skin lesions
COL7A1

Type VII
collagen

Dominant DEB, Autosomal Generalized or localized erosions, blisters, milia, and


pruriginosa dominant, atrophic scars from infancy or later; dystrophic nails
autosomal Pronounced pruritus
recessive

COL7A1
Type VII
collagen

Dominant DEB, Autosomal Nail dystrophy or loss at birth or infancy without skin
nails only dominant involvement

COL7A1

Type VII
collagen

Dominant DEB, Autosomal Generalized blistering from birth or early infancy that
bullous dominant, improves dramatically over time
dermolysis of autosomal Milia, atrophic scarring; dystrophic nails
the newborn recessive

COL7A1

Type VII
collagen

Recessive DEB

Recessive DEB, Autosomal Generalized blistering from birth, atrophic scarring,


generalized recessive milia; sparse hair with scarring alopecia; dystrophic or
severe (formerly absent nails
COL7A1
recessive DEB- Acral pseudosyndactyly and contractures
Hallopeau- Type VII
Involvement of oral cavity (blisters, erosions, scarring,
Siemens) collagen
excessive caries, smooth tongue without papillae),
gastrointestinal (protein losing enteropathy) and
genitourinary tract (glomerulonephritis, IgA
nephropathy, chronic renal failure), conjunctivae
Tremendous risk for development of squamous cell
carcinomas
Cardiomyopathy, osteoporosis, anemia, impaired
growth, delayed puberty
Shortened life expectancy

Recessive DEB, Autosomal Generalized blistering at birth, continue to develop


generalized recessive throughout life; atrophic scarring, milia; nail dystrophy;
intermediate scarring alopecia
COL7A1
Pseudosyndactyly
Type VII
Increased risk for development of squamous cell
collagen
carcinomas
Involvement of oral cavity, gastrointestinal and
genitourinary tract, conjunctivae
Anemia, growth retardation
Recessive DEB, Autosomal Blistering predominantly located in intertriginous
inversa recessive lumbosacral, acral, and axial areas; milia, atrophic
scarring; nail dystrophy;
COL7A1
Involvement of oral cavity (caries), gastrointestinal and
Type VII
genitourinary tract
collagen
Stenosis of meatus acusticus externus
Anemia, growth retardation

Recessive DEB, Autosomal Pretibial blistering at birth or early infancy, involvement


pretibial recessive of hands and feet; milia; atrophic scarring; nail dystrophy

COL7A1 Lichen planus-like skin lesions

Type VII
collagen

Recessive DEB, Autosomal Blistering on hands and feet from early infancy, milia,
localized recessive atrophic scarring; nail dystrophy

COL7A1

Type VII
collagen

Recessive DEB, Autosomal Generalized or localized erosions, blisters, milia, and


pruriginosa recessive atrophic scars from infancy; dystrophic nails

COL7A1 Excessive pruritus

Type VII
collagen

Recessive DEB, Autosomal Pretibial and acral blistering at birth or from early
centripetalis recessive infancy, milia, atrophic scarring; nail dystrophy

COL7A1 Involvement of oral mucosa

Type VII
collagen

Recessive DEB, Autosomal Generalized blistering from birth or early infancy that
bullous recessive improves dramatically over time
dermolysis of Milia, atrophic scarring; dystrophic nails
COL7A1
the newborn
Type VII
collagen

DEB: dystrophic epidermolysis bullosa; IgA: immunoglobulin A.


References:

1. Dang N, Murrell DF. Mutation analysis and characterization of COL7A1 mutations with dystrophic epidermolysis
bullosa. Exp Dermatol 2008; 17:553.
2. Kim J, Loh CH, Murrell DF. Epidermolysis bullosa pruriginosa triggered by scabies infestation. J Dermatol 2013;
40:562.
3. Fine JD, Bruckner-Tuderman L, Eady RA. Inherited epidermolysis bullosa: updated recommendations on diagnosis
and classification. J Am Acad Dermatol 2014; 70:1103.

Graphic 55262 Version 4.0


Genotypic and phenotypic characteristics of Kindler epidermolysis bullosa

Inheritance

Mutated gene
Site of cleavage Clinical features
Targeted gene
product

Intraepidermal, Autosomal Generalized blistering at birth, atrophic scarring,


junctional, or recessive skin fragility diminishes with advancing age;
sublamina densa poikiloderma
FERMT1 (KIND1)
Photosensitivity
Fermitin family
Pseudosyndactyly
homologue 1
Gingival hyperplasia, oral leukokeratosis, colitis,
(kindlin-1)
esophagitis
Involvement of genitourinary tract (genital
leukokeratosis)
Conjunctivitis, ectropium
Mental retardation
Bone malformations (mandibular abnormalities)
Increased risk of cutaneous squamous cell
carcinoma after age 30 years

Graphic 81536 Version 4.0


Genetic classification of oculocutaneous albinism

Type of
MIM # Gene Locus Encoding function Comments
albinism

OCA1* 606933 TYR 11q14- Tyrosinase, which OCA1A: No


11q21 catalyzes several steps melanin
OCA1A 203100
in melanogenesis OCA1B: Varying
OCA1B 606952
amounts of
melanin are
present

OCA2 203200 OCA2 15q11.2- Melanosomal Common in sub-


(previously 15q12 membrane protein Saharan Africa due
called P to 2.7 kb deletion
gene) Includes brown
and rufous OCA

OCA3 203290 TYRP1 9p23 Stabilizes tyrosinase Previously called


and regulates red or rufous OCA
eumelanin production Common in Africa

OCA4 696574 SLC45A2 5p13.2 Membrane transport Minimal to near


(previously protein normal melanin
called Phenotype similar
MATP and to OCA2
AIM1) Common in Japan

OCA5 615312 Unknown 4q24 Unknown Described in a


Pakistani family

OCA6 609802 SLC24A5 15q21.1 Melanosome Described in a


maturation Chinese family

OCA7 615179 C10orf11 10q22.2- Melanocyte Described in


10q22.3 differentiation families on the
Faroe Islands

OCA8 Not DCT/TYRP2 13q32.1 Enzyme catalyst in Described in a


available melanogenesis French girl and a
North African
woman

OCA: oculocutaneous albinism.


* OCA1 includes previously described temperature-sensitive OCA, minimal pigment OCA, and
yellow OCA.

Original figure modified for this publication. From: Summers CG. Albinism. In: Taylor and Hoyt's Pediatric Ophthalmology
and Strabismus, 5th ed, Lambert SR, Lyons CJ (Eds), Elsevier, Atlanta 2017. Table used with the permission of Elsevier Inc.
All rights reserved.

Graphic 115427 Version 2.0


Piebaldism

Patchy areas of depigmentation in a child with piebaldism.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 58857 Version 5.0


Piebaldism

A congenital depigmented patch with white hair is present on the


scalp and forehead of this man with piebaldism. The patient also
had depigmented patches on the trunk and extremities. Multiple
family members had similar lesions.

Copyright © Bernard Cohen, MD, Dermatlas; http://www.dermatlas.org.

Graphic 52591 Version 7.0


Piebaldism

Multiple depigmented patches are present on the trunk and legs in


this patient with piebaldism.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 71514 Version 5.0


Waardenburg syndrome

A white forelock, broad nasal root, and dystopia canthorum in the


parent, and heterochromatic irides in the child affected by
Waardenburg syndrome.

Reproduced with permission from: Nayak CS, Isaacson G. Worldwide distribution of


Waardenburg syndrome. Ann Otol Rhinol Laryngol 2003; 112:817. Copyright © 2003
Annals Publishing Company.

Graphic 56776 Version 7.0


Neurofibromata and café-au-lait macules in a
patient with neurofibromatosis 1 (NF1)

Skin-colored and pink-tan, soft papules and nodules on the back are
neurofibromata. These lesions first appeared during late childhood.
The large, soft, ill-defined, subcutaneous nodule on the right lower
back is a plexiform neuroma. The café-au-lait macules appeared
earlier in childhood. A large one is visible on the middle lower back.

Reproduced with permission from: Fitzpatrick TB, Johnson RA, Wolff K, Suurmond D
(Eds). Color Atlas and Synopsis of Clinical Dermatology, 3 rd ed, McGraw-Hill, New
York. p.461. Copyright © 1997 The McGraw-Hill Companies.

Graphic 69606 Version 4.0


Multiple neurofibromas in an adult patient with
neurofibromatosis type 1

Multiple papules and nodules consistent with neurofibromas are


present on the trunk.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 82613 Version 5.0


Axillary freckling in neurofibromatosis type 1

Multiple hyperpigmented macules are present in the axilla.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 67532 Version 5.0


Tuberous sclerosis complex

Hypomelanotic macule on the torso of a patient with tuberous


sclerosis complex.

Graphic 61601 Version 8.0


Tuberous sclerosis complex

Angiofibromas of the face presenting as erythematous papules in a


child with tuberous sclerosis complex.

Graphic 54868 Version 5.0


Tuberous sclerosis complex

Typical forehead fibrous plaque in a child with tuberous sclerosis.

Graphic 78688 Version 3.0


Pseudoxanthoma elasticum

This woman with pseudoxanthoma elasticum has numerous yellow


asymptomatic papules on her neck and upper chest.

Copyright © Yahia Albaili, DO, Dermatlas; http://www.dermatlas.org.

Graphic 80952 Version 7.0


Pseudoxanthoma elasticum

Symmetric, confluent, yellow papules and redundant skin folds are


present in this 38-year-old woman. The characteristic plucked
chicken skin changes on the neck of patients with pseudoxanthoma
elasticum are seen. An eye examination showed the characteristic
angioid streaks.

Copyright © Rakhesh SV, MD, Dermatlas; http://www.dermatlas.org.

Graphic 59135 Version 7.0


Pseudoxanthoma elasticum

Yellow papules are present on the inner aspect of the lower lip.
Infiltration of the buccal mucosa of the lip is the most common
mucous membrane involvement in pseudoxanthoma elasticum.

Copyright © Franziska Ringpfeil, MD, Dermatlas; http://www.dermatlas.org.

Graphic 70421 Version 7.0


Pseudoxanthoma elasticum

Numerous confluent, yellow papules are present on the neck of this


patient with pseudoxanthoma elasticum.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 76225 Version 5.0


Angioid streaks

Typical angioid streaks in fundus of patient with pseudoxanthoma


elasticum.

Reproduced with permission from: Gold DH, Weingeist TA. Color Atlas of the Eye in
Systemic Disease. Lippincott Williams & Wilkins, Baltimore 2001. Copyright © 2001
Lippincott Williams & Wilkins.

Graphic 77863 Version 2.0


Classification of ectodermal dysplasias

OMIM,
Ectodermal dysplasia (ED) whenever Inheritance
available

Subgroup hair-teeth-nails-sweat glands (n = 38)


Alopecia-contractures-dwarfism mental retardation 203550 AR
syndrome

Ankyloblepharon-ectodermal defects-cleft lip/palate 106260 AD


syndrome (AEC syndrome; Hay-Wells syndrome); Rapp-
Hodgkin syndrome (129400) included

Anonychia with flexural pigmentation 106750 AD

Acrorenal field defect, ED, and lipoatrophic diabetes 207780 AR


(AREDYLD)

Arthrogryposis and ED 601701 AR

Camarena syndrome [1] – AD?; XD?

Cleft lip/palate-ED syndrome (CLPED1 syndrome; 225060 AR


Zlotogora-Ogur syndrome; Margarita Island syndrome)

Curly hair-acral keratoderma-caries syndrome [2] – AD

Dyskeratosis congenita, AD (dyskeratosis congenita, 127550 AD


Scoggins type)

Dyskeratosis congenita, AR 224230 AR

Dyskeratosis congenita, X-linked (Zinsser-Cole-Engman 305000 XR


syndrome)

Ectrodactyly, ED, and cleft lip/palate syndrome (EEC1 129900 AD


syndrome)

Ectrodactyly, ED, and cleft lip/palate syndrome 3 (EEC3 604292 AD


syndrome)

ED hypohidrotic, with acanthosis nigricans (Lelis 608290 ?


syndrome)

ED-syndactyly syndrome 2 (EDSS2) 613576 AR

ED with natal teeth, Turnpenny type 601345 AD

ED, Caratinga type [3] – AD?; XD?

ED, hypohidrotic, with hypothyroidism and agenesis of 225040 AD?; AR?; XD?
the corpus callosum
Focal dermal hypoplasia (FDH) 305600 XD

Hypohidrotic ED, AD (ADHED) 129490 AD

Hypohidrotic ED, AR (ARHED) 224900 AR

Hypohidrotic ED, X-linked (XLHED; Christ-Siemens- 305100 XR


Touraine [CST] syndrome)

Hypohidrotic ED with immune deficiency 300291 XD

Hypohidrotic ED with immunodeficiency, osteopetrosis, 300301 XD


and lymphedema (OLEDAID syndrome)

Hypomelanosis of Ito (HMI, incontinentia pigmenti type 300337 XD


I [IP1])

Keratitis-ichthyosis-deafness syndrome, AD (KID 148210 AD


syndrome, AD)

Keratitis-ichthyosis-deafness syndrome, AR (KID 242150 AR


syndrome, AR)

Naegeli syndrome (Naegeli-Franceschetti-Jadassohn 161000 AD


syndrome [NFJS])

Odonto-onychodermal dysplasia (OODD); Schöpf- 257980 AR


Schulz-Passarge syndrome (224750) included

Odontotrichomelic syndrome (tetramelic deficiencies, 273400 AR


ED, deformed ears, and other abnormalities)

Pachyonychia congenita, type 1 (PC1) 167200 AD

Pachyonychia congenita, type 2 (PC2) 167210 AD

Papillon-Lefevre syndrome 245000 AR

Rosselli-Gulienetti syndrome 225000 AR

Scalp-ear-nipple syndrome (Finlay-Marks syndrome; ED 181270; 129550 AD


with adrenal cyst)

Tricho-odonto-onychodysplasia with pili torti [1] – AD?; XD?

Tricho-onycho-dental dysplasia (TOD) [4] – AD

Xeroderma-talipes-enamel defects (XTE) [5] – AR

Subgroup hair-teeth-nails (n = 34)


Ackerman syndrome 200970 AR

ADULT syndrome 103285 AD

Arthrogryposis, ED, cleft lip/palate, and developmental 301815 XR


delay

Cardiofaciocutaneous syndrome (CFC syndrome) 115150 AD

Cardiomyopathy, dilated, with woolly hair, and 605676 AR


keratoderma (Carvajal syndrome)

Clouston syndrome (ED, hidrotic, AD) 129500 AD

Coffin-Siris syndrome 135900 AD?; AR?; XD?

Costello syndrome 218040 AR

Cranioectodermal dysplasia (CED1; Sensenbrenner 218330 AR


syndrome); CED2 (613610), CED3 (614099), and CED4
(614378) included

Dermoodontodysplasia 125640 AD

Dolichocephaly, dental defects, trichodysplasia [1] – AD

ED-syndactyly syndrome 1 (EDSS1); ED with pillous 613573 AR


anomaly and syndactyly [6] included

ED, trichoodontoonychial type 129510 AD

Ellis-van Creveld syndrome (EVC) 225500 AR

Growth retardation, alopecia, pseudoanodontia, and 230740 AR


optic atrophy syndrome (GAPO syndrome)

GOMBO syndrome 233270 AR

Hidrotic ED, AR (Fried's tooth and nail syndrome) 602401 AR

Hypotrichosis with pili bifurcate [7] – AR?

Incontinentia pigmenti (IP2) 308300 XD

Oculotrichodysplasia (OTD) 257960 AR

Odonto-onychodysplasia-alopecia [8] – AR

Odontotrichoungual-digital-palmar syndrome 601957 AD?; XD?

Pineal hyperplasia, insulin-resistant diabetes mellitus, 262190 AR


and somatic abnormalities

Rothmund-Thomson syndrome (RTS syndrome) 268400 AR

Schinzel-Giedion midface-retraction syndrome 269150 AR?; AD?

Sener syndrome 606156 ?

Split-hand/foot malformation (SHFM1); SHFM3 183600 AD


(246560), SHFM4 (605289), and SHFM5 (606708)
included
Thumb deformity and alopecia 188150 AD

Trichodentoosseus syndrome (TDO) 190320 AD

Tricho-dermodysplasia-dental defects [9] – AD?; XD?

Trichoodontoonychial dysplasia with bone deficiency 275450 AR?

Trichorhinophalangeal syndrome, type I (TRPS1) 190350 AD

Trichothiodystrophy, photosensitive (TTDP) 601675 AR

Witkop syndrome 189500 AD

Subgroup hair-teeth-sweat glands (n = 8)


Böök syndrome 112300 AD

Cleft lip/palate, ED, acral anomalies [10] – AR

Hypohidrotic ED with focal sweating [11] – AR?; XR?

Ichthyosis follicularis, atrichia, and photophobia 308205 XR


syndrome with or without bresheck syndrome (IFAP
syndrome)

Johnson neuroectodermal syndrome 147770 AD

Lenz-Passarge dysplasia [12] – XD

Leukomelanoderma, infantilism, mental retardation, 246500 AR


hypodontia, hypotrichosis

Ulnar-mammary syndrome (UMS) 181450 AD

Subgroup hair-nails-sweat glands (n = 4)


Alopecia-skin atrophy-anonychia-tongue defects [13] – ?

ED, hypohidrotic, with hypothyroidism and ciliary 225050 AR


dyskinesia (HEDH syndrome)

ED/skin fragility syndrome 604536 AR

Fischer-Volavsek syndrome [14] – AD

Subgroup teeth-nails-sweat glands (n = 2)


Ameloonychohypohidrotic syndrome 104570 AD

Limb-mammary syndrome (LMS) 603543 AD

Subgroup hair-teeth (n = 29)


Barber-Say syndrome 209885 AR?; AD?; XD?

Blepharocheilodontic syndrome 119580 AD


Brachymetapody-anodontia-hypotrichosis-albinoidism 211370 AR

Cataract, hypertrichosis, mental retardation syndrome 211770 AR


(CAHMR syndrome)

Cerebellar ataxia and ED 212835 AR

Cleft lip/palate-oligodontia-syndactyly-hair defects [15] – AD?; XD?

Coloboma, congenital heart disease, ichthyosiform 280000 AR


dermatosis, mental retardation, and ear anomalies
syndrome (CHIME syndrome; Zunich neuroectodermal
syndrome)

Congenital atrichia, palmoplantar hyperkeratosis, – AR?


mental retardation, and early loss of teeth [16]

Dubowitz syndrome 223370 AR

ED and neurosensory deafness 224800 AR

ED, Cape Verde [17] – AR

ED, ectrodactyly, and macular dystrophy syndrome (EEM 225280 AR


syndrome); hypotrichosis, congenital, with juvenile
macular dystrophy (HJMD, 601553) included

Gorlin-Chaudhry-Moss syndrome 233500 AR

Hallermann-Streiff syndrome (HSS) 234100 AR

Hypertrichosis terminalis, generalized, with or without 135400 AD


gingival hyperplasia (gingival fibromatosis with
hypertrichosis)

Hypertrichosis universalis 145700 AD

Johanson-Blizzard syndrome (JBS) 243800 AR

Mental retardation, hypotrichosis, and syndactyly [18] – AR?

Oculodentoosseous dysplasia, AR 257850 AR

Oculodentodigital dysplasia (ODDD) 164200 AD

Orofaciodigital syndrome I (OFD1 syndrome) 311200 XD

Pili torti, early onset 261900 AR

Pilodental dysplasia with refractive errors 262020 AR

Progeroid short stature with pigmented nevi (Mulvihill- 176690 AD


Smith syndrome)

Rodrigues blindness (microphthalmia, microcornea, and 268320 AR


sclerocornea with short stature and hair and dental
abnormalities)
Trichodental dysplasia 601453 AD

Trichodysplasia and amelogenesis imperfect [19] – AD?; XD?

Uncombable hair, retinal pigmentary dystrophy, dental 191482 AD


anomalies, and brachydactyly

Walbaum-Dehaene-Schlemmer syndrome [20] – AR

Subgroup hair-nails (n = 25)


Anonychia-onychodystrophy with hypoplasia or 106995 AD
absence of distal phalanges (Cooks syndrome)

AR neurodegenerative disorder with trichorrhexis – AR?


invaginata and ED [21]

Cartilage-hair hypoplasia (CHH) 250250 AR

Curly hair-ankyloblepharon-nail dysplasia syndrome 214350 AR


(CHANDS)

ED hidrotic, Christianson-Fourie type 601375 AD

ED with skin anomalies and mental retardation [22] – AR

ED, "pure" hair-nail type 602032 AD?

Hair-nail dysplasia [23] – AD

Hairy elbows (hypertrichosis cubiti) 139600 AD

Ichthyosis and male hypogonadism 308200 XR?

Ichthyosis with alopecia, eclabion, ectropion, and 242510 AR


mental retardation

Lymphedema-hypoparathyroidism syndrome 247410 AR?; XR?

Monilethrix 158000 AD

Onychotrichodysplasia and neutropenia 258360 AR

Palmoplantar keratoderma and congenital alopecia, AD 104100 AD


(alopecia congenita with keratosis palmoplantaris)

Pili torti and onychodysplasia [24] – AD

Pili torti, alopecia, and onychodysplasia [25] – AR

Polyposis, skin pigmentation, alopecia, and fingernail 175500 ?


changes

Popliteal pterygium syndrome, lethal type 263650 AR

Short stature, onychodysplasia, facial dysmorphism, – AR


and hypotrichosis syndrome (SOFT syndrome) [26]
Syndrome of accelerated skeletal maturation, failure to – AR?; XR?
thrive, and peculiar face (Marshall syndrome II) [27]

T cell immunodeficiency, congenital alopecia, and nail 601705 AR?


dystrophy

Trichomegaly with mental retardation, dwarfism, and 275400 AR


pigmentary degeneration of retina

Tricho-onychodysplasia-xeroderma [28] – AR

Trichothiodystrophy, nonphotosensitive 1 (TTDN1) 234050 AR

Subgroup hair-sweat glands (n = 4)


Dry skin and extranumerary areolae [29] – AD

Focal facial dermal dysplasia (Brauer syndrome); facial 136500 AD


ED (Setleis syndrome, 227260) included

Short stature-kidney insufficiency-ophthalmological – AR?; XR?


anomaly-growth retardation-ED (SKORED) [30]

Tricho-facio-hypohidrotic syndrome [31] – AR?; XR?

Subgroup teeth-nails (n = 14)

Corneodermatoosseous syndrome (CDO syndrome) 122440 AD

Deafness, congenital, and onychodystrophy, AD 124480 AD

Deafness, onychodystrophy, osteodystrophy, and 220500 AR?; AD?


mental retardation syndrome (DOOR syndrome)

Dermatoosteolysis, Kirghizian type 221810 AR

Haim-Munk syndrome (HMS) 245010 AR

Hearing loss, sensorineural, with enamel hypoplasia 234580 AR


and nail defects (Heimler syndrome)

Khan et al. chondroectodermal dysplasia [32] – AR

Lacrimoauriculodentodigital syndrome (LADD 149730 AD


syndrome)

Odontomicronychial dysplasia 601319 AR

Odonto-ungueal dysplasia [33] – AD

Otopalatodigital syndrome, type I (OPD1 syndrome) 311300 XD

Pycnodysostosis 265800 AR

Weyers acrofacial dysostosis 193530 AD

Williams-Beuren syndrome (WBS) 194050 AD


Subgroup teeth-sweat glands (n = 3)
Hypohidrotic ED with mydriasis, iris atrophy, and mental – AD?
retardation [34]

Kohlschutter-Tonz syndrome (epilepsy, dementia, and 226750 AR?; XR?


amelogenesis imperfecta)

Marshall syndrome I 154780 AD

Subgroup nail-sweat glands (n = 2)


Adermatoglyphia with congenital facial milia and acral 129200 AD
blisters, digital contractures, and nail abnormalities (ED,
absent dermatoglyphic pattern, changes in nails, and
simian crease)

Pachyonychia congenita, AR 260130 AR

NOTE: The bold text highlights information not listed in an earlier article. [35]

AR: autosomal recessive; AD: autosomal dominant; ?: unknown; XD: X-linked dominant; XR: X-linked
recessive; ADULT: acro-dermato-ungual-lacrimal-tooth; GOMBO: growth retardation, ocular
abnormalities, microcephaly, brachydactyly, and oligophrenia.

References: ​
1. Freire-Maia N, Pinherio M. Ectodermal Dysplasias: A Clinical and Genetic Study, Alan R Liss, New York 1984. p.251.
2. van Steensel MA, van der Hout AH. Lelis syndrome may be a manifestation of hypohidrotic ectodermal dysplasia.
Am J Med Genet 2009; 149A:1612.
3. Montebelo Filho A, Freire AR, Pinheiro M, Freire-Maia N. Odonto-ungueal dysplasia: An apparently new autosomal
dominant ectodermal dysplasia. Braz J Genet 1996; 19:162.
4. Koshiba H, Kimura O, Nakata M, Witkop CJ Jr. Clinical, genetic, and histologic features of the trichoonychodental
[TOD] syndrome. Oral Surg 1978; 46:376.
5. Moynahan EJ. XTE syndrome (xeroderma, talipes and enamel defect): A new heredo-familial syndrome. Proc R Soc
Med 1970; 63:1.
6. Wiedemann HR, Grosse FR, Dibbern H. Características das síndromes em pediatria: Atlas de diagnostic diferencial,
Schattauer, São Paulo: Manole 1978.
7. Beemer FA, Bruynzell-Koomen C, Happle R. Two cases of hypotrichosis with pili bifurcati. Am J Med Genet 1987;
4:187.
8. Pinheiro M, Freire-Maia N. Ondoto-onicodisplasia com alopecia: Dois casos em uma irmandade. Ciênc Cult 1981;
33:696.
9. Pinheiro M, Freire-Maia DV, Miranda E, et al. Trichodermodysplasia with dental alternations: An apparently new
genetic ectodermal dysplasia of the tricho-odonto-onychial subgroup. Clin Genet 1986; 29:332.
10. Richieri-Costa A, Guion-Almeida ML, Freire-Maia N, Pinherio M. Autosomal recessive cleft lip/palate, ectodermal
dysplasia, and minor acral anomalies: Report of a Brazilian family. Am J Med Genet 1992; 44:158.
11. Gorlin RJ. Selected ectodermal dysplasias. In: Recent Advances in Ectodermal Dysplasias, Salinas CF, Optiz JM, Paul
NW (Eds), Alan R Liss, New York 1988. p.123.
12. Lenz W. Medical Genetics, University of Chicago Press, Chicago 1963. p. 214.
13. Sequeiros J, Sack GH. Linear skin atrophy, scarring alopecia, anonychia and tongue lesion: A "new" syndrome? Am J
Med Genet 1985; 21:669.
14. Fischer H. Familiar hereditäres vorkommen von keratoma palmare et plantare, nagelveränderungen,
haaranomalien und verdickung der endglieder der finger und zehen in 5 generationen. (Die beziehungen dieser
veränderungen zur inneren secretion). Dermatol Zeitschr 1921; 32:114.
15. Martínez BR, Monasterio LA, Pinheiro M, Freire-Maia N. Cleft lip/palate-oligodontia-syndactyly-hair alterations, a
new syndrome: Review of the conditions combining ectodermal dysplasia and cleft lip/palate. Am J Med Genet 1987;
27:23.
16. Steijlen PM, Neumann HA, Der-Kinderen DJ, et al. Congenital atrichia, palmoplantar hyperkeratosis, mental
retardation, and early loss of teeth in four siblings: A new syndrome? J Am Acad Dermatol 1994; 30:893.
17. Werninghaus K. Ectodermal dysplasia in Cape Verdian families. Arch Dermatol 1993; 129:515.
18. Lopes VL, Marques-de-Faria AP. Mental retardation, hypotrichosis and syndactyly: A new entity? Genet Couns 1996;
7:47.
19. Angelos G, Jorgenson RJ. Trichodysplasia and amelogenesis imperfecta. Oral Surg Oral Med Oral Pathol 1993; 75:86.
20. Walbaum R, Dehaene PH, Schlemmer H. Dysplasie ectodermique: Une forme autosomique récessive? Arch Fr Pédiatr
1971; 28:435.
21. Gyure KA, Kurczynski TW, Gunning W, French BN. Autosomal recessive neurodegenerative disorder with trichorrhexis
invaginata and ectodermal dysplasia. Pediatr Neurol 1992; 8:469.
22. Halal F, Setton N, Wang NS. A distinct type of hidrotic ectodermal dysplasia. Am J Med Genet 1991; 38:552.
23. Pinheiro M, Freire-Maia N. Hair-nail dysplasia – A new pure autosomal dominant ectodermal dysplasia. Clin Genet
1992; 41:296.
24. Beare JM. Congenital pilar defect showing features of pili torti. Br J Dermatol 1952; 64:366.
25. Calzavara-Pinton P, Carlino A, Benetti A, de Panfilis G. Pili torti and onychodysplasia. Report of previously
undescribed hidrotoic ectodermal dysplasia. Dermatology 1991; 182:184.
26. Sarig O, Nahum S, Rapaport D, et al. Short stature, onychodysplasia, facial dysmorphism, and hypotrichosis
syndrome is caused by a POC1A mutation. Am J Hum Genet 2012; 91:337.
27. Marshall RE, Graham CB, Scott CR, Smith DW. Syndrome of accelerated skeletal maturation and relative failure to
thrive: A newly recognized clinical growth disorder. J Pediatr 1971; 78:95.
28. Freire-Maia N, Pinheiro M, Fernandes-dos-Santos A. Trichoonychodysplasia with xeroderma, an apparently hitherto
undescribed pure ectodermal dysplasia. Braz J Genet 1985; 8:775.
29. Freire-Maia N, Chautard-Freire-Maia EA. Dry skin extranumerrary areolae. Am J Med Genet 1990; 35:141.
30. Greenstein MA, Poole A, Urbanski M, Saal HM. Ectodermal dysplasia: A new form and consideration of possible
associations. Am J Hum Genet 1985; 37:56.
31. Antely RM, Shields ED, Rosenberg GL, Bixler D. Hypohidrosis with sparse hair, short stature and normal teeth and
nails. Birth Defects Orig Artic Ser 1976; 12:136.
32. Khan B, Basit S, Touseef M, et al. A novel chondroectodermal dysplasia mapped to chromosome 2q24.1-q31.1. Eur J
Med Genet 2012; 55:455.
33. Pinheiro M, Freire-Maia N. Odonto-ungueal dysplasia: An apparently new autosomal dominant ectodermal
dysplasia. Braz J Genet 1996; 19:633.
34. Beyer P, Grosshans E, Vetter JM, et al. Forme inhabituelle de dysplasia ectodermique hypohidrotique avec dês
glandes sudoripares em nombre apparemment normal, mais dysplasiques et dês anomalies morphologiques de la
peau. Pediatrie 1979; 34:341.
35. Visinoni AF, Lisboa-Costa T, Pagnan NA, Chautard-Freire-Maia EA. Ectodermal dysplasias: Clinical and molecular
review. Am J Med Genet A 2009; 149A:1980.
From: Pagnan NA, Visinon ÁF. Update on ectodermal dysplasias clinical classification. Am J Med Genet A 2014;
164A(10):2415-23. https://onlinelibrary.wiley.com/doi/abs/10.1002/ajmg.a.36616. Copyright © 2014 Wiley Periodicals, Inc.
Reproduced with permission of John Wiley & Sons Inc. This image has been provided by or is owned by Wiley. Further
permission is needed before it can be downloaded to PowerPoint, printed, shared or emailed. Please contact Wiley's
permissions department either via email: permissions@wiley.com or use the RightsLink service by clicking on the 'Request
Permission' link accompanying this article on Wiley Online Library (http://onlinelibrary.wiley.com).

Graphic 117077 Version 1.0


Ichthyosis vulgaris

Skin with fine, white, adherent scale is present in this patient with
ichthyosis vulgaris.

Reproduced with permission from: Goodheart HP. Goodheart's Photoguide of


Common Skin Disorders, 2nd Edition, Lippincott Williams & Wilkins, Philadelphia
2003. Copyright © 2003 Lippincott Williams & Wilkins

Graphic 61258 Version 2.0


X-linked ichthyosis

X-linked ichthyosis: legs, infant.

Reproduced with permission from: Stedman's Medical Dictionary. Copyright ©2008


Lippincott Williams & Wilkins.

Graphic 66723 Version 1.0


Lamellar ichthyosis

Large, plate-like, diffuse scaling in a child with lamellar ichthyosis.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights reserved.

Graphic 128200 Version 1.0


Epidermolysis bullosa simplex

Bullae are present on the hands of this infant with epidermolysis


bullosa simplex.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 59364 Version 4.0


Epidermolysis bullosa simplex

Multiple erosions and bullae are present on the foot of this child
with epidermolysis bullosa simplex.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 73201 Version 5.0


Incontinentia pigmenti

Multiple erythematous papules, vesicles, and crusts are present in


linear streaks on the leg of this female infant.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 58769 Version 6.0


Focal dermal hypoplasia

Multiple erythematous areas representing dermal hypoplasia are


present. Note the digital abnormalities involving the foot.

Reproduced with permission from: www.visualdx.com. Copyright VisualDx. All rights


reserved.

Graphic 55714 Version 5.0


Contributor Disclosures
Teresa S Wright, MD, FAAD, FAAP No relevant financial relationship(s) with ineligible companies to
disclose. Helen V Firth, DM, FRCP, FMedSci No relevant financial relationship(s) with ineligible
companies to disclose. Jennifer L Hand, MD No relevant financial relationship(s) with ineligible
companies to disclose. Rosamaria Corona, MD, DSc No relevant financial relationship(s) with ineligible
companies to disclose.

Contributor disclosures are reviewed for conflicts of interest by the editorial group. When found, these
are addressed by vetting through a multi-level review process, and through requirements for
references to be provided to support the content. Appropriately referenced content is required of all
authors and must conform to UpToDate standards of evidence.

Política de conflito de interesses

Você também pode gostar