Open access

Current Genetics in Hair Diseases

Written By

Yutaka Shimomura

Submitted: 11 March 2012 Published: 06 February 2013

DOI: 10.5772/54004

From the Edited Volume

Current Genetics in Dermatology

Edited by Naoki Oiso

Chapter metrics overview

3,491 Chapter Downloads

View Full Metrics

1. Introduction

(HF) is a skin appendage which exists on the entire skin surface, except for palmoplantar and mucosal regions. During embryogenesis, HF development is operated through reciprocal interactions between skin epithelial cells and underlying dermal cells [1]. The first signal to induce HF formation is considered to originate from the dermal cells. The epithelial cells which receive the dermal signal lead to form a placode (Figure 1). Then a signal from the placode results in forming a dermal condensate just beneath the placode (Figure 1). Additional interaction between these structures induces the downgrowth of the placode and forms a hair germ, which is the source of epithelial components of the HF (Figure 2). The dermal condensate is gradually surrounded by the HF epithelium and becomes a dermal papilla. It has been shown that many signaling molecules, such as Wnt, ectodysplasin (Eda), bone morphogenic protein (Bmp), and sonic hedgehog (Shh), play crucial roles in the HF development [1]. After the HF is generated, it undergoes dynamic cell kinetics, known as the hair cycle, throughout postnatal life, which is composed of three phases: catagen (regressing) phase, telogen (resting) phase and anagen (growing) phase [2]. In human scalp HFs, duration of the catagen, telogen, and anagen phases are 1-2 weeks, 2-3 months, and 2-6 years, respectively. The hair cycle, which is an amazing ability of self-renewal, is maintained by the stem cell niche in bulge portion of the HF, as well as the dermal papilla [3, 4].

Figure 1.

Hair follicle placode (mouse embryo; E15.5)

Figure 2.

Hair germ (mouse embryo; E16.5)

Figure 3.

Human anagen hair follicle.

The anagen HF has a highly complex structure with several distinct cell layers (Figure 3). During the anagen phase, cells from the bulge portion migrate downward to matrix region, while making the outer root sheath (ORS). The matrix cells actively proliferate and differentiate into the hair shaft, the inner root sheath (IRS), and the companion layer of the HF (Figure 3) [4]. The hair shaft shares a common structural organization, in which a multicellular cortex is surrounded by a cuticular layer, occasionally with a medulla layer centrally located within the cortex. The hair shaft is strongly keratinized at the level of keratinizing zone, and forms a rigid structure (Figure 3). Growth of the hair shaft is molded and supported by the IRS, the companion layer, and the ORS. The IRS is composed of three distinct layers: the IRS cuticle, Huxley’s layer, and Henle’s layer (Figure 3).

Advertisement

2. Hair follicle

Recent advances in molecular genetics have led to the identification of numerous genes that are expressed in the HF. Furthermore, mutations in some of these genes have been shown to underlie hereditary hair diseases in humans [2]. Causative genes for the diseases encode various proteins with different functions, such as structural proteins, transcription factors, and signaling molecules. This chapter aims to update recent findings regarding the molecular basis of genetic hair diseases.

Advertisement

3. Keratin disorders

Keratins are one of the major structural components of the HF, and are largely divided into type I (acidic) and type II (neutral to basic) keratins. The type I and type II keratins undergo heterodimerization, which leads to form keratin intermediate filaments (KIFs) in the cytoplasm [5]. Based on the amino acid composition, keratins are further classified into two groups: epithelial (soft) keratins and hair (hard) keratins. As compared to the epithelial keratins, the hair keratins show higher sulfur content in their N- and C-terminus, which plays an important role in interacting with hair keratin-associated proteins via disulfide bindings [6, 7]. All the keratin proteins are composed of an N-terminal rod domain, a central rod domain, and a C-terminal tail domain. Importantly, the N-terminal and the C-terminal regions of the rod domain are highly conserved in amino acid sequences, which are called helix initiation motif (HIM) and helix termination motif (HTM), respectively (Figure 4). It is believed that the HIM and the HTM play essential roles in heterodimerization between the keratins. In humans, gene clusters for the type I and type II keratin genes are mapped on chromosomes 17q21 [8] and 12q13 [9], respectively. To date, a total of 54 functional keratin genes (28 type I and 26 type II) have been identified and characterized in humans. It has been shown that during differentiation of the HF, various keratin genes are abundantly and differentially expressed, and contribute to HF keratinization, leading to the formation of a rigid structure [10]. In general, epithelial keratins are mainly expressed in the ORS, the companion layer, the IRS, while hair keratins are predominantly expressed in the hair shaft. In addition, it has recently been reported that some epithelial keratins are expressed in the hair shaft medulla as well [11]. It is noteworthy that mutations in several keratin genes have been reported to underlie hereditary hair disorders in humans (Table 1).

Figure 4.

Structure of keratin proteins.

diseaseinheritance patternOMIM#main symptomsgeneprotein, function
MonilethrixAD158000moniliform hair, perifollicular papulesKRT81
KRT83
KRT86
K81 (basic hair keratin)
K83 (basic hair keratin)
K86 (basic hair keratin)
Pure hair and nail ectodermal dysplasiaAR602032hypotrichosis, spoon nailsKRT85K85 (basic hair keratin)
Autosomal dominant woolly hair (ADWH)/ hypotrichosisAD194300/613981WH/
hypotrichosis
KRT74
KRT71
K74 (basic epithelial keratin)
K71 (basic epithelial keratin)

Table 1.

Hereditary hair disorders caused by mutations in keratin genes. AD, autosomal dominant; AR, autosomal recessive.

Monilethrix is characterized clinically by fragile scalp hair shafts and diffuse perifollicular papules with erythema. As the hair of affected individuals with monilethrix is easily broken, they frequently show sparse hair (hypotrichosis). In most cases, monilethrix shows an autosomal dominant inheritance pattern (MIM 158000), while autosomal recessive forms (MIM 252200) also exist. Under microscopy, the hair shaft of affected individuals with monilethrix dysplays a characteristic anomaly, known as beaded or moniliform hair, which shows periodic changes in hair diameter. As a result, the hair leads to the formation of nodes and internodes (Figure 5) [12]. Autosomal dominant form of the disease is caused by heterozygous mutations in KRT81, KRT83, and KRT86 genes, which encode type II hair keratins K81, K83, and K86, respectively [13, 14]. All the mutations identified to date result in a deleterious amino acid substitution within either the HIM or the HTM of the rod domain. These hair keratins are predominantly expressed in the keratinizing zone of the hair shaft cortex (Figure 6) [15]. Although precise mechanisms to cause moniliform hair remain elusive, mutations in these hair keratin genes are predicted to result in disruption of the KIF formation, leading to an abnormal hair shaft keratinization.

Figure 5.

Moniliform hair. N, node; in, internode.

Figure 6.

Expression of hair keratin K86 in the human hair shaft cortex.

Pure hair and nail ectodermal dysplasia (PHNED; MIM 602032) is characterized by absent or sparse hair, as well as nail dystrophy [16]. Hairs of affected individuals with PHNED are short and thin, and perifollicular papules can also be observed. In addition, their nails typically show koilonychia (spoon nails). The disease can show either an autosomal dominant or recessive inheritance trait. The autosomal recessive form has been mapped to chromosome 17p12-q21.2 [17] and 12p11.1-q21.1 [18] which contain the type I and type II keratin gene clusters, respectively. Subsequently, homozygous mutations in KRT85 gene have been identified in families with autosomal recessive PHNED [18, 19]. The KRT85 gene encodes the type II hair keratin K85, which is abundantly expressed in the matrix region of both the HF and the nail units [15, 20]. Molecular basis for autosomal dominant PHNED is yet unknown.

In addition to hair keratins, it has recently been reported that mutations in HF-specific epithelial keratin genes are associated with hereditary woolly hair (WH)/hypotrichosis. WH is defined as an abnormal variant of tightly curled hair and is considered to be a kind of hair growth deficiency [21]. There are both syndromic and non-syndromic forms of WH. The non-syndromic forms of WH can show either an autosomal-dominant (ADWH; MIM 194300) or -recessive (ARWH; MIM 278150) inheritance pattern. It is well-known that WH is frequently associated with hypotrichosis. Recently, heterozygous mutations in KRT74 and KRT71 genes have been identified in families with ADWH/hypotrichosis (Figure 7) [22-24]. Importantly, the KRT74 and the KRT71 genes encode the IRS-specific type II epithelial keratins K74 and K71, respectively (Figure 8) [25]. It can be postulated that disruption of the KIF formation in the IRS results in a failure to guide the hair growth, and leads to WH phenotype. Interestingly, KRT71 mutations have also been identified in mice, rats, cats, and dogs, all of which show wavy coat phenotypes [26-30]. These data strongly suggest crucial roles of the IRS-specific epithelial keratins in the HF development and hair growth across mammalian species.

Figure 7.

Clinical features of autosomal dominant woolly hair/hypotrichosis caused by a mutation in the KRT71 gene.

Figure 8.

Expression of the IRS-specific karatin K71 in the human hair follicle.

Advertisement

4. Hereditary hair disorders resulting from disruption of cell-cell adhesion molecules

Similar to epidermis, the HF epithelium possess a number of cell-cell adhesion structures, such as desmosomes, corneodesmosomes, adherens junctions, gap junctions, and tight junctions, which play important roles in maintaining the structure and the function of the HF. It has been shown that disruption of any of these structures can result in hereditary hair disorders in humans (Table 2).

Desmosome is a critical structure for cell-cell adhesion in most epithelial tissues, including the HF. The major structural component of the desmosome is the desmosomal cadherin family, which is comprised of the desmogleins (DSGs) and desmocollins (DSCs). In humans, 4 DSG genes (DSG1-DSG4) and 3 DSC genes (DSC1-DSC3) are located on chromosome 18q12. These desmosomal cadherin family members are glycoproteins with single-pass transmembrane domain, and are involved in Ca2+-dependent cell-cell adhesion, connecting with each other using their extracellular domains [31]. Within the cytoplasm, they interact with several other proteins, known as desmosomal plaque proteins, which include plakoglobin, plakophilin, and desmoplakin. The desmosomal plaque proteins contribute to anchor the KIF near the cell membrane. As such, the cell integrity and the cell-cell adhesion are maintained [31]. Recessively-inherited mutations in the DSG4 gene have been shown to cause a non-syndromic form of hereditary hair disorder known as localized autosomal recessive hypotrichosis 1 (LAH1; MIM 607903) [32]. Affected individuals with LAH1 show sparse hairs on the scalp, chest, arms, and legs. The eyebrows and beard are less dense than normal, and the axillary hair, pubic hair, and eyelashes look normal in most cases. It is noteworthy that hair shafts of affected individuals with DSG4 mutations are fragile and often show moniliform hair [33-35]. Therefore, the DSG4 can also be regarded as a causative gene for autosomal recessive monilethrix. DSG4 is the only desmoglein member that is expressed in the hair shaft (Figure 9) [36], and its expression in the hair shaft cortex finely overlaps with K81, K83, and K86, of which mutations cause autosomal dominant monilethrix. More recently, a homozygous nonsense mutation in the DSC3 gene has been identified in a family with an autosomal recessive form of hypotrichosis [37]. The disease is characterized by sparse scalp hairs and small vesicle formation on the scalp and extremities (hypotrichosis and recurrent skin vesicles; MIM 613102) [37], while there is an argument that the vesicles may be keratosis pilaris [38]. In addition, mutations in genes encoding desmosomal plaque proteins can also show hair phenotypes (Table 2). For example, mutations in junctional plakoglobin (JUP) and desmoplakin (DSP) genes are known to underlie Naxos disease (MIM 601214) and Carvajal syndrome (MIM 605676), respectively [39, 40]. Both diseases show an autosomal recessive inheritance pattern and are characterized by woolly hair, palmoplantar keratoderma, and severe cardiomyopathy. Furthermore, loss of function mutations in plakophilin 1 (PKP1) gene cause a rare autosomal recessive disease named ectodermal dysplasia/skin fragility syndrome (MIM 604536) [41].

diseaseinheritance patternOMIM#main symptomsgeneprotein, function
Localized autosomal recessive hypotrichosis 1 (LAH1)/monilethrixAR607903/ 252200hypotrichosis, moniliform hair, perifollicular papulesDSG4desmoglein 4
Hypotrichosis and recurrent skin vesiclesAR613102Hypotrichosis, skin vesicles or keratosis pilarisDSC3desmocollin 3
Naxos diseaseAR601214WH, PPK, right ventricular cardiomyopathyJUPjunctional plakoglobin
Carvajal syndromeAR605676WH, PPK, left ventricular cardiomyopathyDSPdesmoplakin
Ectodermal dysplasia/skin fragility syndromeAR604536Hypotrichosis, fragile skin, nail dystrophyPKP1plakophilin 1
Hypotrichosis simplex of the scalp AD146520Scalp-limited hypotrichosisCDSNcorneodesmosin
Netherton syndromeAR256500ichthyosiform erythroderma, atopic manifestation, bamboo hairSPINK5LEKTI (serine protease inhibitor)
Ichthyosis with hypotrichosisAR610765ichthyosis, hypotrichosisST14matriptase (serine protease)
Hypotrichosis with juvenile macular dystrophyAR601553Hypotrichosis, weak eyesightCDH3P-cadherin
Ectodermal dysplasia, ectrodactyly, macular dystrophy (EEM) syndromeAR225280Hypotrichosis, weak eyesight, ectrodactylyCDH3P-cadherin
Hidrotic ectodermal dysplasia (Clouston syndrome)AD129500hypotrichosis, PPK, nail dystrophyGJB6connexin 30
Keratitis ichthyosis deafness (KID) syndromeAD148210vascularizing keratitis, sensorial deafness, erythrokeratoderma, hypotrichosisGJB2
GJB6
connexin 26
connexin 30
Ichthyosis, leukocyte vacuoles, alopecia, and sclerosing cholangitisAR607626Hypotrichosis, ichthyosis, jaundice, hapatomegaly, CLDN1claudin 1

Table 2.

Hereditary hair disorders caused by disruption of cell-cell adhesion structures and the related molecules.

Figure 9.

Expression of desmoglein 4 (DSG4) in the human hair shaft.

Corneodesmosome is a modified desmosome in the stratum corneum (SC) of the epidermis, and plays a crucial in the desquamation process. One of the major components of the corneodesmosome is corneodesmosin (CDSN). CDSN is secreted by cytoplasmic vesicles into the extracellular core of desmosomes, and is progressively proteolysed by several serine proteases, such as kallikrein-related peptidases, which leads to the loss of cell-cell adhesivity in the SC and causes desquamation [42]. CDSN is also expressed predominantly in the IRS of the HF, and thus is considered to be important for terminal differentiation, as well as subsequent degradation of the IRS [43]. In 2003, heterozygous nonsense mutations in the CDSN gene have been identified in patients with hereditary hypotrichosis simplex of the scalp (HHSS; MIM 146520), which is an autosomal dominant disorder characterized by sparse hairs limited to the scalp region without any obvious hair shaft anomalies (Figure 10) [44]. Histologically, the IRS of the patients’ HF was disturbed, which was consistent with the expression of CDSN in the IRS. Furthermore, aggregates of abnormal CDSN were detected around the HF, as well as in the papillary dermis in patients’ skin [44]. These aggregates have recently been shown to be an amyloid protein derived from the mutant CDSN, which is likely to be toxic to the HF cells [45]. Therefore, the mutant CDSN protein appears to function in a dominant negative manner, affect growth of the HF, and lead to HHSS. In addition to HHSS, it has been reported that mutations in other genes functionally related with CDSN can show some hair phenotypes associated with congenital ichthyosis. Of these, Netherton syndrome (NS; MIM 256500) is a rare autosomal recessive condition characterized by ichthyosiform erythroderma, atopic manifestation, and the hair shaft anomaly, known as bamboo hair (trichorrhexis invaginata) (Figure 11). The NS is caused by loss of function mutations in SPINK5 gene which encodes a serine protease inhibitor named LEKTI (lymphoepithelial Kazal-type-related inhibitor) [46]. Disruption of LEKTI has been shown to result in upregulation of serine proteases and excess desquamation due to premature proteolysis of CDSN [47, 48]. Furthermore, it has been reported that recessively-inherited mutations in ST14 gene, which encodes a member of serine proteases (matriptase), underlie ichthyosis with hypotrichosis syndrome (MIM 610765) [49]. Sum of these genetic data suggest that balanced expression of CDSN, serine proteases, and their inhibitors is critical for the HF differentiation.

Figure 10.

Clinical features of hypotrichosis simplex of the scalp.

Figure 11.

Bamboo hair (trichorrhexis invaginata).

E- and P-cadherins are classical cadherins which are a major component of adherens junctions in the HF. When the HF placode is formed during embryogenesis, the expression of E-cadherin is markedly downregulated, while P-cadherin is simultaneously upregulated, and prominant expression of P-cadherin persists in the proximal portion of the HF. This phenomenon, known as cadherin switching, is believed to be essential for the HF morphogenesis [50]. In addition, P-cadherin has recently been shown to be important for postnatal hair growth and cycling as well [51]. The critical role of these classical cadherins in the HF has been further supported by two hereditary diseases resulting from mutations in the P-cadherin gene (CDH3). First, mutations in the CDH3 gene are known to underlie hypotrichosis with juvenile macular dystrophy (HJMD; MIM 601553), which is an autosomal recessive disease characterized by sparse hair and weak eyesight due to macular dystrophy of the retina [52]. In addition, it has been reported that another disease, ectodermal dysplasia, ectrodactyly and macular dystrophy (EEM syndrome; MIM 225280), is also caused by recessively-inherited mutations in the CDH3 gene [53]. Affected individuals with EEM syndrome show common hair and eye phenotype with HJMD. However, EEM patients also shows split hand/foot malformation (ectrodactyly), suggesting crucial roles of P-cadherin in the development of not only hair and retina, but also the limbs in humans. There are no clear genotype-phenotype correlations in CDH3 mutations, as it has been reported that a same mutation in the CDH3 gene caused HJMD in one family [54], while EEM syndrome in another family [53]. Identification of modifier gene(s) may reveal this paradox in the future.

Gap junction (GJ) is a specialized intercellular structure that provides a pathway for both metabolic and ionic coupling between adjacent cells and maintains tissue homeostasis [55]. Connexins (Cxs) are 4-pass transmembrane proteins and the major component of the GJs. Clouston syndrome (MIM 129500), also known as hidrotic ectodermal dysplasia, is an autosomal dominant condition characterized by hypotrichosis, nail dystrophy, and palmoplantar keratoderma. The disease is caused by mutations in GJB6 gene which encodes Cx30 [56]. In addition, mutations in GJB2 gene encoding Cx26 are known to underlie keratitis-ichthyosis-deafness syndrome (KID; MIM 148210) [57]. The triad of KID is vascularizing keratitis, profound sensorial hearing loss, and erythrokeratoderma. Additionally, patients with KID show severe hypotrichosis in high frequency. Interestingly, it has been reported that a mutation in the GJB6 gene (V37E) can show phenotypes resembling KID [58]. These Cx proteins are mainly expressed in the ORS of the HF (Figure 12) [59, 60], and thus they may play some roles in maintaining the function of the HF stem cells.

Figure 12.

Cx30 expression in the human hair follicle.

In addition to the cell-cell adhesion structures described above, tight junction (TJ) also exists in the HF epithelium and expression patterns of TJ-associated proteins in the HF have previously been characterized [61]. Disruption of CLDN1 gene encoding claudin 1, a major structural component of TJ, has recently been shown to cause a severe autosomal recessive syndrome, known as ichthyosis, leukocyte vacuoles, alopecia, and sclerosing cholangitis (MIM 607626) [62].

Advertisement

5. Hereditary hair disorders associated with transcription factors

During the past 20 years, numerous genes that are expressed in the HF have been identified, and various transcription factors have been shown to be involved in transcriptional regulation of these genes. Of these, p63 is one of the main transcription factors expressed in the HF. During the HF morphogenesis, p63 is abundantly expressed in the HF placode (Figure 13). In the postnatal stage, it is strongly expressed in the ORS and the matrix region of the HF (Figure 14). It has previously been reported that mutations in TP63 gene encoding p63 cause several autosomal dominant diseases including ectodermal dysplasia, ectrodactyly, cleft lip/palate (EEC) syndrome (MIM 604292), ankyloblepharon, ectodermal defects, and cleft lip/palate (AEC) syndrome (MIM 106260) and Rapp-Hodgkin syndrome (MIM 129400) (Table 3) [63-65]. In most cases, patients with these syndromes result in scarring alopecia, and their hair shafts are coarse and twisted (Figure 15). It is noteworthy that affected individuals with TP63 mutations show large phenotypic overlaps in hair and limbs with P-cadherin (CDH3) mutations. p63 colocalizes with P-cadherin in developing HF placode and limb buds during mouse embryogenesis. Importantly, it has been demonstrated that the CDH3 is a direct target gene of p63 [66].

Figure 13.

P63 expression in the developing mouse hair follicle placode.

Figure 14.

p63 expression in the human hair follicle.

diseaseinheritance patternOMIM#main symptomsgeneProtein, function
Ectrodactyly, ectodermal dysplasia, and cleft lip/palate (EEC ) syndromeAD604292hypotrichosis, ectrodactyly, cleft lip/palate, hypodontiaTP63tumor protein p63
Ankyloblepharon, ectodermal defects, and cleft lip/palate (AEC) syndromeAD106260hypotrichosis, ankyloblepharon, skin erosion, cleft lip/palate, hypodontiaTP63tumor protein p63
Rapp-Hodgkin syndromeAD129400hypotrichosis, cleft lip/palate, hypodontiaTP63tumor protein p63
T cell immunodeficiency, congenital alopecia, and nail dystrophy (human nude phenotype)AR601705atrichia, nail dystrophy, T-cell immuodeficiencyFOXN1Forkhead box N1
Atrichia with papular lesionsAR209500atrichia, papulesHRHair less (transcriptional corepressor)
Marie-Unna hereditary hypotrichosis AD146550Hypotrichosis, wiry hairU2HRSmall peptide that regulates translation of the HR protein
Trichorhinophalangeal syndrome type I/type IIIAD190350/190351Hypotrichosis, peer-shaped nose, brachydactyly, clinodactylyTRPS1Zing finger transcription factor
Hypotrichosis-lymphedema-telangiectasia syndromeAD/AR607823Hypotrichosis, lymphedema, telangiectasia (easily visible blood vessels)SOX18SRY-BOX 18
Trichodontoosseous syndromeAD190320WH, hypodontia, bone anomaliesDLX3Distal-less homeobox 3

Table 3.

Hereditary hair disorders resulting from mutations in transcription factors.

FOXN1, also known as WHN, is a transcription factor expressed in the matrix and the hair shaft of the HF, and has been shown to regulate the expression of several hair keratin genes [67]. FOXN1 is expressed in not only the HF, but also in the nail units and thymus. Mutations in the FOXN1 gene have been reported to underlie T-cell immunodeficiency, congenital alopecia, and nail dystrophy (MIM 601705), which is an autosomal recessive disease and represents the human counterpart of the nude mouse phenotype, suggesting the crucial roles of FOXN1 in development of skin appendages, as well as thymus in both humans and mice [68].

Figure 15.

Clinical features of Rapp-Hodgkin syndrome.

Hairless (HR) is a putative single zinc-finger transcription factor which is known to regulate the catagen phase of the hair cycle [69]. Recessively-inherited mutations in the HR gene have been shown to underlie atrichia with popular lesions (APL; MIM 209500) [70]. APL is characterized by early onset of generalized complete hair loss (atrichia), which is followed by papular eruptions due to formation of dermal cyst after an abnormal first catagen phase [71]. Mutations responsible for APL have been found in coding exons or exon-intron boundary sequences of the HR gene, all of which were predicted to result in loss of expression and/or function of the HR protein. Recently, another disease, known as Marie-Unna hypotrichosis (MUH; MIM 146550), has been shown to be associated with the HR gene. MUH is a non-syndromic hereditary hair disorder showing an autosomal dominant inheritance pattern. Affected individuals with MUH typically exhibit sparse scalp and facial hair at birth. Subsequently, coarse, wiry, and twisted hairs develop in early childhood. Hair loss progresses with aging, which leads to a complete alopecia or a phenotype just like androgenetic alopecia. MUH was previously mapped to the HR locus on chromosome 8p21.3 [72]. However, direct sequencing analysis of coding sequences of the HR gene failed to detect mutations. Later on, Wen et al. found that the promoter region of the HR gene has four potential upstream open reading frames (uORFs), which were designated U1HR-U4HR. Strikingly, direct sequencing analysis of the U1HR-U4HR in patients with MUH has led to the identification of mutations within the U2HR sequences, which encode a small peptide of 34 amino acid residues [73]. In vitro studies have suggested that this small peptide encoded by the U2HR downregulates the HR expression at the translational level, and loss-of-function mutations in the U2HR results in overexpression of the HR protein [73]. Besides these findings, actual consequences resulting from U2HR mutations in vivo remain elusive.

TRPS1 is a transcription factor with GATA-type and Ikaros-type zinc finger domains, which has been shown to be abundantly expressed in both epithelial and mesenchymal components in the developing mouse HFs [74]. Furthermore, it has recently been reported that Trps1 plays crucial roles in regulating the expression of several Wnt inhibitors and various transcription factors during vibrissa follicle morphogenesis in mice [75]. In humans, mutations in the TRPS1 gene are known to cause trichorhinophalangeal syndrome type I (TRPS I; MIM 190350) or type III (TRPS III; MIM 190351), both of which show an autosomal dominant inheritance trait, and are characterized by sparse hair and a number of craniofacial and skeletal abnormalities, such as peer-shaped nose and brachydactyly. Hypotrichosis is the most prominent in the temporal region of the scalp (Figure 16) [76, 77].

Figure 16.

Clinical features of TRPS I.

In addition to the transcription factors described above, several other members are also associated with hereditary hair diseases. For instance, both dominantly- and recessively-inherited mutations in SOX18 gene underlie hypotrichosis-lymphedema-telangiectasia syndrome (MIM 607823) [78] and dominantly-inherited mutations in DLX3 gene cause trichodontoosseous syndrome (MIM 190320), respectively (Table 3) [79].

Advertisement

6. Hereditary hair disorders caused by disruption in signaling pathways

It has been shown via analyses using mice models that several signaling pathways play crucial roles in the HF morphogenesis and development. In humans, disruption of these signaling pathways has been demonstrated to underlie various hereditary hair disorders (Table 4). In addition, information obtained from the analysis of hereditary hair diseases has highlighted a novel signaling pathway that had not previously been known to play a role in the HF development.

diseaseinheritance patternOMIM#main symptomsgeneprotein, function
Hypohidrotic ectodermal dysplasiaXR305100Hypotrichosis, hypohidrosis, hypodontiaEDAectodysplasin A1 (EDA-A1)
AD129490EDAR
EDARADD
TRAF6
EDA-A1 receptor
EDAR-associated death domain
TNF receptor-associated factor 6
AR224900EDAR
EDARADD
EDA-A1 receptor
EDAR-associated death domain
Odontoonychodermal dysplasiaAR257980Hypotrichosis, hypodontia, nail dystrophy, PPKWNT10AWnt ligand
Generalized hereditary hypotrichosis simplexAD605389hypotrichosisAPCDD1Wnt inhibitor
Localized autosomal recessive hypotrichosis 2 (LAH2)/autosomal recessive woolly hair 2 (ARWH2)AR604379WH, hypotrichosisLIPHphosphatidic acid-selective phospholipase A1α (PA-PLA1α)
LAH3/ARWH1AR611452/278150WH, hypotrichosisLPAR6LPA6 (LPA receptor)
Inflammatory skin and bowel diseaseAR614328erythema, diarrhea, WH ADAM17Tumor necrosis factor converting enzyme (TACE)

Table 4.

Hereditary hair disorders associated with disruption of signaling pathways.

Hypohidrotic ectodermal dysplasia (HED), also known as Christ-Siemens-Touraine syndrome, is a rare genetic disease characterized by abnormal development of hair, teeth, and sweat glands. Most cases of HED show an X-linked recessive inheritance pattern (MIM 305100), while a minority of HED is inherited as either an autosomal dominant (MIM 129490) or an autosomal recessive trait (MIM 224900). During the last 15 years, the molecular basis for HED has gradually been disclosed. X-linked HED is caused by mutations in ectodysplasin (EDA) gene [80], and autosomal forms of HED are resulting from mutations in either EDA-receptor (EDAR) [81] or EDAR-associated death domain (EDARADD) [82] genes. The EDA gene encodes several isoforms of a type II transmembrane protein via alternative splicing [83]. Of these, ectodysplasin-A1 (EDA-A1) is the longest isoform which belongs to the tumor necrosis factor (TNF) ligand superfamily. EDAR, the receptor of EDA-A1 [84], is a type I transmembrane protein and a member of the TNF receptor superfamily with a potential death domain in its intracellular region. During the development of ectoderm-derived organs, EDA-A1 binds to its receptor EDAR, which subsequently associates with its adaptor EDARADD. Additionally, EDARADD protein interacts with TNF receptor-associated factor 6 (TRAF6), which further forms a complex with TGFβ-activated kinase 1 (TAK1) and TAK1-binding protein 2 (TAB2) within the cytoplasm, leading to activate the downstream NF-κB [85]. Most recently, a heterozygous mutation in the TRAF6 gene has been identified in a patient showing typical clinical features of HED [86]. Since EDA-A1, EDAR, EDARADD, and TRAF6 are closely related to each other in a signaling pathway, mutations in any of these four pathway components result in identical phenotypic characteristics among patients.

Odontoonychodermal dysplasia (OODD; MIM 257980) is an autosomal recessive disease which is characterized by various ectodermal abnormalities including hypotrichosis, hypodontia, nail dystrophy, and palmoplantar keratoderma. It has recently been shown that OODD is caused by loss of function mutations in the WNT10A gene, which encodes a WNT ligand [87]. It is noted that some affected individuals with WNT10A mutations can show phenotypes resembling HED [88], indicating the close relationship between EDA-A1/EDAR signaling and Wnt signaling, which has also been suggested by experiments in mice models [89].

In addition to Wnt ligands, abnormal function of Wnt inhibitors has recently been shown to cause a hereditary hair disorder in humans. Generalized hypotrichosis simplex (GHS; MIM 605389) is an autosomal dominant non-syndromic hair disorder which is characterized by progressive loss of scalp and body hairs starting in the middle of the first decade of life and almost complete baldness by the third decade [90]. In several families with GHS, an identical heterozygous missense mutation (L9R) has been identified in APCDD1 gene on chromosome 18p11.22 [91, 92]. The APCDD1 gene encodes a single-pass transmembrane protein which is abundantly expressed in the dermal papilla, the matrix and the hair shaft of human HF. Functional studies in cultured cells, chick embryos, and xenopus have revealed that APCDD1 inhibits Wnt signaling potentially via interacting Wnt ligands and their co-receptors LRPs [91]. In addition, it has been demonstrated that the L9R-mutant APCDD1 protein functions in a dominant negative manner against wild-type APCDD1 protein [91]. Therefore, Wnt activity is predicted to be upregulated in patients’ HFs. It is postulated that chronic stimulation by Wnt signaling may result in depletion of stem cell pool in the HF bulge, leading to GHS.

Recently, a signaling of lipid mediators has been shown to play essential roles in hair growth. About a decade ago, phosphatidic acid, has been demonstrated to promote hair growth in organ culture system, suggesting a potential role of lipids in hair growth [93]. Later on, it has been reported that mutations in lipase H (LIPH) gene underlies an autosomal recessively-inherited hypotrichosis (Localized autosomal recessive hypotrichosis 2 (LAH2); MIM 604379) [94]. Affected individuals with LAH2 show sparse hair on their scalp and extremities, whereas facial and sexual hairs look normal. In addition, it is noteworthy that patients with LIPH mutations show woolly hair (WH) in high frequency (Figure 17) [95], thus the LIPH can be regarded as a causative gene responsible for autosomal recessive WH (ARWH). Most affected individuals with LIPH mutations showed mainly WH during early childhood, and then exhibited wide variability in the hypotrichosis phenotype with aging [96].

Figure 17.

Clinical features of LAH2/woolly hair caused by mutations in the LIPH gene.

The LIPH gene encodes cell membrane-associated phosphatidic acid-selective phospholipase A1α (PA-PLA1α) which produces 2-acyl lysophosphatidic acid (LPA) from phosphatidic acid [97]. As LPA activates cells through binding with its receptor, the existence of LPA receptor(s) in the HF had been expected, which has been idenitified by the analyses of additional families with ARWH/hypotrichosis without carrying mutations in the LIPH gene. Affected individuals in these families showed WH and associated hypotrichosis (Localized autosomal recessive hypotrichosis 3 (LAH3); MIM611452), which were almost identical phenotypes to those with LIPH mutations. Linkage studies and positional cloning have led to the identification of mutations in LPAR6 gene, also known as P2RY5, in these families [98, 99]. The LPAR6 gene encodes a G protein-coupled receptor LPA6 (P2Y5), which has clearly been proved to be a receptor of LPA [100]. Both PA-PLA1α and LPA6 are mainly expressed in the IRS of human HF [24, 99]. Importantly, their expression overlaps with K71 and K74, of which mutations underlie autosomal dominant WH/hypotrichosis. Sum of these data strongly suggest the crucial roles of PA-PLA1α/LPA/LPA6 pathway in the HF differentiation and hair growth, and its downstream signaling may be involved in regulating expression of the IRS-specific keratins. More recently, significant findings have been reported, which have revealed the downstream signaling of the PA-PLA1α/LPA/LPA6 pathway. Inoue et al. have produced Liph-knockout (KO) mice which exhibited a wavy coat phenotype resembling WH in humans [101]. In addition, a series of expression studies in the mutant mice, as well as detailed in vitro analyses, have demonstrated that the PA-PLA1α/LPA/LPA6 axis regulates differentiation and maturation of mouse HF via a signaling pathway composed of tumor necrosis factor converting enzyme (TACE), transforming growth facor (TGF)-α, and epidermal growth factor receptor (EGFR) [101]. It has been shown that LPA produced by PA-PLA1α stimulated its receptor LPA6, which subsequently activated TACE. Then, TACE induced ectodomain shedding of TGF-α, which resulted in transactivation of EGFR (Figure 18) [101]. Notably, in the HF of the Liph-KO mice, the expression of cleaved TGF-α, tyrosine-phosphorylated EGFR, LPA, and the IRS-specific K71, were significantly reduced [101]. Most recently, a recessively-inherited mutation in ADAM17 gene encoding TACE have been shown to cause inflammatory skin and bowel disease (MIM 614328) in humans, and affected individuals with the ADAM17 mutation appear to show WH phenotypes, similar to patients with LIPH or LPAR6 mutations [102]. These findings strongly suggest that the PA-PLA1α/LPA/LPA6 signaling can be involved in activating TACE in humans as well.

Figure 18.

Schematic representation of the PA-PLA1α/LPA/LPA6 signaling pathway.

Advertisement

7. Conclusions

To identify causative genes responsible for hereditary hair disorders, as well as to disclose the functional relationship between these genes, has provided precious information to better understand the complex mechanisms for the HF development and cycling in humans. It is highly expected that recently-established methods in molecular genetics, especially whole genome sequencing [103], will enable us to find additional causative genes for the diseases. These genes may be associated with not only rare hair disorders, but also determining the hair texture in healthy individuals and/or more common hair diseases, such as alopecia areata and androgenetic alopecia.

Acknowledgement

I appreciate Drs. Atsushi Fujimoto and Hiroki Fujikawa (Niigata University, Japan) for their assistance to make figures. This work was supported in part by the Special Coordination Funds for Promoting Science and Technology from the Ministry of Education, Culture, Sports, Science and Technology of Japan (MEXT) to Y.S.

References

  1. 1. MillarS. EMolecular mechanisms regulating hair follicle development.J Invest Dermatol. 2002118221625
  2. 2. ShimomuraYChristianoA. MBiology and genetics of hair.Annu Rev Genomics Hum Genet. 20101110932
  3. 3. CotsarelisGSunT. TLavkerR. MLabel-retaining cells reside in the bulge area of pilosebaceous unit: implications for follicular stem Cells, hair cycle, and skin carcinogenesisCell. 1990617132937
  4. 4. OshimaHRochatAKedziaCKobayashiKBarrandonYMorphogenesis and renewal of hair follicles from adult multipotent stem cellsCell2001104223345
  5. 5. CoulombeP. AOmaryM. BHard’ and’soft’ principles defining the structure, function and regulation of keratin intermediate filaments. Curr Opin Cell Biol. 200214111022
  6. 6. ShimomuraYItoMHuman hair keratin-associated proteinsJ Investig Dermatol Symp Proc. 20051032303
  7. 7. RogersM. ALangbeinLPraetzel-wunderSWinterHSchweizerJHuman hair keratin-associated proteins (KAPs).Int Rev Cytol. 200625120963
  8. 8. RogersM. AWinterHWolfCHeckMSchweizerJCharacterization of a 190-kilobase pair domain of human type I hair keratin genes.J Biol Chem. 1998273412668391
  9. 9. RogersM. AWinterHLangbeinLWolfCSchweizerJCharacterization of a 300 kbp region of human DNA containing the type II hair keratin gene domainJ Invest Dermatol. 2000114346472
  10. 10. MollRDivoMLangbeinLThe human keratins: biology and pathology. Histochem Cell Biol. 2008129670533
  11. 11. LangbeinLYoshidaHPraetzel-wunderSParryD. ASchweizerJThe keratins of the human beard hair medulla: the riddle in the middle.J Invest Dermatol. 201013015573
  12. 12. ItoMHashimotoKYorderF. WMonilethrix: an ultrastructural study. J Cutan Pathol. 198411651321
  13. 13. WinterHRogersM. ALangbeinLStevensH. PLeighI. MLabrèzeCRoulSTaiebAKriegTSchweizerJMutations in the hair cortex keratin hHb6 cause the inherited hair disease monilethrix.Nat Genet. 19971643724
  14. 14. Van SteenselM. ASteijlenP. MBladergroenR. SVermeerMVan GeelMA missense mutation in the type II hair keratin hHb3 is associated with monilethrixJ Med Genet. 2005e19.
  15. 15. LangbeinLRogersM. AWinterHPraetzelSSchweizerJThe catalog of human hair keratins. II. Expression of the six type II members in the hair follicle and the combined catalog of human type I and II keratins.J Biol Chem. 2001276373512332
  16. 16. BarbareschiMCambiaghiSCrupiA. CTadiniGFamily with "pure" hair-nail ectodermal dysplasia.Am J Med Genet. 1997721913
  17. 17. NaeemMJelaniMLeeKAliGChishtiM. SWaliAGulAJohnPHassanM. JLealS. MAhmadWEctodermal dysplasia of hair and nail type: mapping of a novel locus to chromosome 1712q21Br J Dermatol. 2006
  18. 18. NaeemMWajidMLeeKLealS. MAhmadWA mutation in the hair matrix and cuticle keratin KRTHB5 gene causes ectodermal dysplasia of hair and nail typeJ Med Genet. 20064332749
  19. 19. ShimomuraYWajidMKurbanMSatoNChristianoA. MMutations in the keratin 85 (KRT85/hHb5) gene underlie pure hair and nail ectodermal dysplasia.J Invest Dermatol. 201013038925
  20. 20. PerrinCLangbeinLSchweizerJExpression of hair keratins in the adult nail unit: an immunohistochemical analysis of the onychogenesis in the proximal nail fold, matrix and nail bed. Br J Dermatol. 2004151236271
  21. 21. ChienA. JValentineM. CSybertV. PHereditary woolly hair and keratosis pilarisJ Am Acad Dermatol. 2006Suppl): S359
  22. 22. ShimomuraYWajidMPetukhovaLKurbanMChristianoA. MAutosomal-dominant woolly hair resulting from disruption of keratin 74 (KRT74), a potential determinant of human hair textureAm J Hum Genet. 20108646328
  23. 23. WasifNNaqviS. KBasitSAliNAnsarMAhmadWNovel mutations in the keratin-74 (KRT74) gene underlie autosomal dominant woolly hair/hypotrichosis in Pakistani families.Hum Genet. 2011129441924
  24. 24. FujimotoAFarooqMFujikawaHInoueAOhyamaMEhamaRNakanishiJHagiharaMIwabuchiTAokiJItoMShimomuraYA Missense Mutation within the Helix Initiation Motif of the Keratin K71 Gene Underlies Autosomal Dominant Woolly Hair/Hypotrichosis.J Invest Dermatol. 20121321023429doi:jid.2012.154.
  25. 25. LangbeinLRogersM. APraetzelSWinterHSchweizerJ. KIrsKIrsKIrsand Kirs4 represent the inner-root-sheath-specific type II epithelial keratins of the human hair follicle. J Invest Dermatol. 2003120451222
  26. 26. KikkawaYOyamaAIshiiRMiuraIAmanoTIshiiYYoshikawaYMasuyaHWakanaSShiroishiTTayaCYonekawaHA small deletion hotspot in the type II keratin gene mK6irs1/Krt2-6g on mouse chromosome 15, a candidate for causing the wavy hair of the caracul (Ca) mutation.Genetics. 2003165272133
  27. 27. PetersTSedlmeierRBüssowHRunkelFLüersG. HKorthausDFuchsHHrabé de Angelis M, Stumm G, Russ AP, Porter RM, Augustin M, Franz T. Alopecia in a novel mouse model RCO3 is caused by mK6irs1 deficiency. J Invest Dermatol. 2003121467480
  28. 28. KuramotoTHiranoRKuwamuraMSerikawaTIdentification of the rat Rex mutation as a 7-bp deletion at splicing acceptor site of the Krt71 geneJ Vet Med Sci. 201072790912
  29. 29. GandolfiBOuterbridgeC. ABeresfordL. GMyersJ. APimentelMAlhaddadHGrahnJ. CGrahnR. ALyonsL. AThe naked truth: Sphynx and Devon Rex cat breed mutations in KRT71Mamm Genome. 2010
  30. 30. CadieuENeffM. WQuignonPWalshKChaseKParkerH. GVonholdtB. MRhueABoykoAByersAWongAMosherD. SElkahlounA. GSpadyT. CAndréCLarkK. GCargillMBustamanteC. DWayneR. KOstranderE. ACoat variation in the domestic dog is governed by variants in three genes.Science. 200932659491503
  31. 31. McgrathJ. AInherited disorders of desmosomesAustralas J Dermatol. 20052219
  32. 32. KljuicABazziHSundbergJ. PMartinez-mirAOShaughnessyRMahoneyM. GLevyMMontagutelliXAhmadWAitaV. MGordonDUittoJWhitingDOttJFischerSGilliamT. CJahodaC. AMorrisR. JPanteleyevA. ANguyenV. TChristianoA. MDesmogleinin hair follicle differentiation and epidermal adhesion: evidence from inherited hypotrichosis and acquired pemphigus vulgaris. Cell. 2003113224960
  33. 33. ShimomuraYSakamotoFKariyaNMatsunagaKItoMMutations in the desmoglein 4 gene are associated with monilethrix-like congenital hypotrichosis.J Invest Dermatol. 2006126612815
  34. 34. SchafferJ. VBazziHVitebskyAWitkiewiczAKovichO. IKaminoHShapiroL. SAminS. POrlowS. JChristianoA. MMutations in the desmoglein 4 gene underlie localized autosomal recessive hypotrichosis with monilethrix hairs and congenital scalp erosions.J Invest Dermatol. 20061266128691
  35. 35. ZlotogorskiAMarekDHorevLAbuABen-amitaiDGeradLIngberAFrydmanMReznik-wolfHVardyD. APrasEAn autosomal recessive form of monilethrix is caused by mutations in DSG4: clinical overlap with localized autosomal recessive hypotrichosis.J Invest Dermatol. 2006126612926
  36. 36. BazziHGetzAMahoneyM. GIshida-yamamotoALangbeinLWahlJ. Krd, Christiano AM. Desmoglein 4 is expressed in highly differentiated keratinocytes and trichocytes in human epidermis and hair follicleDifferentiation2006129 EOF140 EOF
  37. 37. AyubMBasitSJelaniMUr Rehman F, Iqbal M, Yasinzai M, Ahmad W. A homozygous nonsense mutation in the human desmocollin-3 (DSC3) gene underlies hereditary hypotrichosis and recurrent skin vesicles.Am J Hum Genet. 200985451520
  38. 38. PayneA. SNo evidence of skin blisters with human desmocollin-3 gene mutation.Am J Hum Genet. 2010292 EOF
  39. 39. MckoyGProtonotariosNCrosbyATsatsopoulouAAnastasakisACoonarANormanMBaboonianCJefferySMckennaW. JIdentification of a deletion in plakoglobin in arrhythmogenic right ventricular cardiomyopathy with palmoplantar keratoderma and woolly hair (Naxos disease)Lancet. 20003559221211924
  40. 40. NorgettE. EHatsellS. JCarvajal-huertaLCabezasJ. CCommonJPurkisP. EWhittockNLeighI. MStevensH. PKelsellD. PRecessive mutation in desmoplakin disrupts desmoplakin-intermediate filament interactions and causes dilated cardiomyopathy, woolly hair and keratoderma.Hum Mol Genet. 200091827616
  41. 41. McgrathJ. AMcmillanJ. RShemankoC. SRunswickS. KLeighI. MLaneE. BGarrodD. REadyR. AMutations in the plakophilin 1 gene result in ectodermal dysplasia/skin fragility syndromeNat Genet. 19971722404
  42. 42. Ishida-yamamotoAIgawaSKishibeMOrder and disorder in corneocyte adhesion.J Dermatol. 201138764554doi:j.1346-8138.2011.01227.x.
  43. 43. MilsVVincentCCrouteFSerreGThe expression of desmosomal and corneodesmosomal antigens shows specific variations during the terminal differentiation of epidermis and hair follicle epithelia.J Histochem Cytochem. 1992409132937
  44. 44. Levy-nissenbaumEBetzR. CFrydmanMSimonMLahatHBakhanTGoldmanBBygumAPierickMHillmerA. MJoncaNToribioJKruseRDewaldGCichonSKubischCGuerrinMSerreGNöthenM. MPrasEHypotrichosis simplex of the scalp is associated with nonsense mutations in CDSN encoding corneodesmosin.Nat Genet. 20033421513
  45. 45. CaubetCBoussetLClemmensenOSouriguesYBygumAChavanasSCoudaneFHsuC. YBetzR. CMelkiRSimonMSerreGA new amyloidosis caused by fibrillar aggregates of mutated corneodesmosinFASEB J. 2010249341626
  46. 46. ChavanasSBodemerCRochatAHamel-teillacDAliMIrvineA. DBonaféJ. LWilkinsonJTaïebABarrandonYHarperJ. IDe ProstYHovnanianAMutations in SPINK5, encoding a serine protease inhibitor, cause Netherton syndrome.Nat Genet. 20002521412
  47. 47. KomatsuNTakataMOtsukiNOhkaRAmanoOTakeharaKSaijohKElevated stratum corneum hydrolytic activity in Netherton syndrome suggests an inhibitory regulation of desquamation by SPINK5-derived peptides.J Invest Dermatol. 2002118343643
  48. 48. YangTLiangDKochP. JHohlDKheradmandFOverbeekP. AEpidermal detachment, desmosomal dissociation, and destabilization of corneodesmosin in Spink5-/- miceGenes Dev. 2004181923548
  49. 49. Basel-vanagaiteLAttiaRIshida-yamamotoARainshteinLBen Amitai D, Lurie R, Pasmanik-Chor M, Indelman M, Zvulunov A, Saban S, Magal N, Sprecher E, Shohat M. Autosomal recessive ichthyosis with hypotrichosis caused by a mutation in ST14, encoding type II transmembrane serine protease matriptaseAm J Hum Genet. 200780346777
  50. 50. JamoraCDasGupta R, Kocieniewski P, Fuchs E. Links between signal transduction, transcription and adhesion in epithelial bud development.Nature2003422692931722
  51. 51. SamuelovLSprecherETsurutaDBíróTKloepperJ. EPaus R. P-Cadherin Regulates Human Hair Growth and Cycling via Canonical Wnt Signaling and Transforming Growth Factor-β2. J Invest Dermatol. 201213210233241doi:jid.2012.171.
  52. 52. SprecherEBergmanRRichardGLurieRShalevSPetroniusDShalataAAnbinderYLeibuRPerlmanICohenNSzargelRHypotrichosis with juvenile macular dystrophy is caused by a mutation in CDH3, encoding P-cadherin.Nat Genet. 20012921346
  53. 53. KjaerK. WHansenLSchwabeG. CMarques-de-fariaA. PEibergHMundlosSTommerupNRosenbergTDistinct CDH3 mutations cause ectodermal dysplasia, ectrodactyly, macular dystrophy (EEM syndrome)J Med Genet. 20054242928
  54. 54. Indelman M, Hamel CP, Bergman R, Nischal KK, Thompson D, Surget MO, Ramon M, Ganthos H, Miller B, Richard G, Lurie R, Leibu R, Russell-Eggitt I, Sprecher E. Phenotypic diversity and mutation spectrum in hypotrichosis with juvenile macular dystrophy. J Invest Dermatol. 2003;121 5 121720 .
  55. 55. RichardGConnexins: a connection with the skin. Exp Dermatol. 2000927796
  56. 56. LamartineJMunhoz Essenfelder G, Kibar Z, Lanneluc I, Callouet E, Laoudj D, Lemaître G, Hand C, Hayflick SJ, Zonana J, Antonarakis S, Radhakrishna U, Kelsell DP, Christianson AL, Pitaval A, Der Kaloustian V, Fraser C, Blanchet-Bardon C, Rouleau GA, Waksman G. Mutations in GJB6 cause hidrotic ectodermal dysplasia.Nat Genet. 20002621424
  57. 57. RichardGRouanFWilloughbyC. EBrownNChungPRyynänenMJabsE. WBaleS. JDiGiovanna JJ, Uitto J, Russell L. Missense mutations in GJB2 encoding connexin-26 cause the ectodermal dysplasia keratitis-ichthyosis-deafness syndromeAm J Hum Genet. 200270513418
  58. 58. JanA. YAminSRatajczakPRichardGSybertV. PGenetic heterogeneity of KID syndrome: identification of a Cx30 gene (GJB6) mutation in a patient with KID syndrome and congenital atrichiaJ Invest Dermatol. 20041225110813
  59. 59. SalomonDMasgrauEVischerSUllrichSDupontESappinoPSauratJ. HMedaPTopography of mammalian connexins in human skinJ Invest Dermatol. 199410322407
  60. 60. EssenfelderG. MLarderetGWaksmanGLamartineJGene structure and promoter analysis of the human GJB6 Geneencoding connexin 30.Gene. 200535013340
  61. 61. BrandnerJ. MMcintyreMKiefSWladykowskiEMollIExpression and localization of tight junction-associated proteins in human hair follicles.Arch Dermatol Res. 2003295521121
  62. 62. Hadj-rabiaSBaalaLVabresPHamel-teillacDJacqueminEFabreMLyonnetSDe ProstYMunnichAHadchouelMSmahiAClaudin-1 gene mutations in neonatal sclerosing cholangitis associated with ichthyosis: a tight junction diseaseGastroenterology20041275138690
  63. 63. CelliJDuijfPHamelB. CBamshadMKramerBSmitsA. PNewbury-ecobRHennekamR. CVan BuggenhoutGVan HaeringenAWoodsC. GVan EssenA. JDe WaalRVriendGHaberD. AYangAMckeonFBrunnerH. GVan BokhovenHHeterozygous germline mutations in the 53homolog p63 are the cause of EEC syndromeCell1999
  64. 64. McgrathJ. ADuijfP. HDoetschVIrvineA. DDe WaalRVanmolkotK. RWessagowitVKellyAAthertonD. JGriffithsW. AOrlowS. JVan HaeringenAAusemsM. GYangAMckeonFBamshadM. ABrunnerH. GHamelB. CVan BokhovenHHay-Wells syndrome is caused by heterozygous missense mutations in the SAM domain of63Hum Mol Genet. 2001
  65. 65. KantaputraP. NHamadaTKumchaiTMcgrathJ. AHeterozygous mutation in the SAM domain of 63underlies Rapp-Hodgkin ectodermal dysplasia.J Dent Res. 2003
  66. 66. ShimomuraYWajidMShapiroLChristiano AM. P-cadherin is a 63target gene with a crucial role in the developing human limb bud and hair follicle.Development. 2008
  67. 67. SchlakeTSchorppMMaul-pavicicAMalashenkoA. MBoehmTForkhead/winged-helix transcription factor Whn regulates hair keratin gene expression: molecular analysis of the nude skin phenotype.Dev Dyn. 2000217436876
  68. 68. FrankJPignataCPanteleyevA. AProwseD. MBadenHWeinerLGaetanielloLAhmadWPozziNCserhalmi-friedmanP. BAitaV. MUyttendaeleHGordonDOttJBrissetteJ. LChristianoA. MExposing the human nude phenotype.Nature199939867274734
  69. 69. PanteleyevA. ABotchkarevaN. VSundbergJ. PChristianoA. MPausRThe role of the hairless (hr) gene in the regulation of hair follicle catagen transformationAm J Pathol. 1999155115971
  70. 70. AhmadWFaiyaz ul Haque M, Brancolini V, Tsou HC, ul Haque S, Lam H, Aita VM, Owen J, deBlaquiere M, Frank J, Cserhalmi-Friedman PB, Leask A, McGrath JA, Peacocke M, Ahmad M, Ott J, Christiano AM. Alopecia universalis associated with a mutation in the human hairless gene. Science. 199827953517204
  71. 71. SprecherEBergmanRSzargelRFriedman-birnbaumRCohenNIdentification of a genetic defect in the hairless gene in atrichia with papular lesions: evidence for phenotypic heterogeneity among inherited atrichias.Am J Hum Genet. 199964513239
  72. 72. Van SteenselMSmithF. JSteijlenP. MKluijtIStevensH. PMessengerAKremerHDunnillM. GKennedyCMunroC. SDohertyV. RMcgrathJ. ACovelloS. PColemanC. MUittoJMcleanW. HThe gene for hypotrichosis of Marie Unna maps between D8S258 and D8S298: exclusion of the hr gene by cDNA and genomic sequencing.Am J Hum Genet. 19996524139
  73. 73. WenYLiuYXuYZhaoYHuaRWangKSunMLiYYangSZhangX. JKruseRCichonSBetzR. CNöthenM. MVan SteenselM. AVan GeelMSteijlenP. MHohlDHuberMDunnillG. SKennedyCMessengerAMunroC. STerrinoniAHovnanianABodemerCDe ProstYPallerA. SIrvineA. DSinclairRGreenJShangDLiuQLuoYJiangLChenH. DLoW. HMcleanW. HHeC. DZhangXLoss-of-function mutations of an inhibitory upstream ORF in the human hairless transcript cause Marie Unna hereditary hypotrichosis.Nat Genet. 200941222833
  74. 74. FantauzzoK. ABazziHJahodaC. AChristianoA. MDynamic expression of the zinc-finger transcription factor Trps1 during hair follicle morphogenesis and cyclingGene Expr Patterns. 200882517
  75. 75. FantauzzoK. AChristianoA. MTrps1 activates a network of secreted Wnt inhibitors and transcription factors crucial to vibrissa follicle morphogenesisDevelopment2012139120314
  76. 76. MomeniPGlöcknerGSchmidtOVon HoltumDAlbrechtBGillessen-kaesbachGHennekamRMeineckePZabelBRosenthalAHorsthemkeBLüdeckeH. JMutations in a new gene, encoding a zinc-finger protein, cause tricho-rhino-phalangeal syndrome type I.Nat Genet. 2000241714
  77. 77. LüdeckeH. JSchaperJMeineckePMomeniPGrossSVon HoltumDHircheHAbramowiczM. JAlbrechtBApacikCChristenH. JClaussenUDevriendtKFastnachtEFordererAFriedrichUGoodshipT. HGreiweMHammHHennekamR. CHinkelG. KHoeltzenbeinMKayseriliHMajewskiFMathieuMMcleodRMidroA. TMoogUNagaiTNiikawaNOrstavikK. HPlöchlESeitzCSchmidtkeJTranebjaergLTsukaharaMWittwerBZabelBGillessen-kaesbachGHorsthemkeBGenotypic and phenotypic spectrum in tricho-rhino-phalangeal syndrome types I and IIIAm J Hum Genet. 20016818191
  78. 78. IrrthumADevriendtKChitayatDMatthijsGGladeCSteijlenP. MFrynsJ. PVan SteenselM. AVikkulaMMutations in the transcription factor gene SOX18 underlie recessive and dominant forms of hypotrichosis-lymphedema-telangiectasia.Am J Hum Genet. 200372614708
  79. 79. PriceJ. ABowdenD. WWrightJ. TPettenatiM. JHartT. CIdentification of a mutation in DLX3 associated with tricho-dento-osseous (TDO) syndrome.Hum Mol Genet. 1998735639
  80. 80. KereJSrivastavaA. KMontonenOZonanaJThomasNFergusonBMunozFMorganDClarkeABaybayanPChenE. YEzerSSaarialho-kereUDe La ChapelleASchlessinger D. X-linked anhidrotic (hypohidrotic) ectodermal dysplasia is caused by mutation in a novel transmembrane protein. Nat Genet. 199613440916
  81. 81. MonrealA. WFergusonB. MHeadonD. JStreetS. LOverbeekP. AZonanaJMutations in the human homologue of mouse dl cause autosomal recessive and dominant hypohidrotic ectodermal dysplasia.Nat Genet. 19992243669
  82. 82. HeadonD. JEmmalS. AFergusonB. MTuckerA. SJusticeM. JSharpeP. TZonanaJOverbeekP. AGene defect in ectodermal dysplasia implicates a death domain adapter in development.Nature200141468669136
  83. 83. BayésMHartungA. JEzerSPispaJThesleffISrivastavaA. KKereJThe anhidrotic ectodermal dysplasia gene (EDA) undergoes alternative splicing and encodes ectodysplasin-A with deletion mutations in collagenous repeatsHum Mol Genet. 199871116619
  84. 84. YanMWangL. CHymowitzS. GSchilbachSLeeJGoddardADe VosA. MGaoW. QDixitV. MTwo-amino acid molecular switch in an epithelial morphogen that regulates binding to two distinct receptors.Science. 200029054915237
  85. 85. MikkolaM. LMolecular aspects of hypohidrotic ectodermal dysplasiaAm J Med Genet A. 2009A(9): 2031 EOF2036 EOF
  86. 86. WisniewskiS. ATrzeciakW. HA rare heterozygous TRAF6 variant is associated with hypohidrotic ectodermal dysplasia.Br J Dermatol. 2012166613536doi:j.1365-2133.2012.10871.x.
  87. 87. AdaimyLChoueryEMegarbaneHMrouehSDelagueVNicolasEBelguithHDe MazancourtPMegarbaneAMutation in WNT10A is associated with an autosomal recessive ectodermal dysplasia: the odonto-onycho-dermal dysplasiaAm J Hum Genet. 20078148218
  88. 88. CluzeauCHadj-rabiaSJambouMMansourSGuiguePMasmoudiSBalEChassaingNVincentM. CViotGClaussFManièreM. CToupenaySLe Merrer M, Lyonnet S, Cormier-Daire V, Amiel J, Faivre L, de Prost Y, Munnich A, Bonnefont JP, Bodemer C, Smahi A. Only four genes (EDA1, EDAR, EDARADD, and WNT10A) account for 90% of hypohidrotic/anhidrotic ectodermal dysplasia cases. Hum Mutat. 2011321702
  89. 89. ZhangYTomannPAndlTGallantN. MHuelskenJJerchowBBirchmeierWPausRPiccoloSMikkolaM. LMorriseyE. EOverbeekP. AScheidereitCMillarS. ESchmidt-ullrichRReciprocal requirements for EDA/EDAR/NF-kappaB and Wnt/beta-catenin signaling pathways in hair follicle induction.Dev Cell. 20091714961
  90. 90. BaumerABelliSTrüebR. MSchinzelAAn autosomal dominant form of hereditary hypotrichosis simplex maps to 1811p11.23 in an Italian familyEur J Hum Genet. 2000
  91. 91. ShimomuraYAgalliuDVonicaALuriaVWajidMBaumerABelliSPetukhovaLSchinzelABrivanlouA. HBarresB. AChristianoA. MAPCDD1 is a novel Wnt inhibitor mutated in hereditary hypotrichosis simplex.Nature2010464729110437
  92. 92. LiMChengRZhuangYYaoZA recurrent mutation in the APCDD1 gene responsible for hereditary hypotrichosis simplex in a large Chinese family.Br J Dermatol. 2012doi:j.13652133x.
  93. 93. TakahashiTKamimuraAHamazono-matsuokaTHondaSPhosphatidic acid has a potential to promote hair growth in vitro and in vivo, and activates mitogen-activated protein kinase/extracellular signal-regulated kinase kinase in hair epithelial cellsJ Invest Dermatol. 2003121344856
  94. 94. KazantsevaAGoltsovAZinchenkoRGrigorenkoA. PAbrukovaA. VMoliakaY. KKirillovA. GGuoZLyleSGinterE. KRogaevE. IHuman hair growth deficiency is linked to a genetic defect in the phospholipase gene LIPH.Science. 200631458019825
  95. 95. ShimomuraYWajidMPetukhovaLShapiroLChristianoA. MMutations in the lipase H gene underlie autosomal recessive woolly hair/hypotrichosis.J Invest Dermatol. 200912936228
  96. 96. ShimomuraYCongenital hair loss disorders: rare, but not too rare.J Dermatol. 2012391310doi:j.1346-8138.2011.01395.x.
  97. 97. SonodaHAokiJHiramatsuTIshidaMBandohKNagaiYTaguchiRInoueKAraiHA novel phosphatidic acid-selective phospholipase A1 that produces lysophosphatidic acid.J Biol Chem. 2002277373425463
  98. 98. PasternackS. MVon KügelgenIAl Aboud K, Lee YA, Rüschendorf F, Voss K, Hillmer AM, Molderings GJ, Franz T, Ramirez A, Nürnberg P, Nöthen MM, Betz RC. G protein-coupled receptor 2Y5and its ligand LPA are involved in maintenance of human hair growth.Nat Genet. 2008
  99. 99. ShimomuraYWajidMIshiiYShapiroLPetukhovaLGordonDChristianoA. MDisruption of 2RY5an orphan G protein-coupled receptor, underlies autosomal recessive woolly hair.Nat Genet. 2008
  100. 100. YanagidaKMasagoKNakanishiHKiharaYHamanoFTajimaYTaguchiRShimizuTIshiiSIdentification and characterization of a novel lysophosphatidic acid receptor, 2y5LPA6.J Biol Chem. 2009
  101. 101. InoueAArimaNIshiguroJPrestwichG. DAraiHAokiJLPA-producing enzyme PA-PLA1α regulates hair follicle development by modulating EGFR signalling. EMBO J. 20113020424860doi:emboj.2011.296.
  102. 102. Blaydon DC, Biancheri P, Di WL, Plagnol V, Cabral RM, Brooke MA, van Heel DA, Ruschendorf F, Toynbee M, Walne A, OToole EA, Martin JE, Lindley K, Vulliamy T, Abrams DJ, MacDonald TT, Harper JI, Kelsell DP. Inflammatory skin and bowel disease linked to ADAM17 deletion. N Engl J Med. 2011;365(16): 1502-8.
  103. 103. WheelerD. ASrinivasanMEgholmMShenYChenLMcguireAHeWChenY. JMakhijaniVRothG. TGomesXTartaroKNiaziFTurcotteC. LIrzykG. PLupskiJ. RChinaultCSongX. ZLiuYYuanYNazarethLQinXMuznyD. MMarguliesMWeinstockG. MGibbsR. ARothbergJ. MThe complete genome of an individual by massively parallel DNA sequencing.Nature200845271898726

Written By

Yutaka Shimomura

Submitted: 11 March 2012 Published: 06 February 2013