Fact-checked by Grok 2 weeks ago

Hypohidrotic ectodermal dysplasia

Hypohidrotic ectodermal dysplasia (HED) is a rare genetic disorder that primarily affects the development of ectodermal tissues, resulting in sparse scalp and body hair (hypotrichosis), reduced or absent sweating (hypohidrosis), and missing or abnormally shaped teeth (hypodontia). This condition disrupts the normal formation of structures derived from the ectoderm, such as skin appendages, leading to challenges in thermoregulation, dental function, and physical appearance. Clinically, individuals with HED often exhibit distinctive facial features, including a prominent , flattened , thick or everted lips, and periorbital wrinkling, alongside dry, smooth due to fewer sweat glands and hair follicles. The reduced sweating can cause recurrent fevers and overheating, particularly in infancy, while dental anomalies may include conical or peg-shaped teeth and delayed eruption. Additional features can involve hoarse voice, and in some cases, or atopic conditions, though intelligence and growth are typically unaffected. Severity varies, with classic forms showing profound manifestations and milder variants displaying subtler traits. HED is caused by pathogenic variants in genes involved in ectodermal development, most commonly EDA on the , which encodes ectodysplasin-A, a protein essential for ectoderm-mesoderm signaling. Other implicated genes include EDAR, EDARADD, and WNT10A, affecting the same signaling pathways. is primarily X-linked recessive, with males more severely affected and females showing variable expression due to ; autosomal recessive and dominant forms also occur. The prevalence of HED is estimated at approximately 1 in 20,000 to 1 in 100,000 births worldwide, making it the most common form of . relies on clinical evaluation of the characteristic triad and is confirmed through molecular . Management focuses on symptomatic relief, including dental prosthetics, cooling strategies for , and multidisciplinary care involving dermatologists, dentists, and geneticists; emerging therapies, such as prenatal recombinant EDA administration, are under investigation.

Overview

Definition

Hypohidrotic ectodermal dysplasia (HED) is a heterogeneous group of genetic disorders characterized by the abnormal development of ectodermal structures, primarily manifesting as hypotrichosis (sparse scalp and ), (reduced or absent ability to sweat), and (missing or that are few in number, small, or conical in shape). These features form the diagnostic core triad of the condition, with affected individuals often experiencing challenges related to temperature regulation due to impaired function and difficulties with and appearance due to and anomalies. HED belongs to the broader category of ectodermal dysplasias, a diverse collection of over 100 inherited disorders that selectively impact tissues derived from the embryonic , including skin appendages such as hair follicles, sweat glands, sebaceous glands, and teeth. The term "hypohidrotic" specifically denotes the reduced sweat production central to the disorder, while "ectodermal" refers to the affected tissue origins and "dysplasia" indicates the underlying developmental malformation. This classification highlights HED as the most common form of . The severity of HED symptoms varies widely among affected individuals, ranging from mild presentations with partial involvement of ectodermal structures to severe cases where the core is prominently expressed from infancy. This variability underscores the condition's clinical heterogeneity, though the triad remains the hallmark for across all forms.

Epidemiology

Hypohidrotic ectodermal dysplasia (HED) is a rare with an estimated ranging from 1 in 100,000 births to 2.99 per 100,000 individuals, based on recent analyses of large-scale health databases. The most common form, X-linked HED, has a birth of approximately 2.8 per 100,000 live births, as reported in a Danish national registry study spanning 1995 to 2011. These figures highlight the disorder's rarity, with overall ectodermal dysplasias affecting about 14.5 per 100,000 births globally. The X-linked form of HED exhibits a higher incidence in males due to its pattern, leading to full expression in affected males while females are typically carriers with milder or no symptoms. In contrast, autosomal forms of HED, which account for a smaller proportion of cases, affect males and females equally. U.S.-based studies using electronic health records from over 64 million individuals confirm these patterns, estimating a period prevalence of 2.99 per 100,000 for HED overall, with data indicating relative stability in occurrence rates over recent decades. HED is reported worldwide across all racial and ethnic groups, with no strong predisposition to specific populations, though cases often cluster within affected families due to its hereditary nature. Underdiagnosis is likely prevalent in regions with limited access to , potentially underestimating true global incidence. Registries and longitudinal studies from and the U.S., covering periods up to 2023, support the consistent rarity of the condition without significant temporal variations.

Clinical Presentation

Primary Ectodermal Features

Hypohidrotic ectodermal dysplasia (HED) is characterized by distinctive abnormalities in ectodermal-derived structures, primarily affecting , teeth, sweat glands, and . These features arise from impaired development of ectodermal tissues and manifest early in life, with varying severity depending on the genetic form and individual factors. Hair abnormalities in HED typically present as hypotrichosis, featuring sparse, fine, and light-colored and that grows slowly and is often brittle or fragile. Eyebrows and eyelashes are frequently reduced, absent, or similarly fine and sparse, contributing to a distinctive appearance. While secondary sexual hair, such as beard, axillary, or , may develop normally after , overall hair density remains diminished compared to unaffected individuals. Dental manifestations are a hallmark of HED, with affecting a significant portion of primary and , ranging from partial absence to complete anodontia in severe cases. Surviving teeth are often conical or pointed in shape, widely spaced, smaller than normal, and may exhibit deficiencies; eruption is characteristically delayed. The may appear underdeveloped with atrophic and a narrow alveolar ridge, leading to protrusive lips and challenges in mastication. On average, affected individuals have about nine , primarily canines and molars. Sweat gland dysfunction in HED results in or anhidrosis due to a reduced number or absence of functional eccrine glands, impairing and causing , recurrent fevers, or hyperpyrexia during exertion or warm environments. Sweat production is generally diminished across the body, though some cases show sparing around the perioral area or , with patchy distribution more common in milder or autosomal forms. Newborns may exhibit or peeling skin related to this . Skin involvement includes dry, thin, and soft texture prone to scaling, peeling (especially in infancy), and chronic eczema, often with or and fine wrinkles. Nail dysplasia occurs in some variants, manifesting as thin, brittle, ridged, or slow-growing nails, though changes are inconsistent and less prominent than other features. Abnormal , such as reduced dermal ridges, may also be observed.

Associated Manifestations

Hypohidrotic ectodermal dysplasia (HED) is associated with a range of secondary clinical features that extend beyond the core ectodermal defects, impacting ocular, respiratory, craniofacial, and other systems. These manifestations arise from the developmental abnormalities in ectoderm-derived structures and can vary in severity, often exacerbating morbidity in affected individuals. Ocular involvement is prominent, characterized by due to or aplasia of the lacrimal glands, which results in diminished tear production and symptoms such as dry eyes with bulbar conjunctival injection and irregular corneal surfaces. This tear deficiency heightens susceptibility to corneal abrasions, , infections, scarring, ulcers, and even in severe cases. Abnormal meibomian glands further contribute to ocular dryness and discomfort. Respiratory manifestations frequently include recurrent upper airway infections stemming from dysfunction, which impairs and leads to chronic , , and nasal obstruction due to crusting. Reduced salivary and bronchial function predisposes patients to frequent and respiratory tract infections, such as chronic and , with potential progression to in severe instances; asthma-like symptoms and obstructive airways disease may also occur, particularly in adulthood. These issues are compounded by the primary , which indirectly affects respiratory comfort through overall thermoregulatory challenges. Craniofacial features contribute to a distinctive appearance, including frontal bossing with a prominent , a deformity featuring a depressed or reduced and underdeveloped nostrils, midface , and thickened or everted lips. Additional traits may involve malar and mandibular , periorbital wrinkling, and a from vocal cord involvement. These structural anomalies can affect breathing, speech, and facial aesthetics, often becoming more evident with age. Among other associated features, temperature dysregulation is a critical concern, manifesting as hyperpyrexia and episodes of overheating, especially in infancy and during physical or environments, due to impaired function and altered thermal perception. This can lead to , convulsions during fever spikes, and increased risk of heat-related illness without interventions like cooling strategies. Occasional and middle ear problems, including , result from deficiencies in auditory tube submucosal glands, which compromise ear ventilation and increase susceptibility; raspy and throat hoarseness are also reported. Atopic dermatitis-like reactions, presenting as eczematous skin conditions, occur in many cases and contribute to illness. In X-linked HED, gender differences are notable, with affected males typically exhibiting more severe, uniform manifestations, while heterozygous females often display milder, mosaic patterns of features—such as patchy , variable dental anomalies, and irregular hair distribution—due to random X-chromosome inactivation (lyonization), which creates functional mosaicism following the lines of Blaschko. Skewed inactivation can occasionally result in more pronounced symptoms in females.

Genetics

X-Linked HED (EDA Gene)

X-linked hypohidrotic ectodermal dysplasia (XLHED), the most common subtype of hypohidrotic ectodermal dysplasia (HED), arises from mutations in the EDA gene located on the long arm of the at Xq13.1. The EDA gene encodes ectodysplasin A, a belonging to the (TNF) ligand superfamily that plays a critical role in signaling pathways essential for the of ectodermal appendages, including sweat glands, follicles, and teeth. This form of HED follows an pattern, meaning affected males, who are hemizygous for the , typically exhibit severe manifestations, while heterozygous females serve as carriers and often display milder or mosaic symptoms due to random X-chromosome inactivation. In carrier females, skewed X-inactivation can lead to variable expressivity, with some experiencing more pronounced features if the normal EDA allele is preferentially inactivated in affected tissues. Mutations in the EDA gene are predominantly loss-of-function variants, including missense, nonsense, frameshift, splice-site alterations, small insertions/deletions, and larger genomic deletions or duplications. Over 300 distinct EDA variants have been identified, many disrupting key functional domains such as the TNF-like homology domain, which is crucial for ligand-receptor binding and downstream signaling. For instance, missense mutations like p.Y304C or splice-site changes such as c.924+1dupG impair protein function and are recurrent in affected families. These mutations lead to deficient ectodysplasin A activity, halting normal ectodermal organogenesis. XLHED accounts for approximately 50%-60% of all HED cases, making it the most common genetic etiology. Family pedigrees characteristically show no male-to-male transmission, with the disorder passing from mothers to sons, consistent with . In affected males, the classic triad of (reduced or absent sweating), hypotrichosis (sparse scalp and ), and (missing or conical teeth) is prominently featured, often accompanied by characteristic facial dysmorphisms such as a prominent and . females may exhibit milder , dental anomalies like in up to 73% of cases, or subtle hair thinning, reflecting the mosaic pattern of .

Autosomal HED (EDAR, EDARADD, WNT10A Genes)

Autosomal forms of (HED) exhibit equal sex distribution, in contrast to the male predominance of X-linked HED. These variants arise from pathogenic variants in non-EDA genes involved in ectodermal signaling, with autosomal recessive inheritance more frequent in consanguineous populations due to the homozygous nature of such mutations. These frequencies may vary by population; for example, EDAR variants are more common in individuals of East Asian descent. Phenotypic overlap among autosomal HED includes prominent and sparse hair, though involvement varies, often being milder in dominant cases. The EDAR gene, located at chromosome 2q13, encodes the ectodysplasin A receptor, a that binds ectodysplasin A (EDA) to initiate NF-κB-mediated signaling critical for ectodermal formation, including hair follicles, teeth, and sweat glands. Pathogenic variants in EDAR cause autosomal dominant or recessive HED, with dominant forms typically resulting from dominant-negative that disrupt ligand binding and downstream signaling. These dominant EDAR variants often produce a milder than X-linked HED, featuring reduced but present sweating capacity, conical teeth, and fine , while recessive variants lead to more severe and oligodontia akin to the classic HED triad. For instance, the missense variant c.338G>A (p.Cys113Tyr) exemplifies a dominant associated with these features. EDAR variants account for 10%-15% of HED cases overall. The EDARADD gene, mapped to 1q42.2-q43, encodes an intracellular adapter protein that couples the EDAR receptor to TRAF6 and downstream activation, facilitating ectodermal development signals for , teeth, and glands. Mutations in EDARADD are rare and primarily autosomal recessive, comprising about 2%-3% of HED cases, and result in severe phenotypes closely resembling X-linked HED, with profound , hypotrichosis, and due to disrupted signaling. Reported variants include missense changes and deletions, such as an 4 deletion, that abolish protein function and pathway activation. The WNT10A gene at 2q35 encodes a secreted glycoprotein in the , which regulates ectodermal and , particularly for teeth, , and . Pathogenic variants in WNT10A predominantly cause autosomal recessive HED, with occasional dominant effects, and represent 15%-20% of cases; these often involve loss-of-function mutations leading to variable , severe oligodontia, and sparse hair. Unlike classic HED, WNT10A-related forms frequently include additional manifestations such as , nail dystrophy, and onychodysplasia, with defects being less consistent or severe in some individuals. A common recessive nonsense variant is c.321C>A (p.Cys107Ter), which truncates the protein and impairs Wnt pathway activity.

Pathophysiology

Molecular Mechanisms

Hypohidrotic ectodermal dysplasia (HED) arises primarily from disruptions in the EDA-EDAR-EDARADD signaling pathway, a (TNF)-like cascade essential for the formation of ectodermal appendages such as sweat glands, hair follicles, and teeth. Ectodysplasin A (EDA), a produced by ectodermal cells, binds to the EDAR receptor on adjacent cells, recruiting the adaptor protein EDARADD to initiate downstream signaling. This complex activates TRAF6, leading to the phosphorylation of TAK1 and the IKK complex, which ultimately liberates the transcription factor from its inhibitor, enabling its translocation to the nucleus to drive the expression of genes required for ectodermal development. Mutations in the genes encoding these pathway components typically result in loss-of-function effects, preventing effective ligand-receptor binding or , while some EDAR variants exert dominant-negative interference by forming non-functional multimers. For instance, missense mutations in EDA often disrupt the domain necessary for trimerization and furin-mediated cleavage, impairing EDA-A1 availability for EDAR engagement. Similarly, EDARADD mutations truncate the TRAF6-binding domain, blocking activation and leading to of . These defects collectively reduce the initiation and patterning of ectodermal structures during critical developmental windows. In autosomal forms of HED, mutations in WNT10A perturb Wnt/β-catenin signaling, which regulates epithelial-mesenchymal interactions vital for ectodermal organogenesis. WNT10A, a secreted , stabilizes β-catenin in the , allowing its nuclear translocation to form complexes with transcription factors like LEF1/TCF, promoting progenitor cell proliferation in dental lamina and hair placodes. Disruptions in this pathway, such as those from WNT10A loss-of-function variants, impair β-catenin-mediated upregulation of genes like for differentiation and DLX3 in for tooth root formation, resulting in defective and oligodontia. The core molecular processes in HED involve impaired ectodermal-mesenchymal signaling during human embryogenesis, particularly between weeks 8 and 12, when placode formation and budding occur for appendages. Reduced activation from EDA pathway defects limits the number and distribution of primordia and follicles, as targets such as Edar itself create a loop for placode maintenance. Wnt signaling complements this by ensuring proper epithelial and mesenchymal , with WNT10A mutations exacerbating in these structures. Animal models have been instrumental in elucidating these mechanisms, with the Tabby mouse (Eda mutant) exhibiting sparse hair, reduced sweat glands, and dental anomalies mirroring human X-linked HED due to absent EDA ligand. The Downless mouse (Edar mutant) similarly shows pathway blockade at the receptor level, confirming the centrality of EDA-EDAR signaling in ectodermal patterning and providing a platform for therapeutic testing, such as recombinant EDA administration to rescue phenotypes.

Phenotypic Variability

Hypohidrotic ectodermal dysplasia (HED) exhibits considerable phenotypic variability, influenced by genetic, environmental, and stochastic factors that modulate the expression of ectodermal structures such as , , , and . This variability manifests in the severity and distribution of core features like hypotrichosis, , and , as well as the presence of additional traits. While the signaling pathway disruption is central to all forms, individual differences arise from modifier effects and inheritance patterns. Genetic background plays a key role in modulating severity, with limited but evident genotype-phenotype correlations across HED subtypes. For instance, missense variants in EDA often correlate with milder dental phenotypes like nonsyndromic , whereas null variants lead to classic severe HED. Similarly, EDAR variants show a spectrum from mild to severe manifestations without strong predictive links to specific mutations. Environmental factors, such as ambient and , can exacerbate , leading to variable ; affected individuals may experience more pronounced symptoms during hot weather, necessitating adaptive strategies like cooling aids. In X-linked HED due to EDA variants, phenotypic variability is particularly pronounced in female carriers owing to random X-chromosome inactivation, resulting in mosaicism. Males typically present with severe, uniform features including near-total anhidrosis, sparse hair from birth, and oligodontia affecting over 20 teeth on average. In contrast, heterozygous females often display milder, patchy distributions of sweat glands and hair, with 60-80% experiencing some degree of hypodontia; skewed X-inactivation can intensify symptoms, mimicking hemizygous male severity in rare cases. Autosomal recessive forms (e.g., EDAR or EDARADD-related) tend to produce more uniform and severe phenotypes in both sexes, closely resembling X-linked male presentations, while autosomal dominant EDAR variants are generally milder with incomplete penetrance, featuring subtle hair thinning and partial hypohidrosis without the mosaic patterns seen in X-linked females. Age-related changes further contribute to phenotypic variability, with many features becoming apparent from infancy but evolving over time. and are often evident early, leading to recurrent fevers in newborns, while dental anomalies like delayed eruption and manifest during childhood as primary teeth fail to develop fully. Hair abnormalities, such as fine and sparse hair, are present at birth but may improve slightly in texture during ; post- progression is minimal, stabilizing the ectodermal defects. Certain HED subtypes show overlap with other syndromes, expanding the phenotypic spectrum. WNT10A-related HED frequently includes additional features like nail dystrophy (e.g., ridging or thickening) and palmoplantar , overlapping with odonto-onycho-dermal ; in some cases, it extends to Schopf-Schulz-Passarge syndrome, characterized by cystic skin lesions and . These overlaps highlight how WNT10A variants can produce a broader phenotype beyond classic HED triad elements.

Diagnosis

Clinical Assessment

Clinical assessment of hypohidrotic ectodermal dysplasia (HED) begins with a detailed and family history to identify patterns suggestive of the condition. A family is essential, particularly to detect X-linked patterns where affected males often have mothers or a history of similarly affected male relatives, while autosomal forms may show recessive or dominant familial clustering. Neonatal history frequently reveals due to impaired , feeding difficulties from reduced production, or skin peeling and irritability. Dental history commonly includes reports of delayed eruption, conical-shaped teeth, or significant , with affected individuals typically having fewer than 20 . Physical examination focuses on ectodermal structures to evaluate the classic triad of hypotrichosis, , and . Scalp and body hair density is assessed for sparseness, lightness, or brittleness, often with reduced eyebrow and eyelash growth. Oral inspection reveals , with an average of nine in affected males, conical crowns, and protuberant lips due to underdeveloped alveolar ridges. Skin evaluation notes dryness, fragility, , or eczematous changes, alongside facial features like a prominent and . Sweating ability is tested using methods such as the starch-iodine test, which highlights active sweat pores with color change, or to quantify sweat production and gland distribution, often showing a markedly reduced number of pores. Diagnostic checklists emphasize the presence of the core triad, with clinical suspicion heightened by the presence of the core triad features, including significant , sparse scalp hair, and reduced density. The Ectodermal Dysplasias Burden of Disease Score, a validated , assesses overall impact through patient-reported outcomes on symptoms like dental anomalies and seasonal , aiding in severity grading. These assessments guide initial , with genetic testing recommended for confirmation. Differential diagnosis requires distinguishing HED from other and syndromes. Hidrotic ectodermal dysplasia is differentiated by preserved sweating and prominent nail dystrophy, without the central to HED. Ectrodactyly-ectodermal dysplasia-clefting (EEC) syndrome is excluded by the absence of limb malformations like or and cleft lip/palate, which are not features of HED.

Genetic Testing

Genetic testing for hypohidrotic ectodermal dysplasia (HED) typically begins with targeted sequencing of the EDA gene in cases suspected to be X-linked, given its role in approximately 50%-60% of HED cases overall and up to 75%-95% of familial X-linked HED. If no pathogenic variant is identified, a multigene panel is recommended, encompassing EDAR, EDARADD, and WNT10A, which account for the remaining autosomal forms, including about 10%-15% for EDAR, 2%-3% for EDARADD, and 15%-20% for WNT10A. In instances where panel testing is negative, whole-exome sequencing may be pursued to detect rare or novel variants in other genes associated with ectodermal dysplasias. The diagnostic yield exceeds 90% for classic HED when appropriate gene selection is applied, with EDA sequencing alone yielding positive results in over 50% of sporadic cases. is available for at-risk pregnancies through (CVS) or to identify known familial pathogenic variants, enabling early diagnosis and options such as preimplantation . Pathogenic variants are classified according to the American College of and (ACMG) guidelines, which categorize them as pathogenic, likely pathogenic, uncertain significance, likely benign, or benign based on criteria including population data, computational predictions, functional studies, and segregation analysis. For carrier detection in females, particularly for X-linked HED, molecular methods such as or next-generation sequencing are used to identify heterozygous variants, while quantitative or (MLPA) helps detect copy number variations or assess . Such testing is widely available through clinical laboratories including and GeneDx, which offer ectodermal dysplasia panels covering EDA, EDAR, EDARADD, and WNT10A among other genes. As of 2025, turnaround times typically range from 10-21 days for panel testing at (average 14 days) to 2-4 weeks at GeneDx for exome-based approaches, with costs around $299 for panels at under their financial assistance programs.

Management

Symptomatic Support

Symptomatic support for hypohidrotic ectodermal dysplasia (HED) primarily focuses on alleviating the daily challenges posed by reduced sweating, dry , sparse , and dry eyes, emphasizing non-invasive strategies to improve . Management begins with to mitigate the risk of due to , as affected individuals have fewer functional sweat glands, impairing the body's ability to cool itself. Recommendations include maintaining access to air-conditioned environments at home, school, and work, wearing lightweight, breathable clothing such as cotton garments, and avoiding hot or humid conditions and strenuous physical activities. Frequent with cool liquids is essential, and tools like misting fans, spray bottles with cool , or cooling vests filled with gel packs can provide additional relief during outdoor activities or in uncooled spaces. For infants and young children, who are particularly vulnerable to overheating, close monitoring for signs such as , flushed , or is critical, with interventions like damp cloths, shaded areas, or chilled items to prevent febrile seizures or long-term complications. Skin and hair care addresses the chronic dryness and fragility resulting from ectodermal dysfunction, which predisposes individuals to xerosis, eczema, and infections. Daily application of emollients, such as or fragrance-free creams like or , is recommended immediately after to lock in and prevent cracking or . Broad-spectrum sunscreens with 30 or higher, containing physical blockers like zinc oxide, should be used regularly to protect sensitive from damage, with reapplication every two hours during sun exposure. For , which is often sparse, fine, and brittle, gentle shampoos and conditioners are advised, along with avoidance of heat styling or chemical treatments to minimize breakage; cosmetic options like wigs or hairpieces can address concerns. To prevent eczema flares, irritants such as harsh soaps should be avoided in favor of mild, superfatted cleansers, and dilute bleach baths may be incorporated weekly under medical supervision to reduce bacterial colonization on the . Ocular symptoms, including dry eyes from reduced tear production and meibomian gland abnormalities, require proactive lubrication to avert corneal abrasions or infections. , preservative-free lubricating , should be applied multiple times daily to maintain eye moisture and alleviate discomfort, grittiness, or light sensitivity. In cases of persistent dryness, punctal plugs—small devices inserted into tear ducts to block drainage—can be trialed to conserve natural , with versions used for longer-term management if effective. Protective , such as wraparound , is beneficial to shield against environmental irritants and wind. A multidisciplinary team, including dermatologists for skin concerns and geneticists for ongoing counseling, coordinates comprehensive care to address the systemic nature of HED. Education on prevention is vital, as dysfunctional glands increase susceptibility to skin and respiratory s; strategies include prompt of minor wounds with topical antibiotics, maintaining with gentle products, and for signs of bacterial overgrowth in dry areas. This holistic approach empowers individuals to manage symptoms effectively throughout life.

Dental and Multidisciplinary Care

Dental management in hypohidrotic ectodermal dysplasia (HED) focuses on addressing , conical teeth, and enamel defects to improve function, aesthetics, and . Early intervention is crucial, with pediatric dentists recommending initial evaluation by age 2-3 years to plan care. Removable partial or , often starting around ages 6-8 when primary teeth are lost, serve as interim prostheses to support mastication, speech, and facial growth; these require relining or replacement every 3-4 years due to ongoing jaw development. Orthodontic treatment is typically initiated in late childhood or to manage spacing anomalies and align existing teeth, often in coordination with prosthetics to prevent root resorption risks associated with . After skeletal maturity around age 18, dental implants offer a definitive solution, particularly in the anterior where volume is adequate; success rates range from 88.5% to 97.6%, though may be needed for atrophic ridges, and mini-implants can be used earlier in select cases. Preventive measures include biannual cleanings, high-fluoride applications, and dietary counseling to mitigate caries risk from hypoplastic . Multidisciplinary care extends beyond to address associated systemic issues. consultation is recommended for recurrent respiratory infections due to reduced sweat glands and nasal patency; interventions may include if chronic or persists. Ophthalmologic evaluation manages leading to dry eyes, with lubricating drops as standard therapy to prevent corneal damage. Speech therapy targets articulation and swallowing difficulties stemming from dental anomalies and reduced , often integrated with prosthetic fitting to enhance outcomes. Nutritional support is vital, particularly in infancy where impaired suckling from missing teeth or nipple in carrier mothers can lead to ; high-calorie formula feeds or thickened textures are advised to ensure adequate caloric intake (e.g., 1800 kcal/day for active school-age children). using standardized charts is essential, with dental prostheses aiding improved oral intake and . Long-term involves lifelong dental follow-up for prosthesis maintenance and , alongside multidisciplinary to track complications. Cosmetic procedures, such as using grafts for deformity, may be considered post-adolescence for psychosocial benefits, often as part of reconstructive efforts involving teams.

Emerging Therapies

Research into emerging therapies for hypohidrotic ectodermal dysplasia (HED) centers on causal interventions targeting the underlying genetic defects, particularly in X-linked HED (XLHED) caused by EDA gene mutations, with a focus on protein replacement to restore ectodysplasin A1 (EDA1) signaling. These approaches aim to induce development of sweat glands, , and teeth during critical embryonic or early postnatal windows, contrasting with lifelong symptomatic management. Biologics, specifically recombinant EDA1 fusion proteins like Fc-EDA (also known as ER004), represent the most advanced emerging option. In mouse models of XLHED, postnatal infusions of Fc-EDA promoted sweat gland neogenesis and improved , with effects persisting into adulthood. Early human studies involving nine male infants with XLHED demonstrated that three weekly subcutaneous doses shortly after birth increased density by up to 10-fold and enhanced sweating capacity, as measured by starch-iodine tests, with benefits lasting up to six years in follow-up. Prenatal administration via intra-amniotic injection has shown even greater potential; in a proof-of-concept of three male fetuses, three doses starting at gestational week 26 led to normalized development and reduced severity postnatally. The ongoing phase 2 EDELIFE (NCT04980638), enrolling up to 20 affected male fetuses, evaluates the safety and efficacy of ER004 prenatal , with primary endpoints including sweat count and respiratory infections at age 13 months. Safety profiles indicate mild, transient infusion reactions, with no serious adverse events linked to the . Stem cell-based strategies for dental manifestations, such as , are in preclinical and early exploratory phases, leveraging induced pluripotent stem cells (iPSCs) to regenerate buds. and animal models of congenital , including those mimicking pathways, have demonstrated that iPSC-derived epithelial and mesenchymal cells can form functional bioengineered germs when combined with scaffolds and growth factors like BMP4, potentially addressing missing in HED patients. These approaches target reactivation of arrested via Wnt and EDA signaling modulation, but no HED-specific human trials are registered as of 2025; broader regenerative dentistry trials for report successful implantation of stem cell-seeded constructs with 70-80% integration rates in small cohorts. As of 2025, comprehensive reviews highlight ongoing preclinical exploration of EDAR-targeted small molecules to mimic EDA1 signaling, with potential for to enhance downstream activation in non-XL HED forms, though no clinical trials are active. lists active studies primarily for XLHED protein therapy, underscoring biologics as the frontrunner, while surgical and ophthalmic advances, such as advanced limbal transplants for associated dry eye, continue to evolve in parallel.

Additional Topics

Genetic Counseling

Genetic counseling for hypohidrotic ectodermal dysplasia (HED) is essential for families following a diagnosis, providing information on inheritance patterns, recurrence risks, and reproductive choices to support informed decision-making. HED primarily follows X-linked recessive inheritance due to variants in the EDA gene, accounting for 50%-60% of cases, with autosomal recessive and dominant forms involving genes such as EDAR, EDARADD, and WNT10A comprising the remainder. For X-linked HED, a carrier mother has a 50% risk of having an affected son and a 50% risk of a carrier daughter per pregnancy, while affected males transmit the variant to all daughters (who become carriers) but none to sons. In autosomal recessive HED, if both parents are carriers, each child has a 25% chance of being affected, a 50% chance of being a carrier, and a 25% chance of being unaffected and non-carrier. Autosomal dominant HED carries a 50% risk to each child of an affected parent, though penetrance can vary. Reproductive options include (PGD), which allows selection of embryos without the pathogenic variant during fertilization, particularly useful for X-linked forms to avoid affected males. , such as performed at 10-12 weeks of gestation, can detect variants in at-risk pregnancies once the familial mutation is identified. Counseling remains non-directive, discussing options like pregnancy continuation with preparation for management or termination, tailored to family values and circumstances. Psychosocial support in genetic counseling addresses the emotional impact of the diagnosis, using pedigree analysis to illustrate recurrence risks and family implications. Counselors help families navigate uncertainties, such as variable expressivity, and connect them with resources like the National Foundation for Ectodermal Dysplasias (NFED), which offers peer support, educational materials, and advocacy for affected individuals. According to 2025 updates in clinical guidelines, early post-diagnosis is recommended to facilitate and long-term psychosocial adjustment.

History and Terminology

Hypohidrotic ectodermal dysplasia (HED) was first documented in the 1840s by , who observed familial patterns of hairlessness, missing teeth, and reduced sweating in affected individuals, noting the condition's hereditary nature across generations. This early account laid the groundwork for recognizing s as inherited disorders affecting . In 1929, A.A. Weech formalized the term "hereditary ectodermal dysplasia" in medical literature, emphasizing congenital defects in ectodermal structures and introducing "anhidrotic" to describe the profound impairment in function observed in many cases. By the 1950s, clinical studies incorporating biopsies and thermoregulatory assessments clarified the variability in sweating ability, leading to the distinction of the hypohidrotic subtype from completely anhidrotic forms. Key research milestones advanced understanding of HED's genetic basis in the late . The EDA gene, responsible for the X-linked form, was cloned in 1996 through positional cloning efforts that identified mutations disrupting ectodysplasin-A, a signaling protein essential for ectodermal development. Concurrently, animal models such as the Tabby mouse, which recapitulates HED phenotypes including sparse hair and absent s, were established in the 1990s, enabling mechanistic studies of ectodermal patterning. Therapeutic progress emerged in the 2010s with the initiation of trials, including prenatal administration of recombinant ectodysplasin to restore formation in affected fetuses. As of 2025, ongoing clinical trials such as the Edelife study are evaluating ER004, a recombinant protein mimicking ectodysplasin-A, for prenatal treatment of X-linked HED to improve development and reduce symptoms. Terminologically, HED was historically known as anhidrotic ectodermal dysplasia due to the emphasis on absent perspiration, but this shifted to "hypohidrotic" in the late 20th century to better reflect the spectrum of reduced, rather than absent, sweating in most patients. The X-linked hypohidrotic form is often termed Christ-Siemens-Touraine syndrome, named after its detailed clinical description in 1936, highlighting the classic triad of hypotrichosis, hypodontia, and hypohidrosis. In contrast, hidrotic ectodermal dysplasia, exemplified by Clouston syndrome, involves normal sweating but nail dystrophy and alopecia, distinguishing it genetically and phenotypically as an autosomal dominant condition caused by GJB6 mutations. From a 2025 perspective, genetic research has increasingly recognized overlaps between HED and odonto-onycho-dermal dysplasia, both linked to WNT10A mutations, which can manifest with shared features like tooth agenesis and abnormalities alongside variable ectodermal defects.

Notable Individuals

, an American character actor born in 1948, lives with hypohidrotic ectodermal dysplasia (HED), a condition that resulted in the absence of sweat glands, scalp and body hair, fingernails, and teeth from birth. His distinctive physical features have shaped his career in film and television, particularly in genres, where he gained prominence for portraying Pluto in (1977) and its 1985 sequel, as well as roles in One Flew Over the Cuckoo's Nest (1975), Weird Science (1985), and Star Trek V: (1989). Berryman's visibility in over 100 projects has helped normalize representations of individuals with rare genetic conditions, indirectly raising public awareness about HED. Beyond acting, he advocates for people with disabilities and environmental causes, including a decade-long residency at a wolf sanctuary in . Aidan Abbott, a resident of Slinger, Wisconsin, was diagnosed as a child with X-linked hypohidrotic ectodermal dysplasia (XLHED), experiencing challenges such as limited sweating, sparse hair, and that complicated access to . By sharing his experiences with insurance denials for essential reconstructive procedures—costing his family thousands—Aidan inspired U.S. Senator to reintroduce the Ensuring Lasting Smiles Act (ELSA) in February 2019 as S.560 in the Senate and H.R.1379 in the House. This bipartisan legislation seeks to require private health insurers to cover medically necessary treatments for congenital anomalies, including those associated with HED, thereby addressing barriers faced by affected individuals. In recognition of his efforts, Aidan received the National Foundation for Ectodermal Dysplasias (NFED) Advocacy Award in 2020 and continues to participate in awareness events as of 2025, contributing to ongoing advocacy for improved care access and research funding.

References

  1. [1]
    Hypohidrotic Ectodermal Dysplasia - GeneReviews® - NCBI Bookshelf
    Apr 28, 2003 · Hypohidrotic ectodermal dysplasia (HED) is characterized by hypotrichosis (sparseness of scalp and body hair), hypohidrosis (reduced ability to sweat), and ...
  2. [2]
    Hypohidrotic ectodermal dysplasia: MedlinePlus Genetics
    Nov 1, 2018 · These disorders result in the abnormal development of ectodermal tissues, particularly the skin, hair, nails, teeth, and sweat glands.
  3. [3]
    Ectodermal Dysplasia - StatPearls - NCBI Bookshelf
    Ectodermal dysplasias (EDs) form a diverse group of inherited disorders characterized by a congenital defect in two or more ectodermal structures.<|control11|><|separator|>
  4. [4]
    Hypohidrotic ectodermal dysplasia | About the Disease | GARD
    Hypohidrotic ectodermal dysplasia (HED) is a genetic skin disease. Common symptoms include sparse scalp and body hair, reduced ability to sweat, and missing ...Missing: definition | Show results with:definition
  5. [5]
    Ectodermal Dysplasia: Background, Pathophysiology, Etiology
    May 29, 2025 · The prevalence of HED, the most common variant, has been estimated to be one case per 100,000 births. The worldwide prevalence of ectodermal ...Background · Pathophysiology · Etiology · Epidemiology
  6. [6]
    Prevalence rates for ectodermal dysplasia syndromes - PubMed
    Aug 9, 2024 · Results: Period prevalence rates were calculated for the following ED disorders: hypohidrotic ectodermal dysplasia, found to be 2.99 per 100, ...
  7. [7]
    Prevalence and Patient Characteristics of Ectodermal Dysplasias in ...
    Mar 13, 2024 · Previous studies have reported prevalence estimates of 10 to 70 cases per 100 000 births. A Danish registry-based study on X-linked hypohidrotic ...
  8. [8]
    Ectodermal Dysplasias - Symptoms, Causes, Treatment | NORD
    The ectodermal dysplasias (EDs) are a heterogeneous group of nearly 100 inherited disorders characterized by anomalies in at least two of the structures ...
  9. [9]
  10. [10]
    Hypohidrotic ectodermal dysplasia: A case report with review and ...
    [2,3] ED syndrome affects both males and females of all races and ethnic groups. The mortality rate is much higher in infancy or early childhood because of ...Missing: global | Show results with:global
  11. [11]
    Natural history of X-linked hypohidrotic ectodermal dysplasia
    Jan 10, 2020 · Prevalence and prevention of severe complications of hypohidrotic ectodermal dysplasia in infancy. Early Hum Dev. 2010;86:397–9. Article ...
  12. [12]
    Hypohidrotic Ectodermal Dysplasia - Symptoms, Causes, Treatment
    Hypohidrotic ectodermal dysplasia (HED) is a rare inherited multisystem disorder that belongs to the group of diseases known as ectodermal dysplasias.
  13. [13]
    Hypohidrotic Ectodermal Dysplasia - NFED
    Hypohidrotic ectodermal dysplasia (HED) is a genetic condition that affects the skin, hair, teeth, nails, and sweat glands. Symptoms of HED may include missing ...Missing: primary reliable
  14. [14]
    Ocular and non-ocular manifestations of hypohidrotic ectodermal ...
    Apr 4, 2011 · Hypohidrotic ectodermal dysplasia (HED) is a group of rare multisystemic genetic syndromes that affects ectodermal structures such as skin, hair ...
  15. [15]
    Ear nose throat manifestations in hypoidrotic ectodermal dysplasia
    Often patients report an increased rate of infections of the upper respiratory tract and of the ear. Nasal obstruction due to the presence of nasal crusting ...
  16. [16]
    Clinical aspects of X-linked hypohidrotic ectodermal dysplasia
    Many suffered severe illness in early childhood and nearly 30% died; many had feeding problems, severe fever, atopic disease, and recurrent respiratory ...Missing: manifestations | Show results with:manifestations
  17. [17]
    Orofacial Features of Hypohidrotic Ectodermal Dysplasia - PMC - NIH
    Hypohidrotic ectodermal dysplasia (HED) is a type of genodermatosis characterized by the abnormal development of sweat glands, teeth, and hair.Missing: definition | Show results with:definition
  18. [18]
    Thermoregulation in Ectodermal Dysplasia: A Case Series - NIH
    Nov 15, 2019 · The lack of functioning sweat glands in those affected with ED leads to high infant mortality and frequent complaints of hyperthermia.Missing: hyperpyrexia dysregulation
  19. [19]
    Role of ectodysplasin signalling in middle ear and nasal pathology ...
    Apr 25, 2019 · Loss of signalling due to mutation of genes encoding any component of this linear pathway leads to hypohidrotic ectodermal dysplasia (HED), ...
  20. [20]
    X-chromosome inactivation: role in skin disease expression - PubMed
    In X-linked skin disorders, Lyonization usually gives rise to a mosaic pattern, as manifest by the appearance of the lines of Blaschko.
  21. [21]
    EDA gene: MedlinePlus Genetics
    Nov 1, 2018 · Genomic Location. The EDA gene is found on the X chromosome. Related Health Topics. Genes and Gene Therapy · Genetic Disorders. MEDICAL ...
  22. [22]
    ECTODERMAL DYSPLASIA 1, HYPOHIDROTIC, X-LINKED; XHED
    ... gene encoding ectodysplasin-A (EDA; 300451) on chromosome Xq13. ▽ Description. Some ectodermal dysplasias are here classified as congenital disorders ...
  23. [23]
    A Novel Ectodysplasin a Gene mutation of X-Linked Hypohidrotic ...
    Jun 25, 2024 · To date, over 300 mutations inEDA have been identified, including missense, nonsense, splicing mutations, gross deletions, smallindels, small ...
  24. [24]
    Functional and clinical analysis of five EDA variants associated with ...
    Jul 17, 2022 · Mutations leading to X-linked hypohidrotic ectodermal dysplasia affect three major functional domains in the tumor necrosis factor family ...
  25. [25]
    A novel EDA1 missense mutation in X-linked hypohidrotic... - Medicine
    We screened the coding sequence of the EDA gene and identified four EDA1 missense mutations, as well as a novel missense mutation (c.1046C>T) in an exon of EDA1 ...
  26. [26]
    Gene Mutations of the Three Ectodysplasin Pathway Key Players ...
    Sep 8, 2021 · The four aforementioned genes contribute to approximately 90% of HED cases in Caucasians, whilst 10% of cases remain unidentified [6].
  27. [27]
    Molecular basis of hypohidrotic ectodermal dysplasia: an update
    Consequently, due to disturbed thermal perception, the patients suffer from overheating (hyperthermia), particularly in the Summer. Dentition abnormalities ...Missing: hyperpyrexia temperature<|separator|>
  28. [28]
  29. [29]
    Structural insights into pathogenic mechanism of hypohidrotic ...
    Feb 11, 2023 · Mutations of EDA have long been known to cause X‐linked hypohidrotic dysplasia ... (hypohidrotic) ectodermal dysplasia is caused by mutation ...
  30. [30]
    WNT10A mutation causes ectodermal dysplasia by impairing ...
    Jun 7, 2017 · Human WNT10A mutations are associated with developmental tooth abnormalities and adolescent onset of a broad range of ectodermal defects.
  31. [31]
    The fundamentals of WNT10A - PMC - PubMed Central - NIH
    WNT10A is a secreted glycoprotein that is involved in signaling pathways essential to ectodermal organogenesis and tissue regeneration.
  32. [32]
    Embryology, Epidermis - StatPearls - NCBI Bookshelf - NIH
    The epidermis begins developing from the surface ectoderm at the end of the fourth week of life as the neural tube separates from the overlying ectoderm.Missing: timing appendages
  33. [33]
    Molecular aspects of hypohidrotic ectodermal dysplasia - Mikkola
    Aug 13, 2009 · The defects in Eda, Edar, and Edaradd mutant mice are highly similar to those seen in humans and include tooth and hair abnormalities. Over 20 ...
  34. [34]
    Ectodysplasin signalling deficiency in mouse models of hypohidrotic ...
    Mouse models with HED also carry Eda, Edar or Edaradd mutations and have defects that map to the same structures. Patients with HED have ear, nose and throat ...
  35. [35]
  36. [36]
  37. [37]
    Rare X-Linked Hypohidrotic Ectodermal Dysplasia in Females ...
    The goal of this study was to identify the pathogenic gene variants in female patients with severe X-linked hypohidrotic ectodermal dysplasia (XLHED).<|control11|><|separator|>
  38. [38]
    Mutational spectrum in 101 patients with hypohidrotic ectodermal ...
    Jun 16, 2016 · Pathogenic mutations were detected in 101 subjects with HED, affecting EDA, EDA1R and EDARADD in 88%, 9% and 3% of the cases, respectively, and ...
  39. [39]
  40. [40]
    WNT10A Mutations Are a Frequent Cause of a Broad Spectrum of ...
    WNT10A mutations are a frequent cause of ectodermal dysplasia and were found in about 9% of an unselected patient cohort.Missing: keratoderma | Show results with:keratoderma
  41. [41]
    Hypohidrotic ectodermal dysplasia - Orphanet
    Hypohidrotic ectodermal dysplasia ; Prevalence: 1-9 / 100 000 ; Inheritance: Autosomal dominant, Autosomal recessive, X-linked recessive ; Age of onset: Infancy, ...<|separator|>
  42. [42]
    The Ectodermal Dysplasias-Burden of Disease Score - NIH
    Aug 30, 2023 · Tooth anomalies and hypohidrosis have negative impact on quality of life. The “burden” assesses disability, social integration, home life and ...
  43. [43]
    Ectodermal Dysplasia Panel - Genetic testing for - Blueprint Genetics
    Nov 13, 2023 · X-linked hypohidrotic ectodermal dysplasia (HED) is caused by EDA mutations and explain 75%-95% of familial HED and 50% of sporadic cases. HED ...
  44. [44]
    Ectodermal Dysplasia Workup - Medscape Reference
    May 29, 2025 · Fetal skin biopsy may help identify the presence of decreased numbers of eccrine sweat glands for antenatal diagnosis of hypohidrotic ectodermal ...Missing: differential | Show results with:differential
  45. [45]
    Standards and Guidelines for the Interpretation of Sequence Variants
    The variant classification approach described in this paper is meant to be applicable to variants in all Mendelian genes whether identified by single gene tests ...
  46. [46]
    Improved definition of carrier status in X-linked hypohidrotic ...
    The detection of carriers of the X-linked disorder hypohidrotic ectodermal dysplasia is problematic because of random X-Inactivation; the diagnosis was ...
  47. [47]
    Invitae Ectodermal Dysplasia and Related Disorders Panel
    The Invitae Ectodermal Dysplasia and Related Disorders Panel analyzes genes that are associated with disorders affecting ectodermal tissues including skin, ...
  48. [48]
    Discover More with GeneDx - Experts in Genome & Exome Testing
    When patients and families come to you for answers, find them with GeneDx genomic testing. We are experts in genome and exome sequencing.Contact Us · Ordering GeneDx genetic testing · FAQs · Genetic testing 101
  49. [49]
    2025 billing assurance and patient program updates - Invitae
    The enhanced financial program offers flat rates of $250 for cancer and $299 for rare disease. New tests for unaffected patients are $250 and $299. Cost ...
  50. [50]
    How long will it take to receive test results? - GeneDx®
    The turnaround time will vary depending on the specific test requested. Please see the specific test details page via our test catalog for additional ...
  51. [51]
    Ectodermal Dysplasia Treatment & Management
    May 29, 2025 · For patients with anhidrosis/hypohidrosis, air conditioning at home, school, and work is advisable. Frequent consumption of cool liquids should ...
  52. [52]
    Improving the Quality of Life in Patients With Hypohidrotic ...
    May 7, 2024 · Recommended measures include ensuring the availability of chilled water, using air conditioning, dressing in breathable fabrics, and limiting ...
  53. [53]
    None
    ### Cooling Strategies for Hypohidrotic Ectodermal Dysplasia (HED)
  54. [54]
    [PDF] Treatment Guide for Skin Issues in Ectodermal Dysplasias
    Swimming is an ideal activity for people with ectodermal dysplasias that include hypohidrosis because cool water reduces the body heat produced by exercise. ...
  55. [55]
    None
    ### Eye Care Measures for Dry Eyes in Ectodermal Dysplasias
  56. [56]
    Anhidrotic (hypohidrotic) ectodermal dysplasia - Dermatology Advisor
    Mar 13, 2019 · The differential diagnosis includes ichthyosis; the neonatal finding of diffuse scaly erythema can be mistaken for an inherited ichthyosis. ...
  57. [57]
    Dental Management of Persons with Ectodermal Dysplasia
    The goals of dental treatment for individuals affected by ED syndromes are to provide an age-appropriate dentition that optimizes chewing function.
  58. [58]
    Dental management of hypohidrotic ectodermal dysplasia - NIH
    This article presents with prosthetic rehabilitation including removable partial and complete denture and implant supported overdenture of two male children
  59. [59]
    X-Linked Hypohidrotic Ectodermal Dysplasia (XLHED): A Case ...
    Jun 13, 2023 · EDA-related HED is inherited in an X-linked manner. On the other hand, HED caused by mutations in EDAR, EDARADD, or WNT10A genes follows an ...
  60. [60]
    [PDF] Dental Management of Persons with Ectodermal Dysplasia
    Oct 4, 2016 · Items that need to be addressed in planning dental treatment include age, psychosocial environment, teeth present, oral hygiene, occlusal ...
  61. [61]
    Growth and Nutrition for Children with Ectodermal Dysplasias - NFED
    Jun 27, 2015 · A well-balanced diet for children with ectodermal dysplasias includes daily servings from all the food groups in amounts sufficient to meet their energy and ...Missing: management | Show results with:management
  62. [62]
    Improvement in the Quality of Life of a Patient of Ectodermal ... - NIH
    A 15-year-old boy, a known case of hypohidrotic ectodermal dysplasia, presented to us in early 2015 with protuberant lips, saddle nose, dental anomalies, fine ...
  63. [63]
    Ectodermal dysplasias: New perspectives on the treatment of so far ...
    Sep 5, 2022 · Drug targeting via the neonatal Fc receptor may cure a so far immedicable genetic deficiency: the life-threatening inability to sweat associated ...
  64. [64]
    A Causal Treatment for X-Linked Hypohidrotic Ectodermal Dysplasia
    Apr 12, 2023 · ... disorder of ectodermal derivatives such as hair, sweat glands, and teeth. The absence of sweat glands and perspiration can evoke life ...<|separator|>
  65. [65]
    A Causal Treatment for X-Linked Hypohidrotic Ectodermal Dysplasia
    Apr 12, 2023 · In our center, treatment with Fc-EDA was considered only for male individuals hemi-zygous for a null mutation in the X-chromosomal gene EDA ( ...
  66. [66]
    Prenatal Correction of X-Linked Hypohidrotic Ectodermal Dysplasia
    Apr 25, 2018 · Genetic deficiency of ectodysplasin A (EDA) causes X-linked hypohidrotic ectodermal dysplasia (XLHED), in which the development of sweat ...
  67. [67]
    Study Details | NCT04980638 | ClinicalTrials.gov - Clinical Trials
    X-linked hypohidrotic ectodermal dysplasia (XLHED) is a rare developmental disease affecting body parts derived from the embryonal ectoderm.
  68. [68]
    Edelife Clinical Trial: A Prenatal Study for XLHED-Affected Boys
    The Edelife Clinical Trial is studying a potential treatment, called ER004, for x-linked hypohidrotic ectodermal dysplasia (XLHED) in unborn boys.
  69. [69]
    Development of tooth regenerative medicine strategies by ...
    Sep 1, 2020 · Targeted molecular therapy could be used to generate teeth in patients with congenital tooth agenesis by stimulating arrested tooth germs.
  70. [70]
    Development of a new antibody drug to treat congenital tooth agenesis
    This study aimed to develop a therapeutic agent promoting teeth regeneration from autologous tissues for congenital tooth agenesis, specifically for hypodontia ...
  71. [71]
    Ectodermal dysplasia: a narrative review of the clinical and ...
    Feb 27, 2025 · Only four genes (EDA1, EDAR, EDARADD, and WNT10A) account for 90% of hypohidrotic/anhidrotic ectodermal dysplasia cases. Hum Mutat. (2011) ...
  72. [72]
  73. [73]
    Ectodermal dysplasias: New perspectives on the treatment of so far ...
    Sep 6, 2022 · Autologous, ex vivo expanded epithelial cells containing limbal stem cells were approved some years ago as an advanced-therapy medicinal product ...Missing: hypodontia | Show results with:hypodontia
  74. [74]
    Combined Preimplantation Genetic Testing for Aneuploidy and ...
    Background: Hypohidrotic ectodermal dysplasia (HED) is a genetic skin condition presenting as hypohidrosis, hypodontia, and hypotrichosis, resulting in an ...
  75. [75]
    Genetic Counseling Helps Make Sense of Ectodermal Dysplasia
    Sep 6, 2016 · The goal of genetic counseling is to help you learn more about the causes of ectodermal dysplasia and how they affect you. Here's what you can ...
  76. [76]
    National Foundation for Ectodermal Dysplasias | NFED
    We empower and connect those touched by ectodermal dysplasias through education, support and research. Find resources, treatment and people like you.<|separator|>
  77. [77]
    An Insight into the Genesis of Hypohidrotic Ectodermal Dysplasia in ...
    Charles Darwin first documented the earliest accessible account of ectodermal dysplasia in English as early as 1840s. However, in the mid-19th century, it ...
  78. [78]
    HEREDITARY ECTODERMAL DYSPLASIA (CONGENITAL ...
    HEREDITARY ECTODERMAL DYSPLASIA (CONGENITAL ECTODERMAL DEFECT): A REPORT OF TWO CASES. A. A. WEECH, M.D.. Author Affiliations. BALTIMORE. From the Harriet Lane ...
  79. [79]
    X-linked anhidrotic (hypohidrotic) ectodermal dysplasia is caused by ...
    We now describe the positional cloning of the gene mutated in EDA. Two exons, separated by a 200-kilobase intron, encode a predicted 135-residue transmembrane ...
  80. [80]
    Cloning of Tabby, the Murine Homolog of the Human EDA Gene
    We have identified a candidate cDNA for the mouse Tabby gene (Ta), which, based on phenotype and syntenic mapping, is postulated to represent the analogous ...Cloning Of Tabby, The Murine... · Results · Materials And Methods
  81. [81]
    Christ–Siemens–Touraine Syndrome: A Rare Case Report - PMC
    Christ–Siemens–Touraine syndrome/hypohidrotic ectodermal dysplasia (HED) is a heterogeneous group of inherited disorders with primary defects in tissues ...
  82. [82]
    Hidrotic Ectodermal Dysplasia 2 - GeneReviews® - NCBI Bookshelf
    Apr 25, 2005 · The clinical features include severely dystrophic nails and thin scalp hair, fine eyebrows and eyelashes, and thin body hair. The associated ...
  83. [83]
    Mutation in WNT10A Is Associated with an Autosomal Recessive ...
    Mutation in WNT10A is associated with an autosomal recessive ectodermal dysplasia: The odonto-onycho-dermal dysplasia.
  84. [84]
    This Day in Horror History: Happy Birthday, Michael Berryman
    Sep 4, 2020 · Berryman was born with hypohidrotic ectodermal dysplasia, a rare ... He is an advocate for environmental protection and lived on a wolf sanctuary ...
  85. [85]
    Michael Berryman: books, biography, latest update - Amazon.com
    Born in 1948, he was diagnosed with a rare genetic disorder called hypohidrotic ectodermal ... He is an advocate for people with disabilities and has used ...
  86. [86]
    ELSA Reintroduced in Congress - NFED
    Feb 26, 2019 · She took action after getting to know Aidan Abbott, a 14-year-old advocate affected by x-linked hypohidrotic ectodermal dysplasia. Aidan ...
  87. [87]
    Teen Advocate Wins Award - NFED
    Jan 2, 2020 · Aidan, who has ectodermal dysplasia, has been sharing his story for years to raise awareness. He, his parents, Becky and Tom, and younger ...
  88. [88]
    Aidan's Story Sparked a Movement When Aidan bravely shared his ...
    Apr 30, 2025 · ... hypohidrotic ectodermal dysplasia (HED), it inspired Senator Tammy Baldwin to write and introduce the Ensuring Lasting Smiles Act (ELSA)—a ...