Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 277
Filtrar
1.
Clin Genet ; 105(5): 581-583, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38379111

RESUMO

A case of mosaic MTOR-associated hemimegalencephaly and hypomelanosis of Ito, died at 33 probably because of sudden unexpected death in epilepsy. Assessment of the variant allele fraction (VAF) in different tissues postmortem showed high variability not correlated with clinical features, representing the most detailed assessment of VAFs in different tissues to date.


Assuntos
Hipopigmentação , Humanos , Hipopigmentação/genética , Alelos , Autopsia , Serina-Treonina Quinases TOR
2.
Lasers Surg Med ; 55(5): 490-502, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37051852

RESUMO

OBJECTIVES: One symptom of hypertrophic scar (HTS) that can develop after burn injury is dyschromia with hyper- and hypopigmentation. There are limited treatments for these conditions. Previously, we showed there is no expression of alpha melanocyte stimulating hormone (α-MSH) in hypopigmented scars, and if these melanocytes are treated with synthetic α-MSH in vitro, they respond by repigmenting. The current study tested the same hypothesis in the in vivo environment using laser-assisted drug delivery (LADD). METHODS: HTSs were created in red Duroc pigs. At Day 77 (pre), they were treated with CO2 fractional ablative laser (FLSR). Synthetic α-MSH was delivered as a topical solution dissolved in  l-tyrosine (n = 6, treated). Control scars received LADD of  l-tyrosine only (n = 2, control). Scars were treated and examined weekly through Week 4. Digital images and punch biopsies of hyper, hypo-, and normally pigmented scar and skin were collected. Digital pictures were analyzed with ImageJ by tracing the area of hyperpigmentation. Epidermal sheets were obtained from punch biopsies through dispase separation and RNA was isolated. qRT-PCR was run for melanogenesis-related genes: tyrosinase (TYR), tyrosinase-related protein-1 (TYRP1), and dopachrome tautomerase (DCT). Two-way ANOVA with multiple comparisons and Dunnett's correction compared the groups. RESULTS: The areas of hyperpigmentation were variable before treatment. Therefore, data is represented as fold-change where each scar was normalized to its own pre value. Within the LADD of NDP α-MSH + l-tyrosine group, hyperpigmented areas gradually increased each week, reaching 1.3-fold over pre by Week 4. At each timepoint, area of hyperpigmentation was greater in the treated versus the control (1.04 ± 0.05 vs. 0.89 ± 0.08, 1.21 ± 0.07 vs. 0.98 ± 0.24, 1.21 ± 0.08 vs. 1.04 ± 0.11, 1.28 ± 0.11 vs. 0.94 ± 0.25; fold-change from pre-). Within the treatment group, pretreatment, levels of TYR were decreased -17.76 ± 4.52 below the level of normal skin in hypopigmented scars. After 1 treatment, potentially due to laser fractionation, the levels decreased to -43.49 ± 5.52. After 2, 3, and 4 treatments, there was ever increasing levels of TYR to almost the level of normally pigmented skin (-35.74 ± 15.72, -23.25 ± 6.80, -5.52 ± 2.22 [p < 0.01, Week 4]). This pattern was also observed for TYRP1 (pre = -12.94 ± 1.82, Week 1 = -48.85 ± 13.25 [p < 0.01], Weeks 2, 3, and 4 = -34.45 ± 14.64, -28.19 ± 4.98, -6.93 ± 3.05 [p < 0.01, Week 4]) and DCT (pre = -214.95 ± 89.42, Week 1 = -487.93 ± 126.32 [p < 0.05], Weeks 2, 3, and 4 = -219.06 ± 79.33, -72.91 ± 20.45 [p < 0.001], -76.00 ± 24.26 [p < 0.001]). Similar patterns were observed for scars treated with LADD of  l-tyrosine alone without NDP α-MSH. For each gene, in hyperpigmented scar, levels increased at Week 4 of treatment compared to Week 1 (p < 0.01). CONCLUSIONS: A clinically-relevant FLSR treatment method can be combined with topical delivery of synthetic α-MSH and l-tyrosine to increase the area of pigmentation and expression of melanogenesis genes in hypopigmented HTS. LADD of  l-tyrosine alone leads to increased expression of melanogenesis genes. Future studies will aim to optimize drug delivery, timing, and dosing.


Assuntos
Cicatriz Hipertrófica , Hiperpigmentação , Hipopigmentação , Lasers de Gás , Animais , Suínos , Cicatriz Hipertrófica/tratamento farmacológico , Cicatriz Hipertrófica/genética , Cicatriz Hipertrófica/patologia , Tirosina , alfa-MSH/uso terapêutico , alfa-MSH/metabolismo , Preparações Farmacêuticas , Pigmentação , Hipopigmentação/tratamento farmacológico , Hipopigmentação/genética , Hiperpigmentação/tratamento farmacológico , Hiperpigmentação/genética , Lasers de Gás/uso terapêutico , Melaninas/metabolismo
3.
Ann Dermatol Venereol ; 150(2): 109-113, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36990862

RESUMO

BACKGROUND: Pigmentary mosaicism (PM), also known as Blaschkoid dyspigmentation, is a rare pigmentary anomaly. Although several case reports have been published describing extracutaneous manifestations associated with PM, there are very few studies on the clinical characteristics of patients with PM. AIM: To describe the clinical characteristics of patients with PM. PATIENTS AND METHODS: This descriptive cross-sectional study was conducted among 47 children examined by a dermatologist and a pediatrician. The pattern and location of the PM, type of pigmentation and extracutaneous manifestations were documented. RESULTS: The most common pattern of PM was narrow-band PM, followed by broad-band and checkerboard patterns. The trunk was the most affected region, followed by the legs and arms. PM manifested as hypopigmentation in 51.1% of cases, as hyperpigmentation in 27.6%, and as hypo/hyperpigmentation in 21.2%. Accompanying diseases were present in 40.4% of patients: neuropsychiatric diseases were the most common, followed by endocrinological or hematological diseases and growth/developmental delay. CONCLUSION: PM has been associated with several extracutaneous findings but there is still some debate whether these associations reflect different PM phenotypes or whether they are simply coincidental. Our study suggests that extracutaneous involvement in PM patients is frequent, thus warranting careful examination of PM patients.


Assuntos
Hiperpigmentação , Hipopigmentação , Humanos , Mosaicismo , Estudos Transversais , Hiperpigmentação/genética , Hipopigmentação/genética , Fenótipo
5.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(12): 1360-1365, 2022 Dec 10.
Artigo em Chinês | MEDLINE | ID: mdl-36453959

RESUMO

OBJECTIVE: To explore the clinical phenotype and genetic basis for a Chinese pedigree affected with familial progressive hyperpigmentation and hypopigmentation (FPHH). METHODS: Clinical data and family history for a child with FPHH were collected. Peripheral blood samples were collected from the child, his parents and two sisters. Following the extraction of DNA, high-throughput sequencing was carried out to screen for genetic variant associated with the disease. Candidate variant was verified by Sanger sequencing of his family members. RESULTS: The main clinical features of the proband have included progressive hyperpigmentation and hypopigmentation. High-throughput sequencing revealed that he has harbored a heterozygous c.105T>A (p.Asn35Lys) variant of the KITLG gene, which was unreported previously. Sanger sequencing confirmed that the variant has co-segregated with the disease phenotype in his pedigree. CONCLUSION: For infants with progressive skin pigmentation and hypopigmentation spots, FPHH should be suspected. The heterozygous c.105T>A (p.Asn35Lys) variant of the KITLG gene probably underlay the FPHH in this pedigree.


Assuntos
Hiperpigmentação , Hipopigmentação , Masculino , Humanos , Linhagem , Hipopigmentação/genética , Fenótipo , Hiperpigmentação/genética , China
6.
Exp Dermatol ; 31(12): 1927-1931, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35960249

RESUMO

Inherited epidermolysis bullosa (EB) simplex is a heterogeneous group of skin fragility disorders caused by mutations in genes encoding cell-cell or cell-matrix adhesion proteins. A recently identified, rare subtype of EB simplex is due to bi-allelic mutations in the EXPH5 gene, which encodes exophilin5, an effector protein of the Rab27B GTPase involved in intracellular vesicle trafficking and exosome secretion. The EXPH5 EB subtype is characterized by early-onset skin blisters and scars, mainly on extremities, and varying degrees of pigmentary alterations. Here, we present a 31-year-old female with diffuse guttate hypopigmentation on the trunk and extremities since early childhood, with no apparent blisters or scars. We employed whole exome sequencing of germline DNA extracted from the patient's leukocytes to determine the genetic aetiology of the phenotype. A novel homozygous variant in EXPH5, c.1153C>T causing a premature stop codon at amino acid Glutamine 385, was identified. Histologic examination after skin pricking disclosed focal keratinocyte detachment typical to EB. Additionally, we identified a deleterious-predicted variant in ENPP1, a gene associated with disturbed transfer of melanosomes to keratinocytes in Cole disease. Our report expands the clinical spectrum of inherited EB simplex with a possible di-genic synergism contributing to co-presentation with guttate leukoderma.


Assuntos
Epidermólise Bolhosa Simples , Epidermólise Bolhosa , Hipopigmentação , Feminino , Pré-Escolar , Humanos , Epidermólise Bolhosa Simples/genética , Epidermólise Bolhosa Simples/patologia , Cicatriz/patologia , Vesícula/patologia , Queratinócitos/metabolismo , Hipopigmentação/genética , Epidermólise Bolhosa/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
7.
BMJ Case Rep ; 15(6)2022 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-35725288

RESUMO

Peripheral demyelinating neuropathy, central dysmyelinating leukodystrophy, Waardenburg syndrome and Hirschsprung disease (PCWH) is a rare manifestation of Waardenburg-Shah syndrome associated with mutations in the SOX10 gene. The phenotypic expression is variable, thus presenting a diagnostic challenge. Clinical manifestations of PCWH may mimic other neurocutaneous syndromes. A thorough history, careful physical examination, appropriate imaging studies and an index of suspicion are needed to diagnose this condition. We describe an adolescent girl with skin hypopigmentation and blue irides associated with sensorineural hearing loss, Hirschsprung disease, as well as seizures with neurological signs, and discuss the challenges in diagnosing PCWH.


Assuntos
Doenças Desmielinizantes , Doença de Hirschsprung , Hipopigmentação , Síndrome de Waardenburg , Adolescente , Criança , Doenças Desmielinizantes/diagnóstico , Feminino , Doença de Hirschsprung/complicações , Doença de Hirschsprung/diagnóstico , Doença de Hirschsprung/genética , Humanos , Hipopigmentação/diagnóstico , Hipopigmentação/genética , Mutação , Fatores de Transcrição SOXE/genética , Síndrome de Waardenburg/complicações , Síndrome de Waardenburg/diagnóstico , Síndrome de Waardenburg/genética
8.
Hum Genet ; 141(11): 1771-1784, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35503477

RESUMO

Pigmentary mosaicism of the Ito type, also known as hypomelanosis of Ito, is a neurocutaneous syndrome considered to be predominantly caused by somatic chromosomal mosaicism. However, a few monogenic causes of pigmentary mosaicism have been recently reported. Eleven unrelated individuals with pigmentary mosaicism (mostly hypopigmented skin) were recruited for this study. Skin punch biopsies of the probands and trio-based blood samples (from probands and both biological parents) were collected, and genomic DNA was extracted and analyzed by exome sequencing. In all patients, plausible monogenic causes were detected with somatic and germline variants identified in five and six patients, respectively. Among the somatic variants, four patients had MTOR variant (36%) and another had an RHOA variant. De novo germline variants in USP9X, TFE3, and KCNQ5 were detected in two, one, and one patients, respectively. A maternally inherited PHF6 variant was detected in one patient with hyperpigmented skin. Compound heterozygous GTF3C5 variants were highlighted as strong candidates in the remaining patient. Exome sequencing, using patients' blood and skin samples is highly recommended as the first choice for detecting causative genetic variants of pigmentary mosaicism.


Assuntos
Hipopigmentação , Mosaicismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Humanos , Hipopigmentação/genética , Serina-Treonina Quinases TOR/genética , Ubiquitina Tiolesterase/genética
9.
J Eur Acad Dermatol Venereol ; 36(9): 1606-1611, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35543077

RESUMO

BACKGROUND: Pathogenic variants in KITLG, a crucial protein involved in pigmentation and neural crest cell migration, cause non-syndromic hearing loss, Waardenburg syndrome type 2, familial progressive hyperpigmentation and familial progressive hyper- and hypopigmentation, all of which are inherited in an autosomal dominant manner. OBJECTIVES: To describe the genotypic and clinical spectrum of biallelic KITLG-variants. METHODS: We used a genotype-first approach through the GeneMatcher data sharing platform to collect individuals with biallelic KITLG variants and reviewed the literature for overlapping reports. RESULTS: We describe the first case series with biallelic KITLG variants; we expand the known hypomelanosis spectrum to include a 'sock-and-glove-like', symmetric distribution, progressive repigmentation and generalized hypomelanosis. We speculate that KITLG biallelic loss-of-function variants cause generalized hypomelanosis, whilst variants with residual function lead to a variable auditory-pigmentary disorder mostly reminiscent of Waardenburg syndrome type 2 or piebaldism. CONCLUSIONS: We provide consolidating evidence that biallelic KITLG variants cause a distinct auditory-pigmentary disorder. We evidence a significant clinical variability, similar to the one previously observed in KIT-related piebaldism.


Assuntos
Perda Auditiva Neurossensorial , Hiperpigmentação , Hipopigmentação , Piebaldismo , Perda Auditiva Neurossensorial/genética , Humanos , Hipopigmentação/genética , Fator de Células-Tronco , Síndrome de Waardenburg
10.
Hum Mutat ; 43(9): 1183-1200, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35475527

RESUMO

ENPP1 encodes ENPP1, an ectonucleotidase catalyzing hydrolysis of ATP to AMP and inorganic pyrophosphate (PPi), and an endogenous plasma protein physiologically preventing ectopic calcification of connective tissues. Mutations in ENPP1 have been reported in association with a range of human genetic diseases. In this mutation update, we provide a comprehensive review of all the pathogenic variants, likely pathogenic variants, and variants of unknown significance in ENPP1 associated with three autosomal recessive disorders-generalized arterial calcification of infancy (GACI), autosomal recessive hypophosphatemic rickets type 2 (ARHR2), and pseudoxanthoma elasticum (PXE), as well as with a predominantly autosomal dominant disorder-Cole disease. The classification of all variants is determined using the latest ACMG guidelines. A total of 140 ENPP1 variants were curated consisting of 133 previously reported variants and seven novel variants, with missense variants being the most prevalent (70.0%, 98/140). While the pathogenic variants are widely distributed in the ENPP1 gene of patientsgen without apparent genotype-phenotype correlation, eight out of nine variants associated with Cole disease are confined to the somatomedin-B-like (SMB) domains critical for homo-dimerization of the ENPP1 protein.


Assuntos
Hipopigmentação , Diester Fosfórico Hidrolases , Pirofosfatases , Raquitismo Hipofosfatêmico , Calcificação Vascular , Humanos , Hipopigmentação/genética , Mutação , Diester Fosfórico Hidrolases/genética , Pirofosfatases/genética , Raquitismo Hipofosfatêmico/complicações , Raquitismo Hipofosfatêmico/genética , Calcificação Vascular/genética
11.
Pediatr Dermatol ; 39(2): 281-287, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35178721

RESUMO

RHOA-related neuroectodermal syndrome is characterised by linear skin hypopigmentation along Blaschko's lines associated with alopecia, leukoencephalopathy, facial and limb hypoplasia, and ocular, dental, and acral anomalies. Herein, we report a patient with patterned cutaneous hypopigmentation with a similar phenotype due to a novel postzygotic RHOA variant (c.210G>T; p.Arg70Ser). This illustrates that the complexity of the orchestration of morphogenesis and organogenesis can be affected by different variants in the same gene.


Assuntos
Hipopigmentação , Mosaicismo , Humanos , Hipopigmentação/genética , Hipopigmentação/patologia , Fenótipo , Pele/patologia , Proteína rhoA de Ligação ao GTP/genética
12.
Pigment Cell Melanoma Res ; 35(2): 212-219, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34870899

RESUMO

Oculocutaneous albinism (OCA) 6 is a non-syndromic type of OCA that has distinct ocular symptoms and variable cutaneous hypopigmentation. The causative gene of OCA6 is SLC24A5, which encodes NCKX5, a K+ -dependent Na+ /Ca2+ exchanger 5. NCKX5 is involved in the maturation of melanosomes, but its function is still unclear. In this study, we characterized a Japanese patient with OCA6. Genetic analysis revealed compound heterozygous variants in SLC24A5, c.590 + 1dupG, and c.598G>A (p.G200R). To clarify the functional significance of the missense variant, we generated a knock-in (KI) mouse model carrying the mouse homolog of the G200R variant using the CRISPR/Cas9 system. Chemical analysis showed decreased amounts of eumelanin in the hair and skin of KI mice, while levels of benzothiazine units in pheomelanin were significantly increased in their hair. Retinal pigment was also decreased in KI mice. Notably, a histopathologic study revealed a significant pigment loss in the retinal pigment epithelium (RPE) but not in the choroid. Immunohistochemically, the expression of NCKX5 in the RPE was decreased but was maintained in the choroid of KI mice. These findings could explain the difference in phenotypic severity between eye symptoms and hypopigmentation in the skin/hair.


Assuntos
Albinismo Oculocutâneo , Hipopigmentação , Epitélio Pigmentado da Retina , Trocador de Sódio e Cálcio , Albinismo Oculocutâneo/genética , Animais , Humanos , Hipopigmentação/genética , Japão , Camundongos , Epitélio Pigmentado da Retina/metabolismo , Trocador de Sódio e Cálcio/metabolismo
13.
Mol Genet Genomic Med ; 9(12): e1841, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34716665

RESUMO

Familial Progressive Hyper- and Hypopigmentation is a pigmentary disorder characterized by a mix of hypo- and hyperpigmented lesions, café-au-lait spots and hypopigmented ash-leaf macules. The disorder was previously linked to KITLG and various mutations have been reported to segregate in different families. Furthermore, association between KITLG mutations and malignancies was also suggested. Exome and SANGER sequencing were performed for identification of KITLG mutations. Functional in silico analyses were additionally performed to assess the findings. We identified a de novo mutation in exon 4 of KITLG gene causing NM_000899.4:c.[329A>T] (chr12:88912508A>T) leading to NP_000890.1:p.(Asp110Val) substitution in the 3rd alpha helix. It was predicted as pathogenic, located in a conserved region and causing an increase in hydrophobicity in the KITLG protein. Our findings clearly confirm an additional hot spot of KITLG mutations in the 3rd alpha helix, which very likely increases the risk of malignancies. To our knowledge the present study provides the strongest evidence of association of the KITLG mutation with both Familial Progressive Hyper- and Hypopigmentation and malignancy due to its' location on somatic cancer mutation locus. Additionally we also address difficulties with classification of the unique phenotype and propose a subtype within broader diagnosis.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Hiperpigmentação/diagnóstico , Hiperpigmentação/genética , Hipopigmentação/diagnóstico , Hipopigmentação/genética , Mutação , Fator de Células-Tronco/genética , Sequência de Aminoácidos , Estudos de Associação Genética/métodos , Humanos , Imuno-Histoquímica , Linhagem , Fenótipo , Análise de Sequência de DNA , Pele/patologia , Fator de Células-Tronco/química
14.
J Dermatol Sci ; 104(1): 48-54, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34551863

RESUMO

BACKGROUND: Amyloidosis cutis dyschromica (ACD) is a rare form of primary cutaneous amyloidosis featured by reticulate dotted hypo- and hyperpigmentation. Recently, loss-of-function mutations in GPNMB, encoding glycoprotein (transmembrane) nonmetastatic melanoma protein B, were found in autosomal-recessive or semi-dominant ACD. OBJECTIVE: This study aims to detect the genetic defect underlying ACD in nine separate cases and to investigate the functional consequences of the mutants. METHODS: Nine ACD cases were collected including eight with autosomal-recessive pattern and one with autosomal-dominant pattern. Whole-exome sequencing or Sanger sequencing of the GPNMB gene was performed to detect the pathogenic mutations. Haplotype analysis was employed to determine the origin of mutation c.565C > T using adjacent highly polymorphic SNPs. Immunoblotting and subcellular localization assessments were performed to evaluate the expression of the mutants using HEK293 cells transfected with the GPNMB constructs. RESULTS: We detected four recurrent mutations (c.393 T > G, p.Y131*; c.565C > T, p.R189*; c.1056delT, p.P353Lfs*20; c.1238 G > C, p.C413S) and two novel mutations (c.935delA, p.N312Tfs*4; c.969 T > A, p.C323*) in GPNMB. Mutation c.565C > T found in six separate ACD cases shared a common haplotype. The two novel mutations caused a decreased abundance of truncated proteins. The c.1238 G > C mutation, which was detected in the autosomal-dominant case, caused abnormal reticular subcellular localization of the protein. A major percentage of wildtype changed its expression pattern when co-expressed with this mutant. CONCLUSIONS: Our findings proved that the recurrent mutation c.565C > T originated from a founder effect. The autosomal-dominant ACD associated mutation p.C413S played its pathogenic role through a dominant-negative effect on wild-type GPNMB. This study expands the genotype and inherited modes of ACD and improves our understanding of the pathogenesis of this disorder.


Assuntos
Amiloidose Familiar/genética , Efeito Fundador , Hiperpigmentação/genética , Hipopigmentação/genética , Glicoproteínas de Membrana/genética , Dermatopatias Genéticas/genética , Adolescente , Idade de Início , Amiloidose Familiar/diagnóstico , Amiloidose Familiar/patologia , Criança , Análise Mutacional de DNA , Feminino , Células HEK293 , Humanos , Hiperpigmentação/diagnóstico , Hiperpigmentação/patologia , Hipopigmentação/diagnóstico , Hipopigmentação/patologia , Padrões de Herança , Masculino , Mutação , Linhagem , Pele/patologia , Dermatopatias Genéticas/diagnóstico , Dermatopatias Genéticas/patologia , Sequenciamento do Exoma
17.
Genet Med ; 23(8): 1484-1491, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33833411

RESUMO

PURPOSE: Hypomelanosis of Ito (HI) is a skin marker of somatic mosaicism. Mosaic MTOR pathogenic variants have been reported in HI with brain overgrowth. We sought to delineate further the pigmentary skin phenotype and clinical spectrum of neurodevelopmental manifestations of MTOR-related HI. METHODS: From two cohorts totaling 71 patients with pigmentary mosaicism, we identified 14 patients with Blaschko-linear and one with flag-like pigmentation abnormalities, psychomotor impairment or seizures, and a postzygotic MTOR variant in skin. Patient records, including brain magnetic resonance image (MRI) were reviewed. Immunostaining (n = 3) for melanocyte markers and ultrastructural studies (n = 2) were performed on skin biopsies. RESULTS: MTOR variants were present in skin, but absent from blood in half of cases. In a patient (p.[Glu2419Lys] variant), phosphorylation of p70S6K was constitutively increased. In hypopigmented skin of two patients, we found a decrease in stage 4 melanosomes in melanocytes and keratinocytes. Most patients (80%) had macrocephaly or (hemi)megalencephaly on MRI. CONCLUSION: MTOR-related HI is a recognizable neurocutaneous phenotype of patterned dyspigmentation, epilepsy, intellectual deficiency, and brain overgrowth, and a distinct subtype of hypomelanosis related to somatic mosaicism. Hypopigmentation may be due to a defect in melanogenesis, through mTORC1 activation, similar to hypochromic patches in tuberous sclerosis complex.


Assuntos
Hipopigmentação , Megalencefalia , Humanos , Hipopigmentação/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Mosaicismo , Fenótipo , Serina-Treonina Quinases TOR/genética
18.
Mol Med Rep ; 23(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33786625

RESUMO

Hereditary leukonychia (HL) is a rare nail dystrophy disease, and several different clinical manifestations and mutations in the phospholipase C δ 1 (PLCD1) gene have been reported. The present study reports on one Chinese family and one sporadic case of with HL. The family members exhibited an autosomal dominant pattern of inheritance with the involvement of all the fingers and toenails in all the patients. Of interest, most of the affected members had koilonychia during their childhood. Thus, the present study first used gene mapping with an aim to identify the pathogenic gene underlying koilonychia. Through genome­wide linkage analysis, the pathogenic area of koilonychia was identified on chromosome 3 with multipoint Log of Odds scores >2. A novel pathogenic mutation c.1384G>A (p.E462K) was identified in the PLCD1 gene in all the patients in the family, which confirmed the diagnosis of hereditary leukonychia. A novel mutation c.770G>A (p.R257H) was also detected in one sporadic case of leukonychia. On the basis of these findings and of previous studies, it is suggested that hereditary leukonychia may initially present as koilonychia, whereas hereditary koilonychia does not progress to leukonychia. Moreover, the present study identified two pathogenic variants of the PLCD1 associated with hereditary leukonychia, and highlights the significance of genetic diagnosis.


Assuntos
Hipopigmentação/genética , Doenças da Unha/congênito , Unhas Malformadas/genética , Fosfolipase C delta/genética , Adulto , Criança , Feminino , Humanos , Hipopigmentação/patologia , Masculino , Mutação de Sentido Incorreto , Doenças da Unha/genética , Doenças da Unha/patologia , Unhas Malformadas/patologia , Linhagem
19.
PLoS One ; 16(3): e0248985, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33765043

RESUMO

There are limited treatments for dyschromia in burn hypertrophic scars (HTSs). Initial work in Duroc pig models showed that regions of scar that are light or dark have equal numbers of melanocytes. This study aims to confirm melanocyte presence in regions of hypo- and hyper-pigmentation in an animal model and patient samples. In a Duroc pig model, melanocyte presence was confirmed using en face staining. Patients with dyschromic HTSs had demographic, injury details, and melanin indices collected. Punch biopsies were taken of regions of hyper-, hypo-, or normally pigmented scar and skin. Biopsies were processed to obtain epidermal sheets (ESs). A subset of ESs were en face stained with melanocyte marker, S100ß. Melanocytes were isolated from a different subset. Melanocytes were treated with NDP α-MSH, a pigmentation stimulator. mRNA was isolated from cells, and was used to evaluate gene expression of melanin-synthetic genes. In patient and pig scars, regions of hyper-, hypo-, and normal pigmentation had significantly different melanin indices. S100ß en face staining showed that regions of hyper- and hypo-pigmentation contained the same number of melanocytes, but these cells had different dendricity/activity. Treatment of hypo-pigmented melanocytes with NDP α-MSH produced melanin by microscopy. Melanin-synthetic genes were upregulated in treated cells over controls. While traditionally it may be thought that hypopigmented regions of burn HTS display this phenotype because of the absence of pigment-producing cells, these data show that inactive melanocytes are present in these scar regions. By treating with a pigment stimulator, cells can be induced to re-pigment.


Assuntos
Queimaduras/patologia , Cicatriz Hipertrófica/patologia , Hipopigmentação/patologia , Melanócitos/patologia , alfa-MSH/metabolismo , Adulto , Animais , Biópsia , Vias Biossintéticas , Queimaduras/complicações , Queimaduras/genética , Células Cultivadas , Cicatriz Hipertrófica/complicações , Cicatriz Hipertrófica/genética , Humanos , Hiperpigmentação/complicações , Hiperpigmentação/patologia , Hipopigmentação/complicações , Hipopigmentação/genética , Masculino , Melaninas/biossíntese , Melanócitos/metabolismo , Pessoa de Meia-Idade , Fenótipo , Pigmentação , Suínos , Regulação para Cima/genética , Adulto Jovem
20.
Pediatr Dermatol ; 38(3): 637-639, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33715178

RESUMO

Partial trisomy-13 mosaicism (PT13M) is a rare condition. Among its possible associated cutaneous features, phylloid hypomelanosis (PH), characterized by leaf-like macules reminiscent of floral ornaments in the form of round or oval spots and patches and oblong lesions, is typical. Two cases of PH associated with hidradenitis suppurativa (HS) have been already reported in the literature. We report a third child with PH due to PT13M associated with HS-like lesions limited to hypomelanotic regions. We hypothesize that follicular occlusion genes may be located in the duplicated part of chromosome 13.


Assuntos
Hidradenite Supurativa , Hipopigmentação , Criança , Humanos , Hipopigmentação/genética , Mosaicismo , Pele , Trissomia/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA