Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
J Hum Genet ; 66(8): 825-829, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33526817

RESUMO

Hypomelanosis of Ito (HMI) is part of a neuroectodermal syndrome characterized by distinctive skin manifestations with or without multisystemic involvements. In our undiagnosed diseases program, we have encountered a 3-year-old girl presenting with characteristic skin hypopigmentation suggesting HMI and developmental delay. An exome and genome approach utilizing next-generation sequencing revealed a heterozygous de novo frameshift variant in the KIF13A gene, i.e., NM_022113.6: c.2357dupA, resulting in nonsense-mediated decay. The low mutant allelic ratio suggested that the mutation has occurred postzygotically leading to embryonic mosaicism. Functionally, K1F3A regulates cell membrane blebbing and migration of neural crest cells by controlling recycling of RHOB to the plasma membrane and is also involved in melanosome biogenesis. Importantly, hypopigmentation of the skin has been reported in chr 6p22.3-p23 microdeletion syndrome supporting the association of KIF13A haploinsufficiency with the novel neuroectodermal syndrome. With the increased availability of genome sequencing, we envisage more genetic causes of HMI will be identified in the future.


Assuntos
Cromossomos Humanos Par 6 , Mutação da Fase de Leitura , Hipopigmentação/genética , Cinesinas/genética , Síndromes Neurocutâneas/genética , Zigoto , Pré-Escolar , Transtornos Cromossômicos/genética , Transtornos Cromossômicos/patologia , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mosaicismo/embriologia , Síndromes Neurocutâneas/patologia , Sequenciamento do Exoma
8.
Clin Genet ; 99(4): 572-576, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33410500

RESUMO

We describe an 11-year-old girl with PLACK Syndrome (peeling skin, leukonychia, acral punctate keratosis, cheilitis, and knuckle pads), who was found to have a novel homozygous variant in CAST, the pathogenicity of which was confirmed using blood-derived RNA. There is no established treatment for PLACK syndrome. However, we demonstrate for the first time that this condition is associated with low levels of vitamin A and essential fatty acids, which prompted us to consider a potential treatment strategy. Indeed, we initiated this patient on intravenous lipid infusion (Vitalipid®; an emulsion of fat-soluble vitamins and lipofundin-MCT/LCT 20%) and the response was dramatic. Following the fourth monthly course of treatment, pruritis disappeared and the skin lesions showed remarkable objective improvement. PLACK syndrome is a very rare genodermatosis and only six families have been described to date with pathogenic CAST variants. This is the first report of an objective response to a therapeutic agent, which suggests that PLACK is a potentially treatable condition. The remarkable response we report and the relative safety of the intervention should prompt healthcare providers who care for PLACK syndrome patients to explore this as a potential treatment strategy in future studies.


Assuntos
Dermatite Esfoliativa/tratamento farmacológico , Hipopigmentação/tratamento farmacológico , Doenças da Unha/congênito , Fosfolipídeos/uso terapêutico , Dermatopatias Genéticas/tratamento farmacológico , Óleo de Soja/uso terapêutico , Vesícula/etiologia , Proteínas de Ligação ao Cálcio/genética , Queilite/tratamento farmacológico , Queilite/genética , Criança , Consanguinidade , Dermatite Esfoliativa/genética , Emulsões/administração & dosagem , Emulsões/uso terapêutico , Feminino , Humanos , Hipopigmentação/genética , Infusões Intravenosas , Ceratose/tratamento farmacológico , Ceratose/genética , Doenças da Unha/tratamento farmacológico , Doenças da Unha/genética , Linhagem , Fosfolipídeos/administração & dosagem , Prurido/tratamento farmacológico , Prurido/genética , Indução de Remissão , Dermatopatias Genéticas/genética , Óleo de Soja/administração & dosagem , Síndrome , Resultado do Tratamento
9.
Retin Cases Brief Rep ; 15(3): 275-280, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-30024478

RESUMO

PURPOSE: Waardenburg syndrome (WS) is a rare condition characterized by six main features. It has been previously observed that WS is also associated with hypopigmentation of the choroid through multimodal imaging. To our knowledge, this is the first report of using swept-source optical coherence tomography angiography (OCTA) on a patient with known WS. METHODS: Report of a single case. The swept-source OCT images were captured using Topcon DRI OCT Triton (Topcon, Inc, Tokyo, Japan), whereas swept-source OCTA images were captured by Optovue AngioVue (Optovue, Inc, Fremont, CA) using DualTrack Motion Correction Technology. RESULTS: In this case, OCTA demonstrated evidence of normal vasculature of all layers (superficial, deep, and choricocapillaris), a normal foveal avascular zone measuring 0.267 mm2 in the right eye and 0.307 mm2 in the left eye, and a normal capillary density measuring 49.8% in the right eye and 52.6% in the left eye. CONCLUSION: There are many conditions that may mimic the hypopigmentation of the choroid associated with WS; it has been documented that these similar conditions such as choroidal nevus, choroidal melanoma, and Vogt-Koyanagi-Harada syndrome all demonstrated abnormal OCTA findings. Unlike these conditions, our patient with WS had unremarkable OCTA findings.


Assuntos
Doenças da Coroide/diagnóstico , Hipopigmentação/diagnóstico , Epitélio Pigmentado da Retina/patologia , Síndrome de Waardenburg/diagnóstico , Adulto , Doenças da Coroide/genética , Angiografia por Tomografia Computadorizada , Feminino , Angiofluoresceinografia , Humanos , Hipopigmentação/genética , Tomografia de Coerência Óptica , Síndrome de Waardenburg/genética
10.
Am J Dermatopathol ; 43(4): 259-265, 2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33201017

RESUMO

BACKGROUND: Hypopigmented mycosis fungoides (HMF) is an uncommon variant of mycosis fungoides. AIMS: To study the clinical and histopathology presentation in children with HMF. METHOD: We reviewed 9 children diagnosed with HMF. The clinical data were collected and analyzed. RESULT: Eight boys and 1 girl were included, with a median onset age of 7.4 year old and median age of diagnosis of 10.5 year old. Multiple hypopigmented patches were observed in all patients, and 5 patients exhibited multiple scaly erythema at the center of hypopigmented patches. Histopathology showed atypical lymphocytes with hyperchromatic, irregular, and cerebriform nuclei, infiltrated in the epidermis and dermis. Pautrier's microabscesses was noted in 6 of 9 patients, and papillary dermal fibroplasia was noted in 6 of 9 patients. CD8 predominance was detected in 4 of 6 patients. Four patients were simultaneously subjected to skin biopsy on hypopigmented patches and scaly erythema simultaneously. Compared with hypopigmented specimens, erythema biopsy detected deeper and denser infiltration of atypical lymphoid cells in 3 of 4 patients, higher CD4+/CD8+ ratio in 4 of 4 patients, more CD5 loss in 2 of 4 patients, and more CD7 loss in 2 of 4 patients. TCR gene monoclonal rearrangement was detected in 2 of 5 patients. Narrowband ultraviolet B phototherapy was applied in 7 patients. One of 7 patients achieved complete response, and 6 of 7 patients achieved partial response. No recurrence was noted with the median follow-up period of 6 months. CONCLUSION: HMF could occur in young patients, with indolent and benign course. HMF could gradually seem as scaly erythema based on hypopigmented patches. The histopathology indicated a more advanced stage of the scaly erythema lesions than hypopigmented patches.


Assuntos
Hipopigmentação/patologia , Micose Fungoide/patologia , Neoplasias Cutâneas/patologia , Pigmentação da Pele , Biomarcadores Tumorais/genética , Criança , Pré-Escolar , Feminino , Rearranjo Gênico do Linfócito T , Genes Codificadores dos Receptores de Linfócitos T , Humanos , Hipopigmentação/genética , Hipopigmentação/imunologia , Hipopigmentação/radioterapia , Linfócitos do Interstício Tumoral/imunologia , Masculino , Micose Fungoide/genética , Micose Fungoide/imunologia , Micose Fungoide/radioterapia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/radioterapia , Pigmentação da Pele/efeitos da radiação , Resultado do Tratamento , Terapia Ultravioleta
12.
13.
J Dermatol ; 47(6): 669-672, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32189379

RESUMO

We herein report a novel mutation in familial progressive hyper- and hypopigmentation (FPHH). The KITLG gene encoding the KIT ligand protein is a disease-causing gene for FPHH. Various disease-causing gain-of-function mutations, which reside within or adjacent to the conserved VTNN motif of this gene, have been described to date. We have now identified a novel KITLG mutation, c.337G>A (p.Glu113Lys), in FPHH which is located within another ligand-receptor interaction site.


Assuntos
Hiperpigmentação/genética , Hipopigmentação/genética , Fator de Células-Tronco/genética , Adulto , Biópsia , Criança , Análise Mutacional de DNA , Feminino , Testes Genéticos , Humanos , Hiperpigmentação/diagnóstico , Hiperpigmentação/patologia , Hipopigmentação/diagnóstico , Hipopigmentação/patologia , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Linhagem , Pele/patologia
14.
Orphanet J Rare Dis ; 14(1): 259, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31730496

RESUMO

BACKGROUND: Pigmentary mosaicism constitutes a heterogeneous group of skin pigmentation alterations associated with multisystem involvement. The aim of this study was to establish a complete cytogenetic and molecular characterization of PM patients, emphasizing on searching for possible low chromosomal mosaicism and on establishing an accurate genotype-phenotype correlation. RESULTS: A total of 73 patients were included (3 months to 18 years of age), 52% male and 48% female. Observed in 69 (95%) patients, the most frequent pattern of pigmentation was fine and whorled BL, which was associated with disseminated skin extent in 41 (59%) patients. Central nervous system (84%) alterations were the most frequent observed in the group of patients, followed by the musculoskeletal (53%) and ophthalmologic (27%) alterations. Considering the pattern of pigmentation, no significant differences in association with skin extent or extracutaneous manifestations were detected. Following a strict cytogenetic analysis strategy, screening metaphases from three different tissues (peripheral blood, hyperpigmented and hypopigmented skin) we found that 23/73 patients had chromosomal abnormalities classified as follows: 1) Mosaic with 2 or more different cell lines with structural alterations n = 19; 2) Polyploidy (mosaic) n = 1 and 3) Alterations in all cells in three different tissues n = 3. SNP array, array CGH and FISH were useful for the complete characterization of the chromosomal aberrations, for the detection of microdeletions in patients with normal karyotype but with strong clinical suspicious of chromosomal alteration, and for a better establishment of genotype-phenotype correlation. In 2 patients we found genes associated with some of the extracutaneous manifestations (SHH, MNX1, PPP2R2C). CONCLUSIONS: This group of 73 patients finely described is the largest series of patients with pigmentary mosaicism reported worldwide. As we showed in this study, the followed analysis strategy allowed the detection of cytogenetic and molecular abnormalities, and made possible the establishment of genotype-phenotype associations in some patients. An important limitation of our study was the analysis of fibroblasts cultures instead of melanocytes and keratinocytes. In some cases the direct molecular DNA analysis of skin biopsy could be another choice.


Assuntos
Hiperpigmentação/genética , Hiperpigmentação/patologia , Hipopigmentação/genética , Hipopigmentação/patologia , Adolescente , Criança , Pré-Escolar , Feminino , Estudos de Associação Genética , Humanos , Lactente , Cariotipagem , Queratinócitos/metabolismo , Masculino , Melanócitos/metabolismo , Pigmentação da Pele/genética
15.
Skinmed ; 17(2): 100-104, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31145059

RESUMO

Nevus depigmentosus, a disorder of hypopigmentation, occurs in both sexes and all races. It most commonly presents in early infancy and childhood as a nonprogressive hypomelanotic macule. It is considered a form of cutaneous mosaicism due to somatic mutation in pigmentary genes, which results in functional impairment of melanocytes. Clinical forms include localized, segmental, and systemized. Rare cases of nevus depigmentosus may be associated with systemic features. Treatment is usually not required, although certain techniques such as suction-blister grafting, excimer laser, and cosmetic camouflage have been tried with variable results. Counseling of parents plays a significant role to allay apprehension and anxiety.


Assuntos
Hipopigmentação/diagnóstico , Nevo/diagnóstico , Neoplasias Cutâneas/diagnóstico , Humanos , Hipopigmentação/genética , Hipopigmentação/patologia , Hipopigmentação/terapia , Mosaicismo , Nevo/genética , Nevo/patologia , Nevo/terapia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia
16.
J Invest Dermatol ; 139(10): 2174-2184.e4, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30998983

RESUMO

Vitiligo is a cutaneous depigmentation disorder caused by the destruction of epidermal melanocytes. The generation and the skin infiltration of autoreactive CD8+ cytotoxic T cells triggered by oxidative stress play a critical role in vitiligo. High-mobility group protein B1 (HMGB1) is a classic damage-associated molecular pattern molecule with strong proinflammatory effects in inflammatory reactions. A previous study reported an enhanced expression of HMGB1 in vitiligo lesions, but the role of HMGB1 in cutaneous inflammation of vitiligo is still unknown. In the present study, we initially found that HMGB1 was released from the nucleus of melanocytes in vitiligo perilesional skin. Furthermore, cultured normal human melanocytes could release HMGB1 under treatment with hydrogen peroxide. Moreover, HMGB1 facilitated the secretion of CXCL16 and IL-8 from keratinocytes by binding to the receptor for advanced glycation end products and activating NF-κB and extracellular signal-regulated kinase signaling pathways. Subsequently, HMGB1 led to the formation of chemotaxis for the migration of CD8+ T cells from patients with vitiligo by increasing the release of CXCL16 from keratinocytes. Additionally, HMGB1 promoted the maturation of dendritic cells from patients with vitiligo. Altogether, our study demonstrates that HMGB1 released from melanocytes contributes to the formation of oxidative stress-induced autoimmunity in vitiligo.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Quimiocina CXCL1/metabolismo , Proteína HMGB1/metabolismo , Estresse Oxidativo/fisiologia , Vitiligo/genética , Autoimunidade/genética , Western Blotting/métodos , Estudos de Casos e Controles , Movimento Celular/imunologia , Células Cultivadas , Ensaio de Imunoadsorção Enzimática/métodos , Citometria de Fluxo/métodos , Humanos , Hipopigmentação/genética , Hipopigmentação/patologia , Queratinócitos/metabolismo , Melanócitos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/métodos , Valores de Referência , Vitiligo/sangue , Vitiligo/imunologia , Vitiligo/patologia
17.
Pigment Cell Melanoma Res ; 32(5): 643-656, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30849202

RESUMO

Although pigment synthesis is well understood, relevant mechanisms of psychologically debilitating dyspigmentation in nascent tissue after cutaneous injuries are still unknown. Here, differences in genomic transcription of hyper- and hypopigmented tissue relative to uninjured skin were investigated using a red Duroc swine scar model. Transcription profiles differed based on pigmentation phenotypes with a trend of more upregulation or downregulation in hyper- or hypopigmented scars, respectively. Ingenuity Pathway Analysis of significantly modulated genes in both pigmentation phenotypes showed pathways related to redox, metabolic, and inflammatory responses were more present in hypopigmented samples, while those related to stem cell development differentiation were found mainly in hyperpigmented samples. Cell-cell and cell-extracellular matrix interactions and inflammation responses were predicted (z-score) active in hyperpigmented and inactive in hypopigmented. The proinflammatory high-mobility group box 1 pathway showed the opposite trend. Analysis of differentially regulated mutually exclusive genes showed an extensive presence of metabolic, proinflammatory, and oxidative stress pathways in hypopigmented scars, while melanin synthesis, glycosaminoglycans biosynthesis, and cell differentiation pathways were predominant in hyperpigmented scar. Several potential therapeutic gene targets have been identified.


Assuntos
Biomarcadores/análise , Cicatriz Hipertrófica/patologia , Cor , Hiperpigmentação/patologia , Hipopigmentação/patologia , Pigmentação da Pele/genética , Ferimentos e Lesões/patologia , Animais , Cicatriz Hipertrófica/genética , Cicatriz Hipertrófica/metabolismo , Modelos Animais de Doenças , Hiperpigmentação/genética , Hipopigmentação/genética , Masculino , Suínos , Transcriptoma , Cicatrização , Ferimentos e Lesões/genética
18.
Cancer Lett ; 442: 299-309, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30445206

RESUMO

Melanoma is an aggressive cancer with poor prognosis, requiring personalized management of advanced stages and establishment of molecular markers. Melanomas derive from melanocytes, which specifically express tyrosinase, the rate-limiting enzyme of melanin-synthesis. We demonstrate that melanomas with high levels of DNp73, a cancer-specific variant of the p53 family member p73 and driver of melanoma progression show, in contrast to their less-aggressive low-DNp73 counterparts, hypopigmentation in vivo. Mechanistically, reduced melanin-synthesis is mediated by a DNp73-activated IGF1R/PI3K/AKT axis leading to tyrosinase ER-arrest and proteasomal degradation. Tyrosinase loss triggers reactivation of the EMT signaling cascade, a mesenchymal-like cell phenotype and increased invasiveness. DNp73-induced depigmentation, Slug increase and changes in cell motility are recapitulated in neural crest-derived melanophores of Xenopus embryos, underscoring a previously unnoticed physiological role of tyrosinase as EMT inhibitor. This data provides a mechanism of hypopigmentation accompanying cancer progression, which can be exploited in precision diagnosis of patients with melanoma-associated hypopigmentation (MAH), currently seen as a favorable prognostic factor. The DNp73/IGF1R/Slug signature in colorless lesions might aid to clinically discriminate between patients with MAH-associated metastatic disease and those, where MAH is indeed a sign of regression.


Assuntos
Transição Epitelial-Mesenquimal , Hipopigmentação/enzimologia , Melaninas/metabolismo , Melanócitos/enzimologia , Melanoma/enzimologia , Monofenol Mono-Oxigenase/metabolismo , Neoplasias Cutâneas/enzimologia , Proteína Tumoral p73/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Humanos , Hipopigmentação/genética , Hipopigmentação/patologia , Melanócitos/patologia , Melanoma/genética , Melanoma/patologia , Camundongos , Monofenol Mono-Oxigenase/genética , Invasividade Neoplásica , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Espécies Reativas de Oxigênio/metabolismo , Receptor IGF Tipo 1 , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Proteína Tumoral p73/genética , Xenopus laevis
19.
Am J Med Genet A ; 179(1): 123-129, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30561107

RESUMO

Terminal osseous dysplasia with pigmentary defects (TODPD; MIM #300244) is an extremely rare, X-linked dominant, in utero male-lethal disease, characterized by skeletal dysplasia of the limbs, pigmentary defects of the skin, and recurrent digital fibromatosis of childhood. Delayed/abnormal ossification of bones of the hands and feet, joint contractures, and dysmorphic facial features may accompany. A single recurrent mutation (c.5217 G>A) of the FLNA gene which causes cryptic splicing was identified as the cause of the disease. We here present the first TODPD case from Turkey with full-blown phenotype who exhibit unique additional findings, hypopigmented patch on the lower extremity following Blaschko's lines and smooth muscle hamartoma of the scalp in review of all the previously reported TODPD cases.


Assuntos
Doenças do Desenvolvimento Ósseo/fisiopatologia , Filaminas/genética , Dedos/anormalidades , Doenças Genéticas Ligadas ao Cromossomo X/fisiopatologia , Deformidades Congênitas dos Membros/fisiopatologia , Osteocondrodisplasias/fisiopatologia , Transtornos da Pigmentação/fisiopatologia , Pele/fisiopatologia , Dedos do Pé/anormalidades , Doenças do Desenvolvimento Ósseo/diagnóstico , Doenças do Desenvolvimento Ósseo/diagnóstico por imagem , Doenças do Desenvolvimento Ósseo/genética , Pré-Escolar , Feminino , Dedos/diagnóstico por imagem , Dedos/fisiopatologia , Doenças Genéticas Ligadas ao Cromossomo X/diagnóstico , Doenças Genéticas Ligadas ao Cromossomo X/diagnóstico por imagem , Doenças Genéticas Ligadas ao Cromossomo X/genética , Mãos/fisiopatologia , Humanos , Hipopigmentação/diagnóstico por imagem , Hipopigmentação/genética , Hipopigmentação/fisiopatologia , Lactente , Deformidades Congênitas dos Membros/diagnóstico , Deformidades Congênitas dos Membros/diagnóstico por imagem , Deformidades Congênitas dos Membros/genética , Mutação , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/diagnóstico por imagem , Osteocondrodisplasias/genética , Fenótipo , Transtornos da Pigmentação/diagnóstico , Transtornos da Pigmentação/diagnóstico por imagem , Transtornos da Pigmentação/genética , Dedos do Pé/diagnóstico por imagem , Dedos do Pé/fisiopatologia , Turquia/epidemiologia
20.
Ann Clin Lab Sci ; 48(2): 248-252, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29678855

RESUMO

Kallmann syndrome (KS) is a clinically and genetically heterogeneous disorder characterized by hypogonadotropic hypogonadism and olfactory dysfunction. Recently, mutations in SOX10, a well-known causative gene of Waardenburg syndrome (WS), have been identified in a few KS patients with additional developmental defects including hearing loss. However, the understanding of SOX10 mutation associates with KS and other clinical consequences remains fragmentary. A 30-year-old Chinese male patient presented with no pubertal sex development when he was at the age of twelve years. Additionally, he showed anosmia, sensory deafness, and blue irises. Last year, he developed clinical symptoms of hyperthyroidism with a fast heartbeat, heat intolerance and weight loss. Blood examinations revealed low levels of FSH, LH, and testosterone. Thyroid function showed high levels of FT3, FT4 and extremely low level of TSH. Molecular analysis detected a de novo (c.565G>T/p.E189X) mutation in SOX10, which has previously been reported in a patient with WS4 (WS with Hirschsprung). The mutation was predicted to be probably damaging. These results highlight the significance of SOX10 haploinsufficiency as a genetic cause of KS. Importantly, our result implies that the same SOX10 mutation can underlie both typical KS and WS, while the correlation between SOX10 and hyperthyroidism still needs to be clarified in the future.


Assuntos
Códon sem Sentido/genética , Surdez/genética , Hipertireoidismo/genética , Hipopigmentação/genética , Síndrome de Kallmann/genética , Fatores de Transcrição SOXE/genética , Adulto , Análise Mutacional de DNA , Surdez/complicações , Surdez/diagnóstico por imagem , Humanos , Hipertireoidismo/complicações , Hipertireoidismo/diagnóstico por imagem , Hipopigmentação/complicações , Hipopigmentação/diagnóstico por imagem , Síndrome de Kallmann/complicações , Síndrome de Kallmann/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA