Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Pathol ; 262(4): 441-453, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38186269

RESUMO

Vitiligo is a depigmented skin disease due to the destruction of melanocytes. Under oxidative stress, keratinocyte-derived chemokine C-X-C motif ligand 16 (CXCL16) plays a critical role in recruiting CD8+ T cells, which kill melanocytes. Autophagy serves as a protective cell survival mechanism and impairment of autophagy has been linked to increased secretion of the proinflammatory cytokines. However, the role of autophagy in the secretion of CXCL16 under oxidative stress has not been investigated. Herein, we initially found that autophagy was suppressed in both keratinocytes of vitiligo lesions and keratinocytes exposed to oxidative stress in vitro. Autophagy inhibition also promoted CXCL16 secretion. Furthermore, upregulated transient receptor potential cation channel subfamily M member 2 (TRPM2) functioned as an upstream oxidative stress sensor to inhibit autophagy. Moreover, TRPM2-mediated Ca2+ influx activated calpain to shear autophagy related 5 (Atg5) and Atg12-Atg5 conjugate formation was blocked to inhibit autophagy under oxidative stress. More importantly, Atg5 downregulation enhanced the binding of interferon regulatory factor 3 (IRF3) to the CXCL16 promoter region by activating Tank-binding kinase 1 (TBK1), thus promoting CXCL16 secretion. These findings suggested that TRPM2-restrained autophagy promotes CXCL16 secretion via the Atg5-TBK1-IRF3 signaling pathway under oxidative stress. Inhibition of TRPM2 may serve as a potential target for the treatment of vitiligo. © 2024 The Pathological Society of Great Britain and Ireland.


Assuntos
Canais de Cátion TRPM , Vitiligo , Humanos , Vitiligo/metabolismo , Vitiligo/patologia , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Linfócitos T CD8-Positivos/patologia , Queratinócitos/patologia , Estresse Oxidativo , Autofagia , Quimiocina CXCL16/metabolismo
2.
J Transl Med ; 21(1): 434, 2023 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-37403086

RESUMO

BACKGROUND: The activation of CD8+ T cells and their trafficking to the skin through JAK-STAT signaling play a central role in the development of vitiligo. Thus, targeting this key disease pathway with innovative drugs is an effective strategy for treating vitiligo. Natural products isolated from medicinal herbs are a useful source of novel therapeutics. Demethylzeylasteral (T-96), extracted from Tripterygium wilfordii Hook F, possesses immunosuppressive and anti-inflammatory properties. METHODS: The efficacy of T-96 was tested in our mouse model of vitiligo, and the numbers of CD8+ T cells infiltration and melanocytes remaining in the epidermis were quantified using whole-mount tail staining. Immune regulation of T-96 in CD8+ T cells was evaluated using flow cytometry. Pull-down assay, mass spectrum analysis, molecular docking, knockdown and overexpression approaches were utilized to identify the target proteins of T-96 in CD8+ T cells and keratinocytes. RESULTS: Here, we found that T-96 reduced CD8+ T cell infiltration in the epidermis using whole-mount tail staining and alleviated the extent of depigmentation to a comparable degree of tofacitinib (Tofa) in our vitiligo mouse model. In vitro, T-96 decreased the proliferation, CD69 membrane expression, and IFN-γ, granzyme B, (GzmB), and perforin (PRF) levels in CD8+ T cells isolated from patients with vitiligo. Pull-down assays combined with mass spectrum analysis and molecular docking showed that T-96 interacted with JAK3 in CD8+ T cell lysates. Furthermore, T-96 reduced JAK3 and STAT5 phosphorylation following IL-2 treatment. T-96 could not further reduce IFN-γ, GzmB and PRF expression following JAK3 knockdown or inhibit increased immune effectors expression upon JAK3 overexpression. Additionally, T-96 interacted with JAK2 in IFN-γ-stimulated keratinocytes, inhibiting the activation of JAK2, decreasing the total and phosphorylated protein levels of STAT1, and reducing the production and secretion of CXCL9 and CXCL10. T-96 did not significantly inhibit STAT1 and CXCL9/10 expression following JAK2 knockdown, nor did it suppress upregulated STAT1-CXCL9/10 signaling upon JAK2 overexpression. Finally, T-96 reduced the membrane expression of CXCR3, and the culture supernatants pretreated with T-96 under IFN-γ stressed keratinocytes markedly blocked the migration of CXCR3+CD8+ T cells, similarly to Tofa in vitro. CONCLUSION: Our findings demonstrated that T-96 might have positive therapeutic responses to vitiligo by pharmacologically inhibiting the effector functions and skin trafficking of CD8+ T cells through JAK-STAT signaling.


Assuntos
Vitiligo , Animais , Camundongos , Vitiligo/tratamento farmacológico , Vitiligo/metabolismo , Linfócitos T CD8-Positivos , Simulação de Acoplamento Molecular , Pele/metabolismo
3.
J Cell Mol Med ; 22(9): 4335-4343, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29974998

RESUMO

In vitiligo, cutaneous depigmentation is accompanied by increased T cell cytolytic activity targeting melanocytes, indicating that autoimmune tolerance is disrupted. The inhibited amount and function of Tregs have been indicated to be involved in the autoimmune intolerance in vitiligo, however, with the conclusion still controversial and the involved mechanism unknown. In this study, we explored the molecular and cellular alterations accounting for the impaired Treg response in vitiligo. Our results showed that the amount of Tregs was drastically reduced in peripheral blood of active vitiligo patients. Furthermore, the immunoregulatory function of Tregs was attenuated, with lower expression of CTLA4, IL-10 and TGF-ß. Moreover, the expression of HO-1, a functional modulator of Tregs, was decreased in vitiligo Tregs, and the concentrations of HO-1 metabolites, including bilirubin, CoHb and iron, were correspondingly decreased in serum of vitiligo patients. In addition, we treated the Tregs from vitiligo patients with Hemin, an agonist of HO-1, and found that enhanced HO-1 expression restored the function of Tregs by up-regulating IL-10 expression. Our study demonstrates the essential role of HO-1 in the impaired Treg response in vitiligo and indicates the potential of HO-1 as a therapeutic target in vitiligo management.


Assuntos
Antígeno CTLA-4/genética , Heme Oxigenase-1/genética , Interleucina-10/genética , Melanócitos/imunologia , Linfócitos T Reguladores/imunologia , Vitiligo/genética , Adolescente , Adulto , Idoso , Bilirrubina/sangue , Bilirrubina/imunologia , Antígeno CTLA-4/imunologia , Carboxihemoglobina/imunologia , Carboxihemoglobina/metabolismo , Progressão da Doença , Feminino , Regulação da Expressão Gênica , Heme Oxigenase-1/imunologia , Hemina/farmacologia , Humanos , Tolerância Imunológica , Interleucina-10/imunologia , Ferro/sangue , Ferro/imunologia , Contagem de Linfócitos , Masculino , Melanócitos/patologia , Pessoa de Meia-Idade , Cultura Primária de Células , Índice de Gravidade de Doença , Transdução de Sinais , Pele/imunologia , Pele/patologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/patologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia , Vitiligo/imunologia , Vitiligo/patologia
4.
Curr Opin Psychol ; 46: 101347, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35500357

RESUMO

The modern marketplace has made consumers' lives better in many ways, offering a multitude of affordable conveniences and luxuries. Why, then, is the prevalence of physical and mental health deficits higher than any other time in history? Here, we articulate an evolutionary mismatch perspective-the idea that the environment we live in has changed dramatically in a short period of time, but the human body and mind have not changed. Consumers' evolved body and mind are interacting with the modern world as if it was an ancestral environment that existed thousands of years ago, leading to many negative outcomes. We discuss three evolutionary mismatches that contribute to or compound consumer vulnerability to disease and dissatisfaction with life. We review emerging research and propose future directions that inform effective strategies to mitigate illness and enhance wellbeing.


Assuntos
Felicidade , Saúde Mental , Humanos
5.
Front Med (Lausanne) ; 9: 818543, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35721084

RESUMO

Background and Objectives: The efficacy of camouflage combined with psychotherapy and the underlying mechanisms are poorly understood in vitiligo management. This study aimed to investigate the joint efficacy and further explore psycho-neuro-endocrine-immune-skin interactions. Patients and Methods: In a prospective, non-randomized and concurrent controlled trial, patients were divided into two groups. Quality of life (QOL) was evaluated using the Chinese version of the Vitiligo Life Quality Index (VLQI-C). Serum levels of neuropeptides and cytokines were detected by enzyme-linked immunosorbent assay. Results: A total of 149 patients were included for final evaluation. After treatment for 4 weeks, total and subcategory quality of life scores in the intervention group were much lower than in the control group. Serum levels of neuropeptide-Y (NPY) and melanin-concentrating hormone (MCH) significantly decreased, and serum level of adrenocorticotropic hormone (ACTH) increased in both active and stable patients of the intervention group, but not in the control group. In addition, the serum levels of interferon-γ (IFN-γ), CXC chemokine ligand 10 (CXCL10), and interleukin-1ß (IL-1ß) decreased in both the active and stable patients of the intervention group and only in the active patients of the control group. Conclusions: The combination of camouflage and psychotherapy provided a clinically meaningful improvement in quality of life and ameliorated the outcome by likely modulating the psycho-neuro-endocrine-immuno-skin system during vitiligo management. Clinical Trial Registration: www.clinicaltrials.gov/ct2/show/NCT03540966, identifier: NCT03540966.

6.
Front Immunol ; 13: 955038, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36203601

RESUMO

Alopecia areata (AA) is an autoimmune disease caused by T cell-mediated destruction of the hair follicle (HF). Therefore, approaches that effectively disrupt pathogenic T cell responses are predicted to have therapeutic benefit for AA treatment. T cells rely on the duality of T cell receptor (TCR) and gamma chain (γc) cytokine signaling for their development, activation, and peripheral homeostasis. Ifidancitinib is a potent and selective next-generation JAK1/3 inhibitor predicted to disrupt γc cytokine signaling. We found that Ifidancitinib robustly induced hair regrowth in AA-affected C3H/HeJ mice when fed with Ifidancitinib in chow diets. Skin taken from Ifidancitinib-treated mice showed significantly decreased AA-associated inflammation. CD44+CD62L- CD8+ T effector/memory cells, which are associated with the pathogenesis of AA, were significantly decreased in the peripheral lymphoid organs in Ifidancitinib-treated mice. We observed high expression of co-inhibitory receptors PD-1 on effector/memory CD8+ T cells, together with decreased IFN-γ production in Ifidancitinib-treated mice. Furthermore, we found that γc cytokines regulated T cell exhaustion. Taken together, our data indicate that selective induction of T cell exhaustion using a JAK inhibitor may offer a mechanistic explanation for the success of this treatment strategy in the reversal of autoimmune diseases such as AA.


Assuntos
Alopecia em Áreas , Doenças Autoimunes , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 3/antagonistas & inibidores , Inibidores de Janus Quinases , Alopecia em Áreas/tratamento farmacológico , Animais , Linfócitos T CD8-Positivos , Citocinas/uso terapêutico , Inibidores de Janus Quinases/farmacologia , Inibidores de Janus Quinases/uso terapêutico , Camundongos , Camundongos Endogâmicos C3H , Receptor de Morte Celular Programada 1
7.
JCI Insight ; 6(7)2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33830087

RESUMO

The Janus kinase/signal transducers and activators of transcription (JAK/STAT) are key intracellular mediators in the signal transduction of many cytokines and growth factors. Common γ chain cytokines and interferon-γ that use the JAK/STAT pathway to induce biological responses have been implicated in the pathogenesis of alopecia areata (AA), a T cell-mediated autoimmune disease of the hair follicle. We previously showed that therapeutic targeting of JAK/STAT pathways using the first-generation JAK1/2 inhibitor, ruxolitinib, and the pan-JAK inhibitor, tofacitinib, was highly effective in the treatment of human AA, as well as prevention and reversal of AA in the C3H/HeJ mouse model. To better define the role of individual JAKs in the pathogenesis of AA, in this study, we tested and compared the efficacy of several next-generation JAK-selective inhibitors in the C3H/HeJ mouse model of AA, using both systemic and topical delivery. We found that JAK1-selective inhibitors as well as JAK3-selective inhibitors robustly induced hair regrowth and decreased AA-associated inflammation, whereas several JAK2-selective inhibitors failed to restore hair growth in treated C3H/HeJ mice with AA. Unlike JAK1, which is broadly expressed in many tissues, JAK3 expression is largely restricted to hematopoietic cells. Our study demonstrates inhibiting JAK3 signaling is sufficient to prevent and reverse disease in the preclinical model of AA.


Assuntos
Alopecia em Áreas/tratamento farmacológico , Janus Quinase 3/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Administração Tópica , Alopecia em Áreas/metabolismo , Alopecia em Áreas/prevenção & controle , Animais , Azetidinas/administração & dosagem , Azetidinas/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Citocinas/metabolismo , Ácidos Isonicotínicos/administração & dosagem , Ácidos Isonicotínicos/farmacologia , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 1/metabolismo , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/metabolismo , Janus Quinase 3/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos C3H , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Nitrilas/farmacologia , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/administração & dosagem , Pirazóis/farmacologia , Piridinas/farmacologia , Pirimidinas/administração & dosagem , Pirimidinas/farmacologia , Pirróis/administração & dosagem , Pirróis/farmacologia , Triazóis/farmacologia
8.
Sci Adv ; 7(14)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33811067

RESUMO

The interleukin-7 (IL-7) signaling pathway plays an important role in regulation of T cell function and survival. We detected overexpression of IL-7 in lesional skin from both humans and C3H/HeJ mice with alopecia areata (AA), a T cell-mediated autoimmune disease of the hair follicle. We found that exogenous IL-7 accelerated the onset of AA by augmenting the expansion of alopecic T cells. Conversely, blockade of IL-7 stopped the progression of AA and reversed early AA in C3H/HeJ mice. Mechanistically, we observed that IL-7Rα blockade substantially reduced the total number of most T cell subsets, but relative sparing of regulatory T cells (Tregs). We postulated that short-term anti-IL-7Rα treatment in combination with a low dose of Treg-tropic cytokines might improve therapeutic efficacy in AA. We demonstrated that short-term IL-7Rα blockade in combination with low doses of Treg-tropic cytokines enhanced therapeutic effects in the treatment of AA, and invite further clinical investigation.

9.
J Invest Dermatol ; 141(12): 2921-2931.e6, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34102211

RESUMO

Vitiligo is a common depigmentation disease characterized by melanocyte death, which is attributed to various mechanisms such as apoptosis and autoimmune destruction. However, whether necroptosis, a newly discovered way of cell death, plays a key role in the pathogenesis of vitiligo is still elusive and has not been well-studied. In this study, we found that necroptosis markers, including phosphorylated RIP3 and phosphorylated-MLKL, were positive in melanocytes from vitiligo perilesional skin, which supported the existence of necroptosis in vitiligo. Furthermore, the expression of RIP1 was remarkably upregulated in melanocytes treated with hydrogen peroxide. Then, RIP1 intervention suppression and MLKL deficiency could significantly enhance the resistance of melanocytes to hydrogen peroxide‒induced necroptosis. Mechanistically, we confirmed that RIP1 and RIP3 could form necrosomes under oxidative stress and further trigger phosphorylated MLKL translocation to the cell membrane, which led to the destruction of melanocytes. Finally, we showed that RIP1-mediated generation of mitochondrial ROS contributed to necrosome formation in melanocytes. Collectively, our study confirms that necroptosis significantly facilitates oxidative stress‒induced melanocyte death through the RIP1 signaling pathway, offering insight into vitiligo.


Assuntos
Melanócitos/patologia , Necroptose/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Estresse Oxidativo/fisiologia , Proteínas de Ligação a RNA/fisiologia , Vitiligo/etiologia , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases/fisiologia , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia , Transdução de Sinais/fisiologia , Vitiligo/patologia
10.
Cell Death Dis ; 11(6): 453, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32532953

RESUMO

Vitiligo is a disfiguring disease featuring chemokines-mediated cutaneous infiltration of autoreactive CD8+ T cells that kill melanocytes. Copious studies have indicated that virus invasion participates in the pathogenesis of vitiligo. IFIH1, encoding MDA5 which is an intracellular virus sensor, has been identified as a vitiligo susceptibility gene. However, the specific role of MDA5 in melanocyte death under virus invasion is not clear. In this study, we first showed that the expression of anti-CMV IgM and MDA5 was higher in vitiligo patients than healthy controls. Then, by using Poly(I:C) to imitate virus invasion, we clarified that virus invasion significantly activated MDA5 and further potentiated the keratinocyte-derived CXCL10 and CXCL16 which are the two vital chemokines for the cutaneous infiltration of CD8+ T cells in vitiligo. More importantly, IFN-ß mediated by the MDA5-MAVS-NF-κB/IRF3 signaling pathway orchestrated the secretion of CXCL10 via the JAK1-STAT1 pathway and MDA5-meidiated IRF3 transcriptionally induced the production of CXCL16 in keratinocytes under virus invasion. In summary, our results demonstrate that MDA5 signaling orchestrates the aberrant skin immunity engaging in melanocyte death via mediating CXCL10 and CXCL16 secretion, which supports MDA5 as a potential therapeutic target for vitiligo under virus invasion.


Assuntos
Quimiocinas/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Queratinócitos/metabolismo , Melanócitos/metabolismo , Vitiligo/genética , Estudos de Casos e Controles , Humanos , Invasividade Neoplásica , Transdução de Sinais , Transfecção
11.
J Dermatol Sci ; 94(1): 236-243, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30987854

RESUMO

BACKGROUND: Oxidative stress plays important roles in the pathogenesis of vitiligo. The removal of hydrogen peroxided (H2O2) has been established to be beneficial to vitiligo patients. Berberine (BBR), a natural isoquinoline alkaloid, has antioxidant activity, however, whether BBR can defend human melanocytes against oxidative injury remains to be elucidated. OBJECTIVE: In the present study, we investigated the potential protective effect of BBR against oxidative stress on an immortalized normal human melanocyte cell line PIG1. METHODS: Generally, PIG1 cells were pretreated with various concentrations of BBR for 1 h followed by exposure to 1.0 mM H2O2 for 24 h. Cell apoptosis, intracellular reactive oxygen species (ROS) levels were assessed through flow cytometry. Cell apoptosis, melanogenesis and the activation of Nrf2-ARE and Mitf signaling pathway were assayed. RESULTS: Our results showed that cell viability rose and intracellular ROS generation, cell apoptosis of melanocytes decreased significantly in response to H2O2 through pretreatment with BBR. Furthermore, We found that BBR can dramatically induce Nrf2 nuclear translocation, increase total Nrf2 levels and enhance ARE activity. Besides, Nrf2-siRNA transfection can abrogate the protection of BBR in melanocytes against oxidative injury. At last, we verified that BBR could facilitate melanogenesis function via modulation of Mitf and its target proteins. CONCLUSION: The results above suggest that BBR can protect melanocytes against oxidative stress via its anti-oxidative activity. Also, we found H2O2-induced activation of NFκB was inhibited by BBR. Therefore, it is worthy of investigation BBR as a potential drug for treatment of vitiligo.


Assuntos
Berberina/farmacologia , Melanócitos/efeitos dos fármacos , Vitiligo/tratamento farmacológico , Berberina/uso terapêutico , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Peróxido de Hidrogênio/toxicidade , Melaninas/biossíntese , Melanócitos/metabolismo , Fator de Transcrição Associado à Microftalmia/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Vitiligo/patologia
12.
Free Radic Biol Med ; 139: 80-91, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31078730

RESUMO

Oxidative stress and effector memory CD8+ T cells have been greatly implicated in vitiligo pathogenesis. However, the crosstalk between these two crucial pathogenic factors has been merely investigated. IL-15 has been regarded as an important cytokine exerting its facilitative effect on memory CD8+ T cells function in various autoimmune diseases. In the present study, we initially discovered that the IL-15 expression was significantly increased in vitiligo epidermis and highly associated with epidermal H2O2 content. In addition, epidermal IL-15 expression was mainly derived from keratinocytes. Then, we showed that oxidative stress promoted IL-15 and IL-15Rα expression as well as IL-15 trans-presentation by activating NF-κB signaling in keratinocytes. What's more, the trans-presented IL-15, rather than the secreted one, was accounted for the potentiation of CD8+ TEMs activation. We further investigated the mechanism underlying trans-presented IL-15 in potentiating CD8+ TEMs activation and found that the blockage of IL-15-JAK-STAT signaling could be a potent therapeutic approach. Taken together, our results demonstrate that oxidative stress-induced IL-15 trans-presentation in keratinocytes contributes to the activation of CD8+ TEMs, providing a novel mechanism by which oxidative stress initiates autoimmunity in vitiligo.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Interleucina-15/genética , Janus Quinase 1/genética , Queratinócitos/metabolismo , Fator de Transcrição STAT3/genética , Vitiligo/genética , Anti-Inflamatórios/farmacologia , Betametasona/análogos & derivados , Betametasona/farmacologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Técnicas de Cocultura , Combinação de Medicamentos , Regulação da Expressão Gênica , Humanos , Peróxido de Hidrogênio/metabolismo , Interleucina-15/metabolismo , Janus Quinase 1/metabolismo , Queratinócitos/efeitos dos fármacos , Queratinócitos/patologia , Ativação Linfocitária/efeitos dos fármacos , Melanócitos/efeitos dos fármacos , Melanócitos/metabolismo , Melanócitos/patologia , Estresse Oxidativo , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Interleucina-15/antagonistas & inibidores , Receptores de Interleucina-15/genética , Receptores de Interleucina-15/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Vitiligo/metabolismo , Vitiligo/patologia
13.
Theranostics ; 9(6): 1614-1633, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31037127

RESUMO

Mitochondrial dysregulation has been implicated in oxidative stress-induced melanocyte destruction in vitiligo. However, the molecular mechanism underlying this process is merely investigated. Given the prominent role of nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase Sirtuin3 (SIRT3) in sustaining mitochondrial dynamics and homeostasis and that SIRT3 expression and activity can be influenced by oxidative stress-related signaling, we wondered whether SIRT3 could play an important role in vitiligo melanocyte degeneration by regulating mitochondrial dynamics. Methods: We initially testified SIRT3 expression and activity in normal and vitiligo melanocytes via PCR, immunoblotting and immunofluorescence assays. Then, cell apoptosis, mitochondrial function and mitochondrial dynamics after SIRT3 intervention were analyzed by flow cytometry, immunoblotting, confocal laser microscopy, transmission electron microscopy and oxphos activity assays. Chromatin immunoprecipitation (ChIP), co-immunoprecipitation (Co-IP), immunoblotting and immunofluorescence assays were performed to clarify the upstream regulatory mechanism of SIRT3. Finally, the effect of honokiol on protecting melanocytes and the underlying mechanism were investigated via flow cytometry and immunoblotting analysis. Results: We first found that the expression and the activity of SIRT3 were significantly impaired in vitiligo melanocytes both in vitro and in vivo. Then, SIRT3 deficiency led to more melanocyte apoptosis by inducing severe mitochondrial dysfunction and cytochrome c release to cytoplasm, with Optic atrophy 1 (OPA1)-mediated mitochondrial dynamics remodeling involved in. Moreover, potentiated carbonylation and dampened peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) activation accounted for SIRT3 dysregulation in vitiligo melanocytes. Finally, we proved that honokiol could prevent melanocyte apoptosis under oxidative stress by activating SIRT3-OPA1 axis. Conclusions: Overall, we demonstrate that SIRT3-dependent mitochondrial dynamics remodeling contributes to oxidative stress-induced melanocyte degeneration in vitiligo, and honokiol is promising in preventing oxidative stress-induced vitiligo melanocyte apoptosis.


Assuntos
Melanócitos/patologia , Dinâmica Mitocondrial , Estresse Oxidativo , Sirtuína 3/metabolismo , Vitiligo/patologia , Adolescente , Adulto , Apoptose , Citocromos c/toxicidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
14.
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
15.
Free Radic Biol Med ; 129: 492-503, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30342186

RESUMO

Vitiligo is a complex disorder characterized by patchy loss of skin pigmentation due to abnormal melanocyte function. Overwhelming evidences have suggested that oxidative stress plays a major role in the loss of melanocytes thereby mediating the onset and progression of vitiligo. The nuclear factor erythroid 2-like factor 2 (Nrf2) is a master regulator of cellular redox homeostasis and the activation of Nrf2 signaling pathway is impaired in the vitiligo melanocytes. Baicalein, as flavonoid extracted from the Scutellaria baicalensis, has been proved to possess the ability to activate Nrf2 signaling pathway in other cell types and mouse model. Our previous data found that baicalein exerts a cytoprotective role in H2O2-induced apoptosis in human melanocytes cell line (PIG1). Based on these founding, we hypothesized that baicalein activates Nrf2 signaling pathway, alleviates H2O2-induced mitochondrial dysfunction and cellular damage, thereby protecting human vitiligo melanocytes from oxidative stress. In the present study, we found that baicalein effectively inhibited H2O2-induced cytotoxicity and apoptosis in human vitiligo melanocytes (PIG3V). Further results demonstrated that baicalein promoted Nrf2 nucleus translocation as well as up-regulated the expression of Nrf2 and its target gene, heme oxygenase-1 (HO-1). Moreover, the protective effects of baicalein against H2O2-induced cellular damage and apoptosis as well as mitochondrial dysfunction were abolished by Nrf2 knockdown. Additionally, we observed that Nrf2 knockdown suppressed proliferation and increased the sensitivity of PIG3V cells to H2O2 treatment. Finally, we explored the mechanism of baicalein associated with Nrf2 activation and found that the phosphorylation of Nrf2 as well as ERK1/2and PI3K/AKT signaling were not involved in the baicalein-induced activation of Nrf2. Taken together, these data clearly suggest that baicalein enhances cellular antioxidant defense capacity of human vitiligo melanocytes through the activation of the Nrf2 signaling pathway, providing beneficial evidence for the application of baicalein in the vitiligo treatment.


Assuntos
Antioxidantes/farmacologia , Flavanonas/farmacologia , Peróxido de Hidrogênio/farmacologia , Melanócitos/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/genética , Transdução de Sinais/efeitos dos fármacos , Antioxidantes/isolamento & purificação , Linhagem Celular , Flavanonas/isolamento & purificação , Regulação da Expressão Gênica , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Humanos , Peróxido de Hidrogênio/antagonistas & inibidores , Melanócitos/metabolismo , Melanócitos/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Extratos Vegetais/química , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Scutellaria baicalensis/química , Transdução de Sinais/genética , Vitiligo/genética , Vitiligo/metabolismo , Vitiligo/patologia
16.
Free Radic Biol Med ; 126: 259-268, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30138713

RESUMO

Abnormal mitochondrial calcium accumulation plays a critical role in oxidative stress-induced apoptosis of melanocytes. Transient receptor potential cation channel subfamily M member 2 (TRPM2) is a calcium channel sensitive to oxidative stress. However, whether TRPM2 participates in melanocyte apoptosis under oxidative stress was unknown before. In the present study, we initially found that hydrogen peroxide (H2O2) induced the demethylation of the promoter region in TRPM2 gene and increased the expression of TRPM2 in normal human melanocytes (NHMs). Meanwhile, TRPM2 was overexpressed in lesional melanocytes of vitiligo that is a skin disease caused by melanocyte loss under oxidative stress. Furthermore, either TRPM2 inhibitors or TRPM2 shRNA could ameliorate H2O2-induced apoptosis, mitochondrial reactive oxygen species (ROS) accumulation and mitochondrial membrane potential (MMP) loss in NHMs, which was similar to the effects of an anti-oxidant. More importantly, TRPM2 mediated the calcium influx into the cytoplasm and the mitochondria of NHMs exposed to H2O2, and a specific mitochondrial Ca2+ uptake inhibitor Ruthenium 360 (Ru360) could also protect NHMs from apoptosis and mitochondrial damages caused by H2O2. Taken together, our findings demonstrate that oxidative stress promotes the expression of TRPM2 and thus facilitates mitochondria-dependent apoptosis of melanocytes by increasing calcium influx. Our study indicates that TRPM2 is a potential target for protecting melanocytes against oxidative damages in vitiligo.


Assuntos
Metilação de DNA/genética , Peróxido de Hidrogênio/metabolismo , Melanócitos/metabolismo , Canais de Cátion TRPM/genética , Vitiligo/genética , Apoptose/genética , Cálcio/metabolismo , Canais de Cálcio/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/toxicidade , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Estresse Oxidativo/genética , Regiões Promotoras Genéticas/genética , Espécies Reativas de Oxigênio/química , Vitiligo/metabolismo , Vitiligo/patologia
17.
J Invest Dermatol ; 138(3): 588-597, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29054606

RESUMO

Autoreactive B-cell activation and antibody production are critical events for the development of bullous pemphigoid (BP). However, the mechanism that is involved in the modulation of B-cell activation and autoantibody generation has not been fully understood. Semaphorin 4D (Sema4D, or CD100) plays important roles in immune regulation related to B cells, but its implications in BP remain obscure. The aim of our study was to characterize Sema4D and the underlying mechanism contributing to the autoimmune features of BP. We found that soluble Sema4D (sSema4D) levels were elevated and correlated with disease severity and activity in serum and blister fluids from patients with BP. Additionally, Sema4D-expressing cells accumulated in subepidermal blisters of BP lesions. In patient-derived peripheral blood mononuclear cells, by promoting the differentiation of B cells into plasmablasts, sSema4D boosted anti-BP180/anti-BP230 antibody production in a time- and dose-dependent manner, which may be attributed to CD72-mediated activation of Akt/NF-κB phosphorylated (p-)65/ERK cascades in B cells. We determined that a disintegrin and metalloproteinase 10 is a proteolytic enzyme for the cleavage of sSema4D from CD15+ granulocytes instead of T cells, which is probably responsible for the high concentration of sSema4D in BP blister fluid and serum. These findings suggest that Sema4D is a crucial participant in BP pathogenesis.


Assuntos
Proteína ADAM10/fisiologia , Antígenos CD/fisiologia , Autoantígenos/imunologia , Fucosiltransferases/análise , Antígenos CD15/análise , Colágenos não Fibrilares/imunologia , Penfigoide Bolhoso/imunologia , Semaforinas/fisiologia , Formação de Anticorpos , Antígenos CD/análise , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Granulócitos/imunologia , Humanos , NF-kappa B/fisiologia , Semaforinas/análise , Colágeno Tipo XVII
18.
Cell Death Dis ; 9(5): 567, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29752469

RESUMO

Keratinocytes are the main epidermal cell type that constitutes the skin barrier against environmental damages, which emphasizes the balance between the growth and the death of keratinocytes in maintaining skin homeostasis. Aberrant proliferation of keratinocytes and the secretion of inflammatory factors from keratinocytes are related to the formation of chronic inflammatory skin diseases like psoriasis. MicroRNA-17-92 (miRNA-17-92 or miR-17-92) is a miRNA cluster that regulates cell growth and immunity, but the role of miR-17-92 cluster in keratinocytes and its relation to skin diseases have not been well investigated. In the present study, we initially found that miR-17-92 cluster promoted the proliferation and the cell-cycle progression of keratinocytes via suppressing cyclin-dependent kinase inhibitor 2B (CDKN2B). Furthermore, miR-17-92 cluster facilitated the secretion of C-X-C motif chemokine ligand 9 (CXCL9) and C-X-C motif chemokine ligand 10 (CXCL10) from keratinocytes by inhibiting suppressor of cytokine signaling 1 (SOCS1), which enhanced the chemotaxis for T lymphocytes formed by keratinocytes. In addition, we detected increased expression of miR-17-92 cluster in psoriatic lesions and the level of lesional miR-17-92 cluster was positively correlated with the disease severity in psoriasis patients. At last, miR-17-92 cluster was increased in keratinocytes by cytokines through the activation of signal transducers and activators of transcription 1 (STAT1) signaling pathway. Our findings demonstrate that cytokine-induced overexpression of miR-17-92 cluster can promote the proliferation and the immune function of keratinocytes, and thus may contribute to the development of inflammatory skin diseases like psoriasis, which implicates miR-17-92 cluster as a potential therapeutic target for psoriasis and other skin diseases with similar inflammatory pathogenesis.


Assuntos
Proliferação de Células , Quimiocinas/biossíntese , Queratinócitos/metabolismo , MicroRNAs/metabolismo , Família Multigênica , Psoríase/metabolismo , Quimiocinas/genética , Feminino , Humanos , Queratinócitos/patologia , Masculino , MicroRNAs/genética , Psoríase/genética , Psoríase/patologia , RNA Longo não Codificante
19.
J Invest Dermatol ; 137(11): 2396-2406, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28760660

RESUMO

Chemokine-mediated CD8+ T-cell recruitment is an essential but not well-established event for the persistence of oral lichen planus (OLP). Semaphorin 4D (Sema4D)/CD100 is implicated in immune dysfunction, chemokine modulation, and cell migration, which are critical aspects for OLP progression, but its implication in OLP pathogenesis has not been determined. In this study, we sought to explicate the effect of Sema4D on human oral keratinocytes and its capacity to drive CD8+ T-cell lesional trafficking via chemokine modulation. We found that upregulations of sSema4D in OLP tissues and blood were positively correlated with disease severity and activity. In vitro observation revealed that Sema4D induced C-X-C motif chemokine ligand 9/C-X-C motif chemokine ligand 10 production by binding to plexin-B1 via protein kinase B-NF-κB cascade in human oral keratinocytes, which elicited OLP CD8+ T-cell migration. We also confirmed using clinical samples that elevated C-X-C motif chemokine ligand 9/C-X-C motif chemokine ligand 10 levels were positively correlated with sSema4D levels in OLP lesions and serum. Notably, we determined matrix metalloproteinase-9 as a new proteolytic enzyme for the cleavage of sSema4D from the T-cell surface, which may contribute to the high levels of sSema4D in OLP lesions and serum. Our findings conclusively revealed an amplification feedback loop involving T cells, chemokines, and Sema4D-dependent signal that promotes OLP progression.


Assuntos
Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL9/genética , Líquen Plano Bucal/genética , Líquen Plano Bucal/imunologia , Semaforinas/metabolismo , Administração Tópica , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Biópsia por Agulha , Linfócitos T CD8-Positivos/imunologia , Estudos de Casos e Controles , Movimento Celular/genética , Células Cultivadas , Humanos , Imuno-Histoquímica , Queratinócitos/metabolismo , Queratinócitos/patologia , Líquen Plano Bucal/tratamento farmacológico , Líquen Plano Bucal/patologia , RNA Interferente Pequeno/análise , Triancinolona/uso terapêutico , Regulação para Cima
20.
J Invest Dermatol ; 137(6): 1286-1296, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28174051

RESUMO

The prevention of hydrogen peroxide (H2O2)-induced oxidative stress has proved to be beneficial to vitiligo patients. Simvastatin possesses antioxidative capacity and has shown protective effect in various oxidative stress-related diseases. However, whether simvastatin can protect human melanocytes against oxidative stress has not been investigated. In this study, we initially found that pretreatment with 0.1 µmol/L to 1.0 µmol/L simvastatin led to increased cell viability and decreased cell apoptosis of melanocytes in response to H2O2. In addition, simvastatin was able to potentiate the activity of antioxidant enzymes and lessen intracellular reactive oxygen species accumulation. Furthermore, we found that simvastatin promoted the activation of nuclear erythroid 2-related factor (Nrf2) and that knockdown of Nrf2 abolished the protective effect of simvastatin against H2O2-induced oxidative damage. More importantly, the mutual enhancement between mitogen-activated protein kinase pathways and p62 contributed to simvastatin-induced Nrf2 activation in melanocytes. Finally, simvastatin showed more antioxidative capacity and better protective effect than aspirin in H2O2-treated melanocytes. Taken together, our results show that simvastatin protects human melanocytes from H2O2-induced oxidative stress by activating Nrf2, thus supporting simvastatin as a potential therapeutic agent for vitiligo.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Peróxido de Hidrogênio/toxicidade , Melanócitos/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , Sinvastatina/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Sobrevivência Celular/genética , Células Cultivadas , Imunofluorescência , Regulação da Expressão Gênica , Humanos , Peróxido de Hidrogênio/farmacologia , Melanócitos/citologia , Fator 2 Relacionado a NF-E2/genética , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Sensibilidade e Especificidade , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa