Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Lasers Surg Med ; 56(5): 425-436, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38769894

RESUMO

OBJECTIVES: Hidradenitis suppurativa (HS) is a chronic inflammatory condition characterized by painful nodules, draining tunnels, and fibrotic scarring in intertriginous, hair-bearing areas. The pathogenesis involves follicular occlusion and subsequent rupture, leading to uncontrolled inflammation. Treatment options for HS are limited and lack universal effectiveness. Laser hair removal (LHR) has been explored as a potential treatment; however, the efficacy and appropriate laser modalities remain unclear. This systematic review examined the efficacy and adverse effects of LHR in HS. METHODS: A comprehensive literature search was conducted from inception to September 2023 in Ovid MEDLINE, Ovid Embase, and The Cochrane Library (Wiley) with predefined inclusion and exclusion criteria, and a meta-analysis was conducted. RESULTS: Ten studies were selected (n = 227 total patients) and included six randomized controlled trials, two nonrandomized experimental studies, and two case series. Various laser modalities, including long-pulsed neodymium-doped yttrium aluminum garnet (Nd:YAG) (n = 115), intense pulsed light (n = 18), Alexandrite (n = 54), intralesional 1064 nm diode (n = 20), and combined fractional CO2 and long-pulsed Nd:YAG laser (n = 20), consistently demonstrated significant improvement in HS disease severity, irrespective of the disease scoring method used. Minimal adverse effects (primarily mild pain and erythema) were reported. A meta-analysis of three studies utilizing long-pulsed Nd:YAG laser demonstrated a standardized mean difference in disease severity of -1.68 (95% confidence interval: -2.99; -0.37), favoring treatment with LHR for HS. CONCLUSIONS: Hair follicles are key in HS pathogenesis and all included studies showed a significant improvement in HS disease severity after LHR regardless of the laser device used, likely related to hair follicle unit destruction. HS is a complex and heterogenous condition, and multiple disease scoring methods complicate outcome comparisons across studies. However, LHR, utilizing various techniques, is an effective treatment option for HS with minimal adverse effects.


Assuntos
Remoção de Cabelo , Hidradenite Supurativa , Humanos , Hidradenite Supurativa/cirurgia , Hidradenite Supurativa/terapia , Remoção de Cabelo/métodos , Resultado do Tratamento , Terapia a Laser/métodos , Lasers de Estado Sólido/uso terapêutico
2.
J Am Acad Dermatol ; 82(3): 709-722, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31306730

RESUMO

BACKGROUND: Hydroxychloroquine is widely used for the treatment of cutaneous lupus erythematosus (CLE). Although new recommendations exist for hydroxychloroquine dosing, there is still uncertainty about the dosage that will elicit a satisfactory response in CLE while limiting adverse effects, specifically retinopathy. OBJECTIVE: To summarize hydroxychloroquine dosages, outcomes, and adverse effects in the treatment of CLE, focusing on retinopathy. METHODS: A comprehensive literature search from inception to December 2018 was performed in Ovid MEDLINE, Ovid Embase, and The Cochrane Library (Wiley). Studies were screened against predefined inclusion and exclusion criteria. RESULTS: Twelve studies were selected and included 5 retrospective studies, 3 prospective studies, 2 case series, and 2 randomized controlled trials. These studies show that a hydroxychloroquine dosage up to 400 mg/d is effective for most CLE patients (range of effectiveness, 50%-97%), with few adverse effects. One incidence of retinopathy, after a very high cumulative dose, was reported across all 12 studies (852 total patients). LIMITATIONS: Because retinopathy and other serious adverse effects may not appear until much later, many of these studies are limited by short follow-up time. CONCLUSIONS: This evidence suggests that hydroxychloroquine for CLE is effective at 400 mg/d, with an exceedingly low incidence of retinopathy and other adverse effects.


Assuntos
Hidroxicloroquina/uso terapêutico , Lúpus Eritematoso Cutâneo/tratamento farmacológico , Humanos
3.
Trends Immunol ; 37(11): 764-777, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27638128

RESUMO

During normal and pathologic immune responses, lymph nodes can swell considerably. The lymph node vascular-stromal compartment supports and regulates the developing immune responses and undergoes dynamic expansion and remodeling. Recent studies have shown that dendritic cells (DCs), best known for their antigen presentation roles, can directly regulate the vascular-stromal compartment, pointing to a new perspective on DCs as facilitators of lymphoid tissue function. Here, we review the phases of lymph node vascular-stromal growth and remodeling during immune responses, discuss the roles of DCs, and discuss how this understanding can potentially be used for developing novel therapeutic approaches.


Assuntos
Apresentação de Antígeno , Células Dendríticas/imunologia , Imunidade , Imunoterapia/métodos , Linfonodos/imunologia , Células Estromais/fisiologia , Remodelação Vascular/imunologia , Animais , Antígenos/imunologia , Antígenos/metabolismo , Comunicação Celular/imunologia , Humanos , Imunoterapia/tendências , Peptídeos/imunologia , Peptídeos/metabolismo
8.
bioRxiv ; 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39071369

RESUMO

The communication between skin and draining lymph nodes is crucial for well-regulated immune responses to skin insults. The skin sends antigen and other signals via lymphatic vessels to regulate lymph node activity, and regulating dermal lymphatic function is another means to control immunity. Here, we show that Langerhans cells (LCs), epidermis-derived antigen-presenting cells, mediate dermal lymphatic expansion and phenotype acquisition postnatally, a function is independent of LC entry into lymphatic vessels. This postnatal LC-lymphatic axis serves in part to control inflammatory systemic T cell responses in adulthood. Our data provide a tissue-based mechanism by which LCs regulate T cells remotely across time and space and raise the possibility that immune diseases in adulthood could reflect compromise of the LC-lymphatic axis in childhood.

9.
Elife ; 132024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38860651

RESUMO

The autoimmune disease lupus erythematosus (lupus) is characterized by photosensitivity, where even ambient ultraviolet radiation (UVR) exposure can lead to development of inflammatory skin lesions. We have previously shown that Langerhans cells (LCs) limit keratinocyte apoptosis and photosensitivity via a disintegrin and metalloprotease 17 (ADAM17)-mediated release of epidermal growth factor receptor (EGFR) ligands and that LC ADAM17 sheddase activity is reduced in lupus. Here, we sought to understand how the lupus skin environment contributes to LC ADAM17 dysfunction and, in the process, differentiate between effects on LC ADAM17 sheddase function, LC ADAM17 expression, and LC numbers. We show through transcriptomic analysis a shared IFN-rich environment in non-lesional skin across human lupus and three murine models: MRL/lpr, B6.Sle1yaa, and imiquimod (IMQ) mice. IFN-I inhibits LC ADAM17 sheddase activity in murine and human LCs, and IFNAR blockade in lupus model mice restores LC ADAM17 sheddase activity, all without consistent effects on LC ADAM17 protein expression or LC numbers. Anti-IFNAR-mediated LC ADAM17 sheddase function restoration is associated with reduced photosensitive responses that are dependent on EGFR signaling and LC ADAM17. Reactive oxygen species (ROS) is a known mediator of ADAM17 activity; we show that UVR-induced LC ROS production is reduced in lupus model mice, restored by anti-IFNAR, and is cytoplasmic in origin. Our findings suggest that IFN-I promotes photosensitivity at least in part by inhibiting UVR-induced LC ADAM17 sheddase function and raise the possibility that anifrolumab ameliorates lupus skin disease in part by restoring this function. This work provides insight into IFN-I-mediated disease mechanisms, LC regulation, and a potential mechanism of action for anifrolumab in lupus.


Assuntos
Proteína ADAM17 , Células de Langerhans , Lúpus Eritematoso Sistêmico , Pele , Proteína ADAM17/metabolismo , Proteína ADAM17/genética , Animais , Humanos , Células de Langerhans/metabolismo , Camundongos , Pele/metabolismo , Pele/patologia , Pele/efeitos da radiação , Lúpus Eritematoso Sistêmico/metabolismo , Raios Ultravioleta/efeitos adversos , Feminino , Modelos Animais de Doenças , Transtornos de Fotossensibilidade/metabolismo , Interferons/metabolismo , Camundongos Endogâmicos MRL lpr
10.
J Rheumatol ; 49(9): 1026-1030, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35649551

RESUMO

OBJECTIVE: In antiphospholipid antibody (aPL) nephropathy, activation of the mammalian target of rapamycin (mTOR) contributes to endothelial cell proliferation, a key finding of aPL microvascular disease. Here, we examined mTOR activation in the skin of aPL-positive patients with livedo. METHODS: Three patient groups with livedo were studied: (1) persistently aPL-positive with systemic lupus erythematosus (SLE); (2) persistently aPL-positive without SLE; and (3) aPL-negative SLE (control). After collecting aPL-related medical history, two 5-mm skin biopsies of livedo were performed on each patient: (1) peripheral (erythematous-violaceous lesion); and (2) central (nonviolaceous area). We stained specimens for phosphorylated protein kinase B (p-AKT) and phosphorylated S6 ribosomal protein (p-S6RP) as mTOR activity markers, CD31 to identify endothelial cells, and Ki-67 to show cellular proliferation. We counted cells in the epidermis and compared mTOR-positive cell counts between peripheral and central samples, and between patient groups, using Freidman test and Wilcoxon signed-rank test. RESULTS: Ten patients with livedo reticularis were enrolled: 4 aPL-positive without SLE (antiphospholipid syndrome [APS] classification met, n = 3), 4 aPL-positive SLE (APS classification met, n = 3), and 2 aPL-negative SLE (control). In all aPL-positive patients, epidermal p-AKT and p-S6RP staining were significantly increased in both peripheral and central skin samples when compared to aPL-negative SLE controls; both were more pronounced in the lower basal layers of epidermis. CONCLUSION: Our study demonstrates increased mTOR activity in livedoid lesions of aPL-positive patients with or without SLE compared to aPL-negative patients with SLE, with more prominent activity in the lower basal layers of the epidermis. These findings may serve as a basis for further investigating the mTOR pathway in aPL-positive patients.


Assuntos
Síndrome Antifosfolipídica , Livedo Reticular , Lúpus Eritematoso Sistêmico , Serina-Treonina Quinases TOR , Humanos , Anticorpos Antifosfolipídeos , Células Endoteliais , Antígeno Ki-67 , Proteínas Proto-Oncogênicas c-akt , Proteínas Ribossômicas , Sirolimo
11.
J Invest Dermatol ; 142(3 Pt B): 924-935, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34606886

RESUMO

Hidradenitis suppurativa (HS), also known as acne inversa, is a debilitating inflammatory skin disorder that is characterized by nodules that lead to the development of connected tunnels and scars as it progresses from Hurley stages I to III. HS has been associated with several autoimmune diseases, including inflammatory bowel disease and spondyloarthritis. We previously reported dysregulation of humoral immune responses in HS, characterized by elevated serum total IgG, B-cell activation, and antibodies recognizing citrullinated proteins. In this study, we characterized IgG autoreactivity in HS sera and lesional skin compared with those in normal healthy controls using an array-based high-throughput autoantibody screening. The Cy3-labeled anti-human assay showed the presence of autoantibodies against nuclear antigens, cytokines, cytoplasmic proteins, extracellular matrix proteins, neutrophil proteins, and citrullinated antigens. Most of these autoantibodies were significantly elevated in stages II‒III in HS sera and stage III in HS skin lesions compared with those of healthy controls. Furthermore, immune complexes containing both native and citrullinated versions of antigens can activate M1 and M2 macrophages to release proinflammatory cytokines such as TNF-α, IL-8, IL-6, and IL-12. Taken together, the identification of specific IgG autoantibodies that recognize circulating and tissue antigens in HS suggests an autoimmune mechanism and uncovers putative therapeutic targets.


Assuntos
Hidradenite Supurativa , Antígenos , Autoanticorpos , Citocinas/metabolismo , Hidradenite Supurativa/diagnóstico , Humanos , Imunoglobulina G , Macrófagos/metabolismo , Índice de Gravidade de Doença
12.
Curr Opin Immunol ; 64: 63-70, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32387902

RESUMO

The recent advent of single-cell technologies has fast-tracked the discovery of multiple fibroblast subsets in tissues affected by autoimmune disease. In recent years, interest in lymph node fibroblasts that support and regulate immune cells has also grown, leading to an expanding framework of stromal cell subsets with distinct spatial, transcriptional, and functional characteristics. Inflammation can drive tissue fibroblasts to adopt a lymphoid tissue stromal cell phenotype, suggesting that fibroblasts in diseased tissues can have counterparts in lymphoid tissues. Here, we examine fibroblast subsets in tissues affected by autoimmunity in the context of knowledge gained from studies on lymph node fibroblasts, with the ultimate aim to better understand stromal cell heterogeneity in these immunologically reactive tissues.


Assuntos
Autoimunidade , Fibroblastos , Humanos , Linfonodos , Tecido Linfoide , Células Estromais
13.
Sci Immunol ; 5(45)2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-32198221

RESUMO

Nonhematopoietic stromal cells in lymph nodes such as fibroblastic reticular cells (FRCs) can support the survival of plasmablasts and plasma cells [together, antibody-forming cells (AFCs)]. However, a regulatory function for the stromal compartment in AFC accumulation has not been appreciated. Here, we show that chemokine ligand 2 (CCL2)-expressing stromal cells limit AFC survival. FRCs express high levels of CCL2 in vessel-rich areas of the T cell zone and the medulla, where AFCs are located. FRC CCL2 is up-regulated during AFC accumulation, and we use lymph node transplantation to show that CCL2 deficiency in BP3+ FRCs and lymphatic endothelial cells increases AFC survival without affecting B or germinal center cell numbers. Monocytes are key expressers of the CCL2 receptor CCR2, as monocyte depletion and transfer late in AFC responses increases and decreases AFC accumulation, respectively. Monocytes express reactive oxygen species (ROS) in an NADPH oxidase 2 (NOX2)-dependent manner, and NOX2-deficient monocytes fail to reduce AFC numbers. Stromal CCL2 modulates both monocyte accumulation and ROS production, and is regulated, in part, by manipulations that modulate vascular permeability. Together, our results reveal that the lymph node stromal compartment, by influencing monocyte accumulation and functional phenotype, has a regulatory role in AFC survival. Our results further suggest a role for inflammation-induced vascular activity in tuning the lymph node microenvironment. The understanding of stromal-mediated AFC regulation in vessel-rich environments could potentially be harnessed to control antibody-mediated autoimmunity.


Assuntos
Anticorpos/imunologia , Quimiocina CCL2/imunologia , Células Estromais/imunologia , Animais , Formação de Anticorpos/imunologia , Linfonodos/citologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
Front Immunol ; 10: 519, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30949174

RESUMO

Lymphatic vessels are critical for clearing fluid and inflammatory cells from inflamed tissues and also have roles in immune tolerance. Given the functional association of the lymphatics with the immune system, lymphatic dysfunction may contribute to the pathophysiology of rheumatic autoimmune diseases. Here we review the current understanding of the role of lymphatics in the autoimmune diseases rheumatoid arthritis, scleroderma, lupus, and dermatomyositis and consider the possibility that manual therapies such as massage and acupuncture may be useful in improving lymphatic function in autoimmune diseases.


Assuntos
Artrite Reumatoide/imunologia , Vasos Linfáticos/imunologia , Animais , Artrite Reumatoide/patologia , Humanos , Vasos Linfáticos/patologia
15.
Sci Transl Med ; 10(454)2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30111646

RESUMO

Photosensitivity, or skin sensitivity to ultraviolet radiation (UVR), is a feature of lupus erythematosus and other autoimmune and dermatologic conditions, but the mechanistic underpinnings are poorly understood. We identify a Langerhans cell (LC)-keratinocyte axis that limits UVR-induced keratinocyte apoptosis and skin injury via keratinocyte epidermal growth factor receptor (EGFR) stimulation. We show that the absence of LCs in Langerin-diphtheria toxin subunit A (DTA) mice leads to photosensitivity and that, in vitro, mouse and human LCs can directly protect keratinocytes from UVR-induced apoptosis. LCs express EGFR ligands and a disintegrin and metalloprotease 17 (ADAM17), the metalloprotease that activates EGFR ligands. Deletion of ADAM17 from LCs leads to photosensitivity, and UVR induces LC ADAM17 activation and generation of soluble active EGFR ligands, suggesting that LCs protect by providing activated EGFR ligands to keratinocytes. Photosensitive systemic lupus erythematosus (SLE) models and human SLE skin show reduced epidermal EGFR phosphorylation and LC defects, and a topical EGFR ligand reduces photosensitivity. Together, our data establish a direct tissue-protective function for LCs, reveal a mechanistic basis for photosensitivity, and suggest EGFR stimulation as a treatment for photosensitivity in lupus erythematosus and potentially other autoimmune and dermatologic conditions.


Assuntos
Citoproteção/efeitos da radiação , Queratinócitos/citologia , Queratinócitos/efeitos da radiação , Células de Langerhans/citologia , Células de Langerhans/efeitos da radiação , Raios Ultravioleta , Proteína ADAM17/metabolismo , Animais , Apoptose/efeitos da radiação , Modelos Animais de Doenças , Epiderme/metabolismo , Epiderme/efeitos da radiação , Receptores ErbB/metabolismo , Humanos , Ligantes , Lúpus Eritematoso Sistêmico/patologia , Camundongos Endogâmicos C57BL , Fosforilação/efeitos da radiação
16.
F1000Res ; 6: 196, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28344775

RESUMO

Tertiary lymphoid organs are found at sites of chronic inflammation in autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis. These organized accumulations of T and B cells resemble secondary lymphoid organs and generate autoreactive effector cells. However, whether they contribute to disease pathogenesis or have protective functions is unclear. Here, we discuss how tertiary lymphoid organs can generate potentially pathogenic cells but may also limit the extent of the response and damage in autoimmune disease.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA