Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
J Allergy Clin Immunol ; 143(4): 1426-1443.e6, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30240702

RESUMEN

BACKGROUND: Atopic dermatitis (AD) is associated with epidermal barrier defects, dysbiosis, and skin injury caused by scratching. In particular, the barrier-defective epidermis in patients with AD with loss-of-function filaggrin mutations has increased IL-1α and IL-1ß levels, but the mechanisms by which IL-1α, IL-1ß, or both are induced and whether they contribute to the aberrant skin inflammation in patients with AD is unknown. OBJECTIVE: We sought to determine the mechanisms through which skin injury, dysbiosis, and increased epidermal IL-1α and IL-1ß levels contribute to development of skin inflammation in a mouse model of injury-induced skin inflammation in filaggrin-deficient mice without the matted mutation (ft/ft mice). METHODS: Skin injury of wild-type, ft/ft, and myeloid differentiation primary response gene-88-deficient ft/ft mice was performed, and ensuing skin inflammation was evaluated by using digital photography, histologic analysis, and flow cytometry. IL-1α and IL-1ß protein expression was measured by means of ELISA and visualized by using immunofluorescence and immunoelectron microscopy. Composition of the skin microbiome was determined by using 16S rDNA sequencing. RESULTS: Skin injury of ft/ft mice induced chronic skin inflammation involving dysbiosis-driven intracellular IL-1α release from keratinocytes. IL-1α was necessary and sufficient for skin inflammation in vivo and secreted from keratinocytes by various stimuli in vitro. Topical antibiotics or cohousing of ft/ft mice with unaffected wild-type mice to alter or intermix skin microbiota, respectively, resolved the skin inflammation and restored keratinocyte intracellular IL-1α localization. CONCLUSIONS: Taken together, skin injury, dysbiosis, and filaggrin deficiency triggered keratinocyte intracellular IL-1α release that was sufficient to drive chronic skin inflammation, which has implications for AD pathogenesis and potential therapeutic targets.


Asunto(s)
Dermatitis Atópica/metabolismo , Inflamación/metabolismo , Interleucina-1alfa/metabolismo , Proteínas de Filamentos Intermediarios/deficiencia , Queratinocitos/metabolismo , Animales , Dermatitis Atópica/inmunología , Dermatitis Atópica/microbiología , Disbiosis/inmunología , Disbiosis/metabolismo , Proteínas Filagrina , Inflamación/inmunología , Inflamación/microbiología , Interleucina-1alfa/inmunología , Queratinocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
2.
Clin Immunol ; 195: 88-92, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30099194

RESUMEN

Mutations in filaggrin are associated with atopic dermatitis. Filaggrin-deficient flaky tail (Flgft/ft) mice develop spontaneous inflammatory skin lesion that wax and wane. We show that loss of MyD88 promotes the persistence of skin lesions in Flgft/ft mice and exaggerates their expression of the Th17-associated cytokines Il7a and Il22. The development and persistence of skin lesions in Flgft/ft mice was independent of the microbiota. MyD88-mediated signals are shown to be important for the accumulation of T regulatory cells (Tregs) in lesional skin of Flgft/ft mice. Adoptive transfer of WT Tregs dampened the severity of skin lesions in MyD88-/-/Flgft/ft mice. These results suggest that MyD88 signaling in Treg cells by endogenous ligands attenuates skin inflammation in filaggrin deficiency.


Asunto(s)
Dermatitis Atópica/inmunología , Proteínas de Filamentos Intermediarios/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Piel/inmunología , Linfocitos T Reguladores/fisiología , Células Th17/inmunología , Traslado Adoptivo , Animales , Dermatitis Atópica/genética , Modelos Animales de Enfermedad , Proteínas Filagrina , Humanos , Interleucina-17/metabolismo , Interleucinas/metabolismo , Proteínas de Filamentos Intermediarios/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Factor 88 de Diferenciación Mieloide/genética , Transducción de Señal , Piel/patología , Interleucina-22
3.
Clin Exp Med ; 24(1): 122, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38856863

RESUMEN

Regulatory T cells (Tregs) are known to facilitate tumor progression by suppressing CD8+ T cells within the tumor microenvironment (TME), thereby also hampering the effectiveness of immune checkpoint inhibitors (ICIs). While systemic depletion of Tregs can enhance antitumor immunity, it also triggers undesirable autoimmune responses. Therefore, there is a need for therapeutic agents that selectively target Tregs within the TME without affecting systemic Tregs. In this study, as shown also by others, the chemokine (C-C motif) receptor 8 (CCR8) was found to be predominantly expressed on Tregs within the TME of both humans and mice, representing a unique target for selective depletion of tumor-residing Tregs. Based on this, we developed BAY 3375968, a novel anti-human CCR8 antibody, along with respective surrogate anti-mouse CCR8 antibodies, and demonstrated their in vitro mode-of-action through induction of potent antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) activities. In vivo, anti-mouse CCR8 antibodies effectively depleted Tregs within the TME primarily via ADCP, leading to increased CD8+ T cell infiltration and subsequent tumor growth inhibition across various cancer models. This monotherapeutic efficacy was significantly enhanced in combination with ICIs. Collectively, these findings suggest that CCR8 targeting represents a promising strategy for Treg depletion in cancer therapies. BAY 3375968 is currently under investigation in a Phase I clinical trial (NCT05537740).


Asunto(s)
Receptores CCR8 , Linfocitos T Reguladores , Microambiente Tumoral , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Receptores CCR8/inmunología , Receptores CCR8/antagonistas & inhibidores , Animales , Ratones , Humanos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Femenino , Citotoxicidad Celular Dependiente de Anticuerpos , Depleción Linfocítica , Línea Celular Tumoral , Fagocitosis/efectos de los fármacos , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico
4.
J Exp Clin Cancer Res ; 40(1): 288, 2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34517894

RESUMEN

BACKGROUND: Patients with advanced colorectal cancer (CRC) have a poor prognosis. Combinations of immunotherapies and anti-angiogenic agents are currently being evaluated in clinical trials. In this study, the multikinase inhibitor regorafenib (REG) was combined with an anti-programmed cell death protein 1 (aPD1) antibody in syngeneic murine microsatellite-stable (MSS) CT26 and hypermutated MC38 colon cancer models to gain mechanistic insights into potential drug synergism. METHODS: Growth and progression of orthotopic CT26 and subcutaneous MC38 colon cancers were studied under treatment with varying doses of REG and aPD1 alone or in combination. Sustained effects were studied after treatment discontinuation. Changes in the tumor microenvironment were assessed by dynamic contrast-enhanced MRI, and histological and molecular analyses. RESULTS: In both models, REG and aPD1 combination therapy significantly improved anti-tumor activity compared with single agents. However, in the CT26 model, the additive benefit of aPD1 only became apparent after treatment cessation. The combination treatment efficiently prevented tumor regrowth and completely suppressed liver metastasis, whereas the anti-tumorigenic effects of REG alone were abrogated soon after drug discontinuation. During treatment, REG significantly reduced the infiltration of immunosuppressive macrophages and regulatory T (Treg) cells into the tumor microenvironment. aPD1 significantly enhanced intratumoral IFNγ levels. The drugs synergized to induce sustained M1 polarization and durable reduction of Treg cells, which can explain the sustained tumor suppression. CONCLUSIONS: This study highlights the synergistic immunomodulatory effects of REG and aPD1 combination therapy in mediating a sustained inhibition of colon cancer regrowth, strongly warranting clinical evaluation in CRC, including MSS tumors.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Inmunoterapia/métodos , Compuestos de Fenilurea/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Piridinas/uso terapéutico , Animales , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Ratones , Compuestos de Fenilurea/farmacología , Piridinas/farmacología
6.
J Exp Med ; 213(10): 2147-66, 2016 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-27551155

RESUMEN

Atopic dermatitis (AD) is a Th2-dominated inflammatory skin disease characterized by epidermal thickening. Serum levels of IL-22, a cytokine known to induce keratinocyte proliferation, are elevated in AD, and Th22 cells infiltrate AD skin lesions. We show that application of antigen to mouse skin subjected to tape stripping, a surrogate for scratching, induces an IL-22 response that drives epidermal hyperplasia and keratinocyte proliferation in a mouse model of skin inflammation that shares many features of AD. DC-derived IL-23 is known to act on CD4(+) T cells to induce IL-22 production. However, the mechanisms that drive IL-23 production by skin DCs in response to cutaneous sensitization are not well understood. We demonstrate that IL-23 released by keratinocytes in response to endogenous TLR4 ligands causes skin DCs, which selectively express IL-23R, to up-regulate their endogenous IL-23 production and drive an IL-22 response in naive CD4(+) T cells that mediates epidermal thickening. We also show that IL-23 is released in human skin after scratching and polarizes human skin DCs to drive an IL-22 response, supporting the utility of IL-23 and IL-22 blockade in AD.


Asunto(s)
Polaridad Celular , Células Dendríticas/citología , Inmunización , Interleucina-23/metabolismo , Interleucinas/metabolismo , Queratinocitos/metabolismo , Piel/inmunología , Receptor Toll-Like 4/metabolismo , Adulto , Animales , Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Movimiento Celular , Proliferación Celular , Células Dendríticas/metabolismo , Epidermis/patología , Femenino , Hematopoyesis , Humanos , Ligandos , Ganglios Linfáticos/metabolismo , Ratones Endogámicos BALB C , Piel/patología , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA