Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 98: 107904, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34214886

RESUMO

Dysregulation of phosphoinositide 3-kinase δ (PI3Kδ) signaling pathway has been implicated in the pathogenesis of inflammatory and autoimmune diseases. Parsaclisib (INCB050465) represents a potent and selective PI3Kδ inhibitor, which is being clinically investigated for treatment of autoimmune hemolytic anemia and hematological malignancies. We characterized the potential of parsaclisib to ameliorate autoimmune mechanisms implicated in the pathophysiology of systemic lupus erythematosus (SLE) and Sjögren's syndrome (SS). Spontaneous mouse models of SLE and SS were utilized to elucidate the efficacy of orally administered parsaclisib on autoreactive B-cell-mediated antibody-driven disease. Parsaclisib significantly reduced disease symptoms and pathology in three distinct mouse models of SLE. Parsaclisib effectively preserved renal function as measured by glomerular filtration rate, abrogated histopathological evidence of nephritis, modulated discrete immune cell subsets, and decreased anti-dsDNA antibody level. Furthermore, parsaclisib demonstrated efficacy in two spontaneous mouse models of SS. Oral parsaclisib treatment ameliorated the severity of salivary gland inflammation and reduced circulating levels of autoantibodies. Parsaclisib mediated improvement of salivary gland inflammation coincided with reduced B-cell activating cytokine (BAFF) in saliva. Transcriptomic analysis of kidney and salivary gland tissues revealed a downregulation in inflammatory gene expression consistent with PI3Kδ pathway inhibition. Parsaclisib reduced autoreactive B-cells and autoantibody levels, and significantly improved nephritis and salivary gland inflammation. These data provide the scientific rationale for PI3Kδ inhibition as a therapeutic strategy for treatment of B-cell-mediated antibody-driven autoimmune diseases.


Assuntos
Autoanticorpos/sangue , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Pirazóis/farmacologia , Pirimidinas/farmacologia , Pirrolidinas/farmacologia , Síndrome de Sjogren/tratamento farmacológico , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Modelos Animais de Doenças , Humanos , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Pirrolidinas/uso terapêutico , Síndrome de Sjogren/sangue , Síndrome de Sjogren/imunologia
2.
Front Pharmacol ; 12: 650295, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33981229

RESUMO

Hyperinflammatory syndromes comprise a heterogeneous group of disorders characterized by severe inflammation, multiple organ dysfunction, and potentially death. In response to antigenic stimulus (e.g., SARS-CoV-2 infection), overactivated CD8+ T-cells and macrophages produce high levels of proinflammatory cytokines, such as IFN-γ, TNF-α, IL-6, and IL-12. Multiple inflammatory mediators implicated in hyperinflammatory syndromes utilize the Janus kinase-signal transducers and activators of transcription (JAK-STAT) cascade to propagate their biological function. Our findings demonstrate that oral ruxolitinib dosing designed to mimic clinically relevant JAK-STAT pathway inhibition significantly reduces the harmful consequences of immune overactivation in multiple hyperinflammatory models. In contrast to monoclonal antibody therapies targeting a single cytokine, ruxolitinib effectively downregulates the functional effect of multiple cytokines implicated in hyperinflammatory states, without broad immunosuppression.

3.
Front Immunol ; 11: 620098, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33658996

RESUMO

The goal of this study was to elucidate the anti-pruritic and anti-inflammatory efficacy of ruxolitinib cream in experimentally-induced dermatitis. Atopic dermatitis (AD), the most common chronic relapsing inflammatory skin disease, significantly impairs patients' quality of life, with pruritus being a common complaint. The sensation of itch results from the interplay between epidermal barrier dysfunction, upregulated immune signaling and the activation of the central nervous system. The Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway plays a central role in pro-inflammatory cytokine signaling in AD. Ruxolitinib cream is a potent and selective JAK1/2 inhibitor currently undergoing clinical evaluation in adults with mild-to-moderate AD (NCT03745638, NCT03920852 and NCT03745651). The efficacy of ruxolitinib cream was tested in murine models of acute and chronic dermatitis and was also characterized in an ex vivo human skin dermatitis model. Ruxolitinib cream was highly effective at ameliorating disease symptoms in multiple murine dermatitis models through downregulation of T helper (Th)2-driven inflammation, resulting in reduced skin thickening and decreased itch. Pathway analysis of mouse ear tissue and human skin explants underscored the role for ruxolitinib in ameliorating inflammation and reducing itch via modulation of the JAK-STAT pathway. Together, the data offer a strong rationale for the use of ruxolitinib cream as a potent therapeutic agent for the clinical management of atopic dermatitis.


Assuntos
Dermatite/tratamento farmacológico , Inibidores de Janus Quinases/uso terapêutico , Prurido/tratamento farmacológico , Pirazóis/uso terapêutico , Administração Cutânea , Animais , Betametasona/administração & dosagem , Betametasona/uso terapêutico , Clobetasol/administração & dosagem , Clobetasol/uso terapêutico , Citocinas/biossíntese , Citocinas/genética , Citocinas/toxicidade , Modelos Animais de Doenças , Toxidermias/tratamento farmacológico , Toxidermias/imunologia , Avaliação Pré-Clínica de Medicamentos , Feminino , Fluoresceína-5-Isotiocianato/toxicidade , Asseio Animal/efeitos dos fármacos , Humanos , Técnicas In Vitro , Interleucina-33/genética , Inibidores de Janus Quinases/administração & dosagem , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Nitrilas , Pomadas , Técnicas de Cultura de Órgãos , Pirazóis/administração & dosagem , Pirimidinas , Distribuição Aleatória , Transdução de Sinais/efeitos dos fármacos , Pele/efeitos dos fármacos , Organismos Livres de Patógenos Específicos , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Transcriptoma , Linfopoietina do Estroma do Timo
4.
Front Immunol ; 9: 2231, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30364107

RESUMO

IgE is the key mediator of allergic responses. Omalizumab, an IgE-specific monoclonal antibody that depletes IgE, is effective for treating severe allergic asthma. The need for frequent administration of the expensive drug, however, limits its applications. Taking advantage of T cell memory, adoptive T cell therapy (ACT) targeting IgE-producing cells has the potential to achieve long-term suppression of IgE and relief of symptoms for severe allergic diseases. The transmembrane form of IgE (mIgE), which is present on all IgE-producing cells, serves as an excellent molecular target for ACT that employs chimeric antigen receptors (CARs). Here, we designed and tested CARs that use the extracellular domain of high affinity IgE receptor, FcεRIα, for mIgE recognition. When expressed on Jurkat T cells, FcεRIα-based CARs mediated robust responses in terms of CD69 upregulation to U266 myeloma cells expressing low levels of mIgE. FcεRIα-based CARs specifically recognized cells expressing mIgE, but not cells with secreted IgE captured through Fcε receptors. CAR+ Jurkat cells did not respond to LAD2 mast cells with secreted IgE bound through FcεRI or Ramos cells with secreted IgE bound through FcεRII. Co-culture of CAR+ Jurkat cells and LAD2 mast cells with IgE bound did not trigger LAD2 cell degranulation. The activity of CAR using wild type FcεRIα for mIgE binding was inhibited by the presence secreted IgE, which likely blocked CAR-mIgE interaction. The activities of CARs using low affinity mutants of FcεRIα, however, tolerated secreted IgE at relatively high concentrations. Moreover, primary human CD8+ T cells expressing a low affinity mutant CAR responded to U266 cells with INFγ production and cytotoxicity despite the presence of secreted IgE. The potency, specificity, and robustness of our CAR design, combined with repaid advances in the safety of ACT, hold promise for novel and highly effective cell-based therapies against severe allergic diseases.


Assuntos
Imunoglobulina E/imunologia , Receptores de Antígenos Quiméricos/imunologia , Receptores de IgE/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Afinidade de Anticorpos/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Asma/imunologia , Asma/terapia , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Imunoglobulina E/genética , Imunoglobulina E/metabolismo , Imunoterapia Adotiva/métodos , Células Jurkat , Mutação , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Receptores de IgE/genética , Receptores de IgE/metabolismo , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo
5.
Int J Nanomedicine ; 10: 6931-41, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26609231

RESUMO

Nanoshell-mediated photothermal therapy (PTT) is currently being investigated as a standalone therapy for the treatment of cancer. The cellular effects of PTT include loss of membrane integrity, so we hypothesized that nanoshell-mediated PTT could potentiate the cytotoxicity of chemotherapy by improving drug accumulation in cancer cells. In this work, we validated our hypothesis using doxorubicin as a model drug and SUM149 inflammatory breast cancer cells as a model cancer subtype. In initial studies, SUM149 cells were exposed to nano-shells and near-infrared light and then stained with ethidium homodimer-1, which is excluded from cells with an intact plasma membrane. The results confirmed that nanoshell-mediated PTT could increase membrane permeability in SUM149 cells. In complementary experiments, SUM149 cells treated with nanoshells, near-infrared light, or a combination of the two to yield low-dose PTT were exposed to fluorescent rhodamine 123. Analyzing rhodamine 123 fluorescence in cells via flow cytometry confirmed that increased membrane permeability caused by PTT could enhance drug accumulation in cells. This was validated using fluorescence microscopy to assess intracellular distribution of doxorubicin. In succeeding experiments, SUM149 cells were exposed to subtherapeutic levels of doxorubicin, low-dose PTT, or a combination of the two treatments to determine whether the additional drug uptake induced by PTT is sufficient to enhance cell death. Analysis revealed minimal loss of viability relative to controls in cells exposed to subtherapeutic levels of doxorubicin, 15% loss of viability in cells exposed to low-dose PTT, and 35% loss of viability in cells exposed to combination therapy. These data indicate that nanoshell-mediated PTT is a viable strategy to potentiate the effects of chemotherapy and warrant further investigation of this approach using other drugs and cancer subtypes.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Inflamatórias Mamárias/tratamento farmacológico , Nanoconchas/química , Fototerapia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Feminino , Humanos , Hipertermia Induzida , Raios Infravermelhos , Nanoconchas/ultraestrutura , Rodaminas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA