Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Inflammopharmacology ; 31(3): 1341-1359, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37010718

RESUMO

Diosmin is a flavonoid with promising anti-inflammatory and antioxidant properties. However, it has difficult physicochemical characteristics since its solubility demands a pH level of 12, which has an impact on the drug's bioavailability. The aim of this work is the development and characterization of diosmin nanocrystals using anti-solvent precipitation technique to be used for topical treatment of psoriasis. Results revealed that diosmin nanocrystals stabilized with hydroxypropyl methylcellulose (HPMC E15) in ratio (diosmin:polymer; 1:1) reached the desired particle size (276.9 ± 16.49 nm); provided promising colloidal properties and possessed high drug release profile. Additionally, in-vivo assessment was carried out to evaluate and compare the activities of diosmin nanocrystal gel using three different doses and diosmin powder gel in alleviating imiquimod-induced psoriasis in rats and investigating their possible anti-inflammatory mechanisms. Herein, 125 mg of 5% imiquimod cream (IMQ) was applied topically for 5 consecutive days on the shaved backs of rats to induce psoriasis. Diosmin nanocrystal gel especially in the highest dose used offered the best anti-inflammatory effect. This was confirmed by causing the most statistically significant reduction in the psoriasis area severity index (PASI) score and the serum inflammatory cytokines levels. Furthermore, it was capable of maintaining the balance between T helper (Th17) and T regulatory (Treg) cells. Moreover, it tackled TLR7/8/NF-κB, miRNA-31, AKT/mTOR/P70S6K and elevated the TNFAIP3/A20 (a negative regulator of NF-κB) expression in psoriatic skin tissues. This highlights the role of diosmin nanocrystal gel in tackling imiquimod-induced psoriasis in rats, and thus it could be a novel promising therapy for psoriasis.


Assuntos
Diosmina , MicroRNAs , Nanopartículas , Psoríase , Ratos , Animais , Camundongos , NF-kappa B/metabolismo , Imiquimode/efeitos adversos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor 7 Toll-Like/metabolismo , Receptor 7 Toll-Like/uso terapêutico , Diosmina/efeitos adversos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/uso terapêutico , Transdução de Sinais , Psoríase/induzido quimicamente , Psoríase/tratamento farmacológico , Pele , Serina-Treonina Quinases TOR/metabolismo , Anti-Inflamatórios/uso terapêutico , Modelos Animais de Doenças , Camundongos Endogâmicos BALB C
2.
Acta Neuropathol Commun ; 10(1): 144, 2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36180898

RESUMO

BACKGROUND: Regulatory T cells (Tregs) play a neuroprotective role by suppressing microglia and macrophage-mediated inflammation and modulating adaptive immune reactions. We previously documented that Treg immunomodulatory mechanisms are compromised in Alzheimer's disease (AD). Ex vivo expansion of Tregs restores and amplifies their immunosuppressive functions in vitro. A key question is whether adoptive transfer of ex vivo expanded human Tregs can suppress neuroinflammation and amyloid pathology in a preclinical mouse model. METHODS: An immunodeficient mouse model of AD was generated by backcrossing the 5xFAD onto Rag2 knockout mice (5xFAD-Rag2KO). Human Tregs were expanded ex vivo for 24 days and administered to 5xFAD-Rag2KO. Changes in amyloid burden, microglia characteristics and reactive astrocytes were evaluated using ELISA and confocal microscopy. NanoString Mouse AD multiplex gene expression analysis was applied to explore the impact of ex vivo expanded Tregs on the neuroinflammation transcriptome. RESULTS: Elimination of mature B and T lymphocytes and natural killer cells in 5xFAD-Rag2KO mice was associated with upregulation of 95 inflammation genes and amplified number of reactive microglia within the dentate gyrus. Administration of ex vivo expanded Tregs reduced amyloid burden and reactive glial cells in the dentate gyrus and frontal cortex of 5xFAD-Rag2KO mice. Interrogation of inflammation gene expression documented down-regulation of pro-inflammatory cytokines (IL1A&B, IL6), complement cascade (C1qa, C1qb, C1qc, C4a/b), toll-like receptors (Tlr3, Tlr4 and Tlr7) and microglial activations markers (CD14, Tyrobp,Trem2) following Treg administration. CONCLUSIONS: Ex vivo expanded Tregs with amplified immunomodulatory function, suppressed neuroinflammation and alleviated AD pathology in vivo. Our results provide preclinical evidences for Treg cell therapy as a potential treatment strategy in AD.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Inflamação/metabolismo , Interleucina-6/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/patologia , Doenças Neuroinflamatórias , Receptores Imunológicos/metabolismo , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Receptor 3 Toll-Like/metabolismo , Receptor 3 Toll-Like/uso terapêutico , Receptor 4 Toll-Like/metabolismo , Receptor 7 Toll-Like/metabolismo , Receptor 7 Toll-Like/uso terapêutico
3.
Clin Cancer Res ; 28(20): 4574-4586, 2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-35943291

RESUMO

PURPOSE: DNMT3A mutations confer a poor prognosis in acute myeloid leukemia (AML), but the molecular mechanisms downstream of DNMT3A mutations in disease pathogenesis are not completely understood, limiting targeted therapeutic options. The role of miRNA in DNMT3A-mutant AML pathogenesis is understudied. EXPERIMENTAL DESIGN: DNA methylation and miRNA expression was evaluated in human AML patient samples and in Dnmt3a/Flt3-mutant AML mice. The treatment efficacy and molecular mechanisms of TLR7/8-directed therapies on DNMT3A-mutant AML were evaluated in vitro on human AML patient samples and in Dnmt3a/Flt3-mutant AML mice. RESULTS: miR-196b is hypomethylated and overexpressed in DNMT3A-mutant AML and is associated with poor patient outcome. miR-196b overexpression in DNMT3A-mutant AML is important to maintain an immature state and leukemic cell survival through repression of TLR signaling. The TLR7/8 agonist resiquimod induces dendritic cell-like differentiation with costimulatory molecule expression in DNMT3A-mutant AML cells and provides a survival benefit to Dnmt3a/Flt3-mutant AML mice. The small molecule bryostatin-1 augments resiquimod-mediated AML growth inhibition and differentiation. CONCLUSIONS: DNMT3A loss-of-function mutations cause miRNA locus-specific hypomethylation and overexpression important for mutant DNMT3A-mediated pathogenesis and clinical outcomes. Specifically, the overexpression of miR-196b in DNMT3A-mutant AML creates a novel therapeutic vulnerability by controlling sensitivity to TLR7/8-directed therapies.


Assuntos
Leucemia Mieloide Aguda , MicroRNAs , Animais , Briostatinas/uso terapêutico , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Metilases de Modificação do DNA/genética , Humanos , Imunidade Inata , Leucemia Mieloide Aguda/tratamento farmacológico , Camundongos , MicroRNAs/genética , MicroRNAs/uso terapêutico , Mutação , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/uso terapêutico
4.
J Immunol ; 184(9): 5360-7, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20308630

RESUMO

Multiple TLR agonists have been shown to have antitumor effects in animal models. However, the therapeutic efficacy of TLR agonist monotherapy in cancer treatment has been limited, and the mechanisms of failure remain unknown. We demonstrate that topical treatment with a TLR-7 agonist, imiquimod, can elicit significant regression of spontaneous breast cancers in neu transgenic mice, a model of human HER-2/neu(+) breast cancer. However, tumor growth progressed once imiquimod therapy was ended. Gene expression analysis using tumor-derived RNA demonstrated that imiquimod induced high levels of IL-10 in addition to TNF-alpha and IFN-gamma. Elevated levels of circulating IL-10 were also detected in sera from imiquimod-treated mice. Elevated serum IL-10 appeared to be derived from IL-10 and dual cytokine secreting (IFN-gamma(+) and IL-10(+)) CD4(+) T cells rather than CD4(+)CD25(+)Foxp3(+) T regulatory cells, which were also induced by imiquimod treatment. Blockade of IL-10, but not TGF-beta, enhanced the antitumor effect of imiquimod by significantly prolonging survival in treated mice. These data suggest that the excessive inflammation induced by TLR agonists may result in a self-regulatory immunosuppression via IL-10 induction and that blocking IL-10 could enhance the therapeutic efficacy of these agents.


Assuntos
Mediadores da Inflamação/fisiologia , Interleucina-10/antagonistas & inibidores , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Glicoproteínas de Membrana/agonistas , Invasividade Neoplásica/patologia , Receptor 7 Toll-Like/agonistas , Doença Aguda , Administração Tópica , Aminoquinolinas/metabolismo , Aminoquinolinas/uso terapêutico , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Inibidores do Crescimento/agonistas , Inibidores do Crescimento/metabolismo , Inibidores do Crescimento/uso terapêutico , Imiquimode , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/uso terapêutico , Interleucina-10/sangue , Ligantes , Neoplasias Mamárias Experimentais/imunologia , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/uso terapêutico , Camundongos , Camundongos Transgênicos , Invasividade Neoplásica/imunologia , Invasividade Neoplásica/prevenção & controle , Distribuição Aleatória , Receptor ErbB-2/genética , Receptor 7 Toll-Like/metabolismo , Receptor 7 Toll-Like/uso terapêutico , Falha de Tratamento , Regulação para Cima/imunologia
5.
J Immunol ; 182(9): 5217-24, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380767

RESUMO

Topical application of tumors with the TLR7 agonist imiquimod is an effective adjunct treatment for a range of primary dermatological cancers. However, for therapy to be effective against a broad range of solid tumor types, it must promote a strong systemic antitumor response that targets metastases in addition to primary tumor. We therefore investigated the potential of locally delivered imiquimod to stimulate an effective systemic antitumor response in a murine model of malignant mesothelioma (AB1-HA) with primary and distal tumors (dual tumor). Persistent delivery of imiquimod into primary tumor significantly retarded tumor growth in all treated mice compared with vehicle control. This local antitumor immune response required both CD8 T cells and NK cells, but not CD4 T cells, and was reliant on type I IFN induction. In vivo CTL studies and Ly6A/E staining of lymphocytes suggested that local imiquimod treatment had indeed induced a systemic, Ag-specific CD8 response. However, notably this response was not sufficient to retard the growth of an untreated distal tumor. Because local imiquimod treatment did not induce significant CD4 T cell responses, we investigated the efficacy of combining imiquimod with agonistic CD40 Ab (as a surrogate for CD4 T cell help). Combination of locally delivered imiquimod with systemic anti-CD40 immunotherapy not only significantly enhanced the local antitumor response, with 30% complete resolution, but it was also effective at significantly retarding growth of distal tumor. These results demonstrate that antitumor responses induced by locally delivered TLR7 agonists can be harnessed systemically for treating distal tumor.


Assuntos
Aminoquinolinas/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Antígenos CD40/imunologia , Glicoproteínas de Membrana/agonistas , Mesotelioma/imunologia , Mesotelioma/terapia , Receptor 7 Toll-Like/agonistas , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/uso terapêutico , Aminoquinolinas/uso terapêutico , Animais , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Quimioterapia Combinada , Feminino , Imiquimode , Interferon Tipo I/administração & dosagem , Interferon gama/administração & dosagem , Células Matadoras Naturais/imunologia , Ligantes , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/uso terapêutico , Mesotelioma/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Camundongos Transgênicos , Receptor 7 Toll-Like/metabolismo , Receptor 7 Toll-Like/uso terapêutico
6.
Oncologist ; 13(8): 859-75, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18701762

RESUMO

The importance of Toll-like receptors (TLRs) in stimulating innate and adaptive immunity is now well established. In view of this, TLR ligands have become interesting targets to use as stand-alone immunotherapeutics or vaccine adjuvants for cancer treatment. TLR7 and TLR8 were found to be closely related, sharing their intracellular endosomal location, as well as their ligands. In this review, we describe the agonists of TLR7 and TLR8 that are known so far, as well as their contribution to antitumor responses by affecting immune cells, tumor cells, and the tumor microenvironment. The major benefit of TLR7/8 agonists as immune response enhancers is their simultaneous stimulation of several cell types, resulting in a mix of activated immune cells, cytokines and chemokines at the tumor site. We discuss the studies that used TLR7/8 agonists as stand-alone immunotherapeutics or cancer vaccine adjuvants, as well as the potential of TLR7/8 ligands to enhance antitumor responses in passive immunotherapy approaches.


Assuntos
Vacinas Anticâncer/imunologia , Neoplasias/imunologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Humanos , Imunoterapia/métodos , Ligantes , Neoplasias/prevenção & controle , Receptor 7 Toll-Like/uso terapêutico , Receptor 8 Toll-Like/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA