Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Commun ; 14(1): 771, 2023 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-36774352

RESUMO

Glioblastoma, the most common and aggressive primary brain tumor type, is considered an immunologically "cold" tumor with sparse infiltration by adaptive immune cells. Immunosuppressive tumor-associated myeloid cells are drivers of tumor progression. Therefore, targeting and reprogramming intratumoral myeloid cells is an appealing therapeutic strategy. Here, we investigate a ß-cyclodextrin nanoparticle (CDNP) formulation encapsulating the Toll-like receptor 7 and 8 (TLR7/8) agonist R848 (CDNP-R848) to reprogram myeloid cells in the glioma microenvironment. We show that intravenous monotherapy with CDNP-R848 induces regression of established syngeneic experimental glioma, resulting in increased survival rates compared with unloaded CDNP controls. Mechanistically, CDNP-R848 treatment reshapes the immunosuppressive tumor microenvironment and orchestrates tumor clearing by pro-inflammatory tumor-associated myeloid cells, independently of T cells and NK cells. Using serial magnetic resonance imaging, we identify a radiomic signature in response to CDNP-R848 treatment and ultrasmall superparamagnetic iron oxide (USPIO) imaging reveals that immunosuppressive macrophage recruitment is reduced by CDNP-R848. In conclusion, CDNP-R848 induces tumor regression in experimental glioma by targeting blood-borne macrophages without requiring adaptive immunity.


Assuntos
Glioma , Nanopartículas , Receptor 7 Toll-Like , Receptor 8 Toll-Like , Humanos , Adjuvantes Imunológicos , Glioma/tratamento farmacológico , Macrófagos , Linfócitos T , Receptor 7 Toll-Like/agonistas , Microambiente Tumoral , Receptor 8 Toll-Like/agonistas
2.
Adv Healthc Mater ; 11(12): e2102781, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35285581

RESUMO

In situ anti-tumor vaccination is an attractive type of cancer immunotherapy which relies on the effectiveness of dendritic cells (DCs) to engulf tumor antigens, become activated, and present antigens to T cells in lymphoid tissue. Here, a multifunctional nanocomplex based on calcium crosslinked polyaspartic acid conjugated to either a toll-like receptor (TLR)7/8 agonist or a photosensitizer is reported. Intratumoral administration of the nanocomplex followed by laser irradiation induces cell killing and hence generation of a pool of tumor-associated antigens, with concomitant promotion of DCs maturation and expansion of T cells in tumor-draining lymph nodes. Suppression of tumor growth is observed both at the primary site and at the distal site, thereby hinting at successful induction of an adaptive anti-tumor response. This strategy holds promise for therapeutic application in a pre-operative and post-operative setting to leverage to mutanome of the patient's own tumor to mount immunological memory to clear residual tumor cells and metastasis.


Assuntos
Vacinas Anticâncer , Neoplasias , Receptor 7 Toll-Like , Receptor 8 Toll-Like , Adjuvantes Imunológicos/uso terapêutico , Animais , Antígenos de Neoplasias , Cálcio , Vacinas Anticâncer/administração & dosagem , Células Dendríticas , Sistemas de Liberação de Medicamentos , Imunidade , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanopartículas , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Vacinação
3.
Front Immunol ; 11: 580974, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33262759

RESUMO

Novel adjuvants, such as Toll-like receptors (TLRs) agonists, are needed for the development of new formulations able to circumvent limitations of current vaccines. Among TLRs, TLR7/8 agonists represent promising candidates, as they are well described to enhance antigen-specific antibody responses and skew immunity toward T helper (TH) 1 responses. We find here that the incorporation of the synthetic TLR7/8 ligand 3M-052 in a cationic DOEPC-based liposome formulation shifts immunity toward TH1 responses and elicits strong and long-lasting germinal center and follicular T helper cell responses in adult mice. This reflects the prolonged recruitment of innate cells toward the site of immunization and homing of activated antigen-loaded monocytes and monocyte-derived dendritic cells toward draining lymph nodes. We further show that this adjuvanticity is independent of type I IFN but NF-κB-dependent. Overall, our data identify TLR7/8 agonists incorporated in liposomes as promising and effective adjuvants to enhance TH1 and germinal center responses.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Glicoproteínas de Membrana/agonistas , Monócitos/imunologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Animais , Linfócitos B/imunologia , Células Dendríticas/imunologia , Composição de Medicamentos , Centro Germinativo/imunologia , Compostos Heterocíclicos com 3 Anéis/administração & dosagem , Imunidade Inata , Interferon Tipo I/imunologia , Ligantes , Lipossomos/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/deficiência , NF-kappa B/genética , NF-kappa B/imunologia , Fosfatidilcolinas/administração & dosagem , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais/imunologia , Ácidos Esteáricos/administração & dosagem , Células Th1/imunologia
4.
Bioorg Med Chem Lett ; 30(6): 126984, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-32001135

RESUMO

Toll-like receptors 7 and 8 (TLR7/8) agonists are potent immunostimulants that are attracting considerable interest as vaccine adjuvants. We recently reported the synthesis of a new series of 2-O-butyl-8-oxoadenines substituted at the 9-position with various linkers and N-heterocycles, and showed that TLR7/8 selectivity, potency and cytokine induction could be modulated by varying the alkyl linker length and the N-heterocyclic ring. In the present study, we further optimized the oxoadenine scaffold by investigating the effect of different substituents at the 2-position of the oxoadenine on TLR7/8 potency/selectivity, cytokine induction and DC maturation in human PBMCs. The results show that introducing a 1-(S)-methylbutoxy group at the 2-position of the oxoadenine significantly increased potency for TLR7/8 activity, cytokine induction and DC maturation.


Assuntos
Adenina/análogos & derivados , Adjuvantes Imunológicos/química , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Adenina/química , Adenina/imunologia , Adjuvantes Imunológicos/metabolismo , Citocinas/metabolismo , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Quinolinas/química , Relação Estrutura-Atividade
5.
J Control Release ; 306: 165-176, 2019 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-31173789

RESUMO

The toll-like receptor 7 and 8 (TLR7/8) agonist Resiquimod (R848) has been recognized as a promising immunostimulator for the treatment of cutaneous cancers in multiple clinical trials. However, systemic administration of R848 often results in strong immune-related toxicities while having limited therapeutic effects to the tumor. In the present study, a prodrug-based nanocarrier delivery system was developed that exhibited high therapeutic efficiency. R848 was conjugated to α-tocopherol to constitute an R848-Toco prodrug, followed by formulating with a tocopherol-modified hyaluronic acid (HA-Toco) as a polymeric nano-suspension. In vitro evaluation showed that the delivery system prolonged the release kinetics while maintaining TLR agonist activities. When administered subcutaneously, the nano-suspension formed a depot at the injection site, inducing localized immune responses without systemic expansion. This formulation also suppressed tumor growth and recruited immune cells to the tumor in a murine model of head and neck cancer. In a preclinical canine study of spontaneous mast cell tumors, the treatment led to a 67% response rate (three partial remissions and one complete remission).


Assuntos
Antineoplásicos/farmacologia , Imidazóis/administração & dosagem , Fatores Imunológicos/farmacologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Animais , Citocinas/metabolismo , Cães , Composição de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Liberação Controlada de Fármacos , Humanos , Imidazóis/química , Imidazóis/farmacologia , Camundongos , Camundongos Endogâmicos C3H , Coelhos , Suspensões
6.
Vaccine ; 35(6): 916-922, 2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-28089548

RESUMO

Diarrheal infectious diseases represent a major cause of global morbidity and mortality. There is an urgent need for vaccines against diarrheal pathogens, especially parasites. Modern subunit vaccines rely on combining a highly purified antigen with an adjuvant to increase their efficacy. In the present study, we evaluated the ability of a nanoliposome adjuvant system to trigger a strong mucosal immune response to the Entamoeba histolytica Gal/GalNAc lectin LecA antigen. CBA/J mice were immunized with alum, emulsion or liposome based formulations containing synthetic TLR agonists. A liposome formulation containing TLR4 and TLR7/8 agonists was selected based on its ability to generate intestinal IgA, plasma IgG2a/IgG1, IFN-γ and IL-17A. Immunization with a mucosal prime followed by a parenteral boost generated a high mucosal IgA response that inhibited adherence of parasites to mammalian cells. Inclusion of the immune potentiator all-trans retinoic acid in the regimen further improved the mucosal IgA response. Immunization protected from infection with up to 55% efficacy. Our results show that a nanoliposome delivery system containing TLR agonists is a promising prospect for the development of vaccines against enteric pathogens, especially when a multifaceted immune response is desired.


Assuntos
Anticorpos Antiprotozoários/biossíntese , Entamoeba histolytica/efeitos dos fármacos , Entamebíase/prevenção & controle , Imunidade nas Mucosas/efeitos dos fármacos , Lipossomos/imunologia , Vacinas Protozoárias/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/química , Compostos de Alúmen/administração & dosagem , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Entamoeba histolytica/crescimento & desenvolvimento , Entamoeba histolytica/imunologia , Entamebíase/imunologia , Entamebíase/parasitologia , Imunização , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-17/biossíntese , Interleucina-17/imunologia , Lectinas/química , Lectinas/imunologia , Lipopolissacarídeos/administração & dosagem , Lipossomos/administração & dosagem , Lipossomos/química , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos CBA , Oligodesoxirribonucleotídeos/administração & dosagem , Polissorbatos/administração & dosagem , Vacinas Protozoárias/química , Vacinas Protozoárias/imunologia , RNA/administração & dosagem , Esqualeno/administração & dosagem , Receptor 3 Toll-Like/agonistas , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/genética , Receptor 8 Toll-Like/imunologia , Tretinoína/administração & dosagem , Vacinas de Subunidades Antigênicas
7.
Int Immunopharmacol ; 40: 410-418, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27697724

RESUMO

ß,ß-dimethylacryloyl alkannin (DMA) is a key component of Lithospermum and possesses good efficacy for treating psoriasis. DMA inhibits activated dendritic cells (DCs), but the mechanism is unknown. Therefore, this study aimed to explore the modulation of the TLR7/8 pathway by DMA in psoriasis-activated DCs. Models of psoriasis-like skin lesions were established using BALB/c mice; 8 mice were treated with DMA (2.5mg/kg). Bone marrow cells were isolated and induced into DCs using R848, a TLR7/8 agonist. Splenic CD11c+ cells were detected by flow cytometry. Skin CD11c+ cells were detected by immunofluorescence. TLR7, TLR8, MYD88, and IRAKM proteins were detected by Western blot. The effects of DMA on surface molecules of DCs were observed by flow cytometry. mRNA expression of inflammatory factors was detected by qRT-PCR. Secreted cytokines were detected by cytometric bead array. Compared with the model group, psoriasis-like skin lesions were alleviated by DMA, the splenic CD11c+ cells were significantly decreased (P<0.01), and CD11c+ cell numbers in skin lesions were decreased (P<0.01). Expression levels of TLR7, MYD88, and IRAKM were significantly decreased (P<0.05). R848-stimulated DCs showed increased expression of I-A/I-E, CD80, and CD86 (P<0.01), increased IL-23 and IL-1ß mRNA and secretion (P<0.05), and increased TLR7, TLR8, MYD88, and IRAKM expression (P<0.01); DMA inhibited all of these effects of the TLR7/8 pathway activation by R848 (P<0.05). In conclusion, DMA could inhibit psoriasis-activated DCs via the TLR7/8 pathway.


Assuntos
Células Dendríticas/efeitos dos fármacos , Lithospermum , Glicoproteínas de Membrana/metabolismo , Naftoquinonas/uso terapêutico , Psoríase/tratamento farmacológico , Pele/efeitos dos fármacos , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Dendríticas/imunologia , Modelos Animais de Doenças , Humanos , Imidazóis/farmacologia , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-23/genética , Interleucina-23/metabolismo , Masculino , Glicoproteínas de Membrana/agonistas , Camundongos , Camundongos Endogâmicos BALB C , Fator 88 de Diferenciação Mieloide/metabolismo , Naftoquinonas/química , Pele/imunologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas
8.
J Med Chem ; 58(19): 7833-49, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26351878

RESUMO

Human Toll-like receptor 8 (hTLR8) is expressed in myeloid dendritic cells, monocytes, and monocyte-derived dendritic cells. Engagement by TLR8 agonists evokes a distinct cytokine profile which favors the development of type 1 helper T cells. Crystal structures of the ectodomain of hTLR8 cocrystallized with two regioisomers of a dual TLR7/8-agonistic N1-substituted imidazoquinolines showed subtle differences in their interactions in the binding site of hTLR8. We hypothesized that the potency of a previously reported best-in-class pure TLR8 agonist, 3-pentylquinoline-2-amine, could be further enhanced by "designing in" functional groups that would mimic key intermolecular interactions that we had observed in the crystal structures. We performed a focused exploration of decorating the quinoline core with alkylamino groups at all possible positions. These studies have led to the identification of a novel TLR8 agonist that was ∼ 20-fold more potent than the parent compound and displays prominent adjuvantic activity in a rabbit model of immunization.


Assuntos
Adjuvantes Imunológicos/farmacologia , Relação Estrutura-Atividade , Receptor 8 Toll-Like/agonistas , Imunidade Adaptativa/efeitos dos fármacos , Adjuvantes Imunológicos/química , Animais , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Humanos , Conformação Proteica , Quinolinas/química , Coelhos , Receptor 8 Toll-Like/química , Receptor 8 Toll-Like/genética
9.
PLoS Pathog ; 10(6): e1004210, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24967632

RESUMO

The ability of innate immune cells to sense and respond to impending danger varies by anatomical location. The liver is considered tolerogenic but is still capable of mounting a successful immune response to clear various infections. To understand whether hepatic immune cells tune their response to different infectious challenges, we probed mononuclear cells purified from human healthy and diseased livers with distinct pathogen-associated molecules. We discovered that only the TLR8 agonist ssRNA40 selectively activated liver-resident innate immune cells to produce substantial quantities of IFN-γ. We identified CD161(Bright) mucosal-associated invariant T (MAIT) and CD56(Bright) NK cells as the responding liver-resident innate immune cells. Their activation was not directly induced by the TLR8 agonist but was dependent on IL-12 and IL-18 production by ssRNA40-activated intrahepatic monocytes. Importantly, the ssRNA40-induced cytokine-dependent activation of MAIT cells mirrored responses induced by bacteria, i.e., generating a selective production of high levels of IFN-γ, without the concomitant production of TNF-α or IL-17A. The intrahepatic IFN-γ production could be detected not only in healthy livers, but also in HBV- or HCV-infected livers. In conclusion, the human liver harbors a network of immune cells able to modulate their immunological responses to different pathogen-associated molecules. Their ability to generate a strong production of IFN-γ upon stimulation with TLR8 agonist opens new therapeutic opportunities for the treatment of diverse liver pathologies.


Assuntos
Adjuvantes Imunológicos/farmacologia , Imunidade Inata/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Fígado/efeitos dos fármacos , Oligorribonucleotídeos/farmacologia , Receptor 8 Toll-Like/agonistas , Regulação para Cima/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Enterococcus faecalis/imunologia , Enterococcus faecalis/metabolismo , Enterococcus faecalis/patogenicidade , Escherichia coli/imunologia , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Hepacivirus/imunologia , Hepacivirus/patogenicidade , Hepatite B/imunologia , Hepatite B/metabolismo , Hepatite B/patologia , Hepatite B/virologia , Vírus da Hepatite B/imunologia , Vírus da Hepatite B/patogenicidade , Hepatite C/imunologia , Hepatite C/metabolismo , Hepatite C/patologia , Hepatite C/virologia , Humanos , Testes de Liberação de Interferon-gama , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Fígado/imunologia , Fígado/microbiologia , Fígado/patologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/metabolismo , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade , Riboflavina/biossíntese , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Receptor 8 Toll-Like/metabolismo
10.
Inflamm Allergy Drug Targets ; 13(2): 144-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24773332

RESUMO

BACKGROUND/PURPOSE(S): There is a growing interest in the targeting of Toll-like receptors (TLRs) for the treatment of allergic diseases. TLRs7/8 ligands are future candidates of therapeutic value in allergic rhinitis (AR). This study focus on TLRs7/8 ligand; resiquimod (R848) as an adjuvant to immunotherapy (IT) in AR patient. METHODS: Peripheral blood mononuclear cells (PBMCs) were obtained from atopic donors and non atopic donors. PBMCs were cultured in the absence and presence of date palm pollen allergen (Phoenix dactylifera; Pho d) and/or R848. Interleukin-4 (IL-4), IL-10, IL-13 and interferon gamma (IFN-γ) were measured in the culture supernatants. RESULTS: R848 was able to significantly increase the anti-inflammatory response in atopic donors more than non atopic donors. Nevertheless, the combination of both; R848 and Pho d provides inferior stimulus as compared to R848 alone in both atopic and non atopic donors. CONCLUSION: Invitro treatment of PBMCs with R484 hijacks the pro inflammatory immune process triggered by TLRs7/8 to mediate anti-inflammatory response. This may provide a conception about the activity and efficacy of TLRs7/8 ligands in AR and open the gate for them to be applied in clinically in humans.


Assuntos
Antialérgicos/farmacologia , Anti-Inflamatórios/farmacologia , Imidazóis/farmacologia , Rinite Alérgica Sazonal/tratamento farmacológico , Rinite Alérgica/tratamento farmacológico , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Adulto , Alérgenos , Células Cultivadas , Citocinas/metabolismo , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Pessoa de Meia-Idade , Phoeniceae/imunologia , Proteínas de Plantas/imunologia , Pólen/imunologia , Rinite Alérgica/imunologia , Rinite Alérgica/metabolismo , Rinite Alérgica Sazonal/imunologia , Rinite Alérgica Sazonal/metabolismo , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/metabolismo
11.
PLoS One ; 8(8): e72112, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977222

RESUMO

Using a yeast-based assay, a previously unsuspected antiprion activity was found for imiquimod (IQ), a potent Toll-like receptor 7 (TLR7) agonist already used for clinical applications. The antiprion activity of IQ was first detected against yeast prions [PSI (+) ] and [URE3], and then against mammalian prion both ex vivo in a cell-based assay and in vivo in a transgenic mouse model for prion diseases. In order to facilitate structure-activity relationship studies, we conducted a new synthetic pathway which provides a more efficient means of producing new IQ chemical derivatives, the activity of which was tested against both yeast and mammalian prions. The comparable antiprion activity of IQ and its chemical derivatives in the above life forms further emphasizes the conservation of prion controlling mechanisms throughout evolution. Interestingly, this study also demonstrated that the antiprion activity of IQ and IQ-derived compounds is independent from their ability to stimulate TLRs. Furthermore, we found that IQ and its active chemical derivatives inhibit the protein folding activity of the ribosome (PFAR) in vitro.


Assuntos
Aminoquinolinas/farmacologia , Glutationa Peroxidase/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , Doenças Priônicas/tratamento farmacológico , Príons/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Aminoquinolinas/síntese química , Animais , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Guanosina/análogos & derivados , Guanosina/farmacologia , Humanos , Imidazóis/farmacologia , Imiquimode , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/metabolismo , Camundongos , Proteínas PrPSc/metabolismo , Doenças Priônicas/metabolismo , Dobramento de Proteína , Saccharomyces cerevisiae/efeitos dos fármacos , Relação Estrutura-Atividade , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/metabolismo
12.
Med Sci (Paris) ; 28(1): 96-102, 2012 Jan.
Artigo em Francês | MEDLINE | ID: mdl-22289837

RESUMO

Toll-like receptors (TLR) sense a variety of microbial products and play an important role in the mounting of innate and adaptive immune responses. TLR1 to TLR9 are common in mice and humans and recognize similar ligands in both species, with the exception of TLR8. Human TLR7 and TLR8 and mouse TLR7 detect viral single-stranded RNA and imidazoquinoline compounds, while mouse TLR8 not. Based on this discrepancy, for long time it was believed that mouse TLR8 is not functional and as a consequence the contribution of TLR8 to innate immunity remained poorly understood. Our recent studies revealed an important role for TLR8 in the regulation of TLR7-mediated autoimmunity in the mouse. This review illustrates our current understanding regarding the function of TLR8 and its potential for future clinical use for the treatment and/or prevention of various pathological conditions.


Assuntos
Glicoproteínas de Membrana/imunologia , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/fisiologia , Imunidade Adaptativa , Aminoquinolinas/farmacologia , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/imunologia , Ensaios Clínicos Fase I como Assunto , Citocinas/fisiologia , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica , Imiquimode , Imunidade Inata , Ligantes , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Modelos Imunológicos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , RNA Viral/imunologia , Transdução de Sinais/fisiologia , Especificidade da Espécie , Receptor 3 Toll-Like/fisiologia , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/fisiologia , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/genética , Viroses/imunologia
13.
Antivir Ther ; 16(5): 751-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21817197

RESUMO

BACKGROUND: Upper respiratory tract infection is a frequent cause of morbidity worldwide. Although the course of infection is usually mild, it is responsible for enormous social and economic costs. Immunostimulation with bacterial extracts consisting of ribosomal RNA and proteoglycans, such as Ribomunyl, was introduced into the clinic in the 1980s as a new treatment concept, but did not achieve widespread application. Ribomunyl has been proposed to activate innate immunity, but the contribution of its RNA content as well as its antiviral potential has not been studied. METHODS: Peripheral blood mononuclear cells from healthy donors and immune cells from adenoids were incubated with Ribomunyl either by itself or formulated in a complex with cationic polypeptides such as poly-l-arginine or protamine, and induction of cytokines was quantified by ELISA. RESULTS: Ribomunyl in complex with either poly-l-arginine or protamine, but not on its own, was able to strongly induce interferon-α (P<0.01) and interleukin-12 (P<0.01) in peripheral blood mononuclear cells, whereas induced tumour necrosis factor-α and interleukin-6 levels were independent of the formulation. Comparable results were obtained in immune cells from adenoids, suggesting efficacy also in virus-affected tissue. Cell sorting, RNase digests and selective receptor expression show that the RNA in Ribomunyl acts as an agonist of Toll-like receptor (TLR)7 and TLR8. CONCLUSIONS: Ribomunyl is, in principle, able to potently induce antiviral interferon-α and interleukin-12 via TLR7 and TLR8, respectively, but only when formulated in a complex with cationic polypeptides.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos de Bactérias/farmacologia , Antivirais/farmacologia , Poliaminas/química , Tonsila Faríngea/efeitos dos fármacos , Adjuvantes Imunológicos/metabolismo , Adjuvantes Imunológicos/uso terapêutico , Antígenos de Bactérias/metabolismo , Antígenos de Bactérias/uso terapêutico , Antivirais/metabolismo , Antivirais/uso terapêutico , Arginina/química , Arginina/metabolismo , Citocinas/análise , Citocinas/efeitos dos fármacos , Relação Dose-Resposta a Droga , Composição de Medicamentos , Células HEK293 , Humanos , Imunização/métodos , Leucócitos Mononucleares/efeitos dos fármacos , Terapia de Alvo Molecular , Poliaminas/metabolismo , Polieletrólitos , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/imunologia , Infecções Respiratórias/patologia , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/imunologia , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/imunologia , Receptor 8 Toll-Like/metabolismo
14.
Vaccine ; 27(23): 3045-52, 2009 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-19428918

RESUMO

Agonists for TLR7, TLR8, and TLR9 have been shown to enhance vaccine immunogenicity. We evaluated the impact of TLR activation on RSV disease in a murine model by administering TLR7/8 and TLR9 agonists during FI-RSV immunization or RSV infection. CpG administered during immunization reduced disease following challenge as evidenced by decreased lung pathology, illness, and cytokines. In marked contrast, TLR7/8 agonist had little impact. To evaluate potential therapeutic use, TLR agonists were administered during primary infection. Although type 2 cytokine responses decreased and type 1 cytokines and MIP-1-alpha/beta increased, both TLR7/8 and TLR9 agonists increased clinical symptoms and pulmonary inflammation when administered during primary infection. Thus, TLR9-induced signaling during FI-RSV immunization reduced vaccine-enhanced disease whereas immunostimulatory properties of TLR agonists enhanced disease severity when used during RSV infection. Immunomodulation elicited by TLR9 agonist confirms the adjuvant potential of TLR agonists during RSV immunization. However, in contrast to work done with HIV-1 vaccines, the inability of TLR7/8 agonist to boost type 1 vaccine-induced RSV immunity demonstrates pathogen-TLR specificity. These data reveal that the timing of administration of immunomodulatory agents is critical. Furthermore, these data underscore that amplification of anti-viral immune responses may result in immunopathology rather than immune-mediated protection.


Assuntos
Glicoproteínas de Membrana/agonistas , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/uso terapêutico , Vírus Sinciciais Respiratórios/patogenicidade , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Receptor Toll-Like 9/agonistas , Adjuvantes Imunológicos/uso terapêutico , Animais , Formação de Anticorpos , Linhagem Celular , Glicina/análogos & derivados , Glicina/farmacologia , Humanos , Imidazóis/farmacologia , Imunidade Celular , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia , Receptor Toll-Like 9/imunologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA