Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomaterials ; 295: 122030, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36758340

RESUMO

Since the progression of osteoarthritis (OA) is closely associated with synovitis and cartilage destruction, the inhibition of inflammatory responses in synovial macrophages and reactive oxygen species (ROS) induced apoptosis in chondrocytes is crucial for OA amelioration. However, most of the current anti-inflammatory and antioxidant drugs are small molecules apt to be eliminated in vivo. Herein, mesoporous polydopamine nanoparticles (DAMM NPs) doped with arginine and manganese (Mn) ions were prepared to load dexamethasone (DEX) for OA intervention. A series of in vitro studies showed that the sustained release of DEX from DAMM NPs suppressed synovial inflammation and simultaneously inhibited toll-like receptor 3 (TLR-3) production in chondrocytes, contributing to prevention of chondrocyte apoptosis through the inflammatory factor-dependent TLR-3/NF-κB signaling pathway via modulation of macrophage-chondrocyte crosstalk. In addition, DAMM NPs exerted a predominant role in removal of ROS generated in chondrocytes. Therefore, the DEX-loaded DAMM NPs significantly attenuated OA development in mice model. Importantly, the T1-T2 magnetic contrast capabilities of DAMM NPs allowed an MRI-trackable delivery, manifesting a distinct feature widely regarded to boost the potential of nanomedicines for clinical applications. Together, our developed antioxidant-enhanced DAMM NPs with MRI-visible signals may serve as a novel multifunctional nanocarriers for prevention of OA progression.


Assuntos
Nanopartículas , Osteoartrite , Camundongos , Animais , Antioxidantes/uso terapêutico , Antioxidantes/metabolismo , Receptor 3 Toll-Like/metabolismo , Receptor 3 Toll-Like/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Osteoartrite/diagnóstico por imagem , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , NF-kappa B/metabolismo , Condrócitos/metabolismo
2.
Environ Sci Pollut Res Int ; 30(10): 28118-28132, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36394807

RESUMO

Rhinovirus infection frequently causes COPD and asthma exacerbations. Impaired anti-viral signaling and reduced viral clearance have both been seen in sick bronchial epithelium, potentially increasing exacerbations. Polyinosinic:polycytidylic acid (Poly(I:C)), a Toll-like receptor-3 (TLR3) ligand, has been shown to cause a viral exacerbation of severe asthma by detecting double-stranded RNA (dsRNA). The purpose of this work was to determine the effect of a TLR3/dsRNA complex inhibitor-Calbiochem drug in the prevention of Poly(I:C)-induced airway inflammation following TLR3 activation and to uncover a potential pathway for the cure of asthma through TLR3 inhibition. Mice were sensitized with Poly(I:C) as an asthma model before being challenged by PBS and ovalbumin (OVA) chemicals. The mice were administered a TLR3/dsRNA complex inhibitor. Throughout the trial, the mice's body weight was measured after each dosage. Biochemical methods are used to analyze the protein as well as enzyme composition in airway tissues. BALF specimens are stained using Giemsa to identify inflammatory cells and lung histopathology to determine morphological abnormalities in lung tissues. By using the ELISA approach, cytokine levels such as TNF-α, IL-13, IL-6, IL-5, and IgE antibody expression in lung tissue and blood serum were assessed. TLR3/dsRNA complex inhibitor drug significantly lowered the number of cells in BALF and also on Giemsa staining slides. It also downregulated the level of TNF-α and IL-6 in contrast to OVA and Poly(I:C) administered in animals. A TLR3/dsRNA complex inhibitor decreased the fraction of oxidative stress markers (MDA, GSH, GPx, and CAT) in lung tissues while keeping the mice's body weight constant during the treatment period. By decreasing alveoli, bronchial narrowing, smooth muscle hypertrophy, and granulocyte levels, the TLR3/dsRNA complex blocker significantly reduced the histopathological damage caused by OVA and Poly(I:C) compounds. In an animal model utilizing ovalbumin, TLR3/dsRNA complex inhibitors similarly reduced the bronchial damage produced by Poly(I:C). A novel TLR3/dsRNA complex inhibitor is expected to be employed in clinical studies since it suppresses airway inflammation without inducing antiviral approach resistance.


Assuntos
Asma , Fator de Necrose Tumoral alfa , Camundongos , Animais , Fator de Necrose Tumoral alfa/metabolismo , Ovalbumina , RNA de Cadeia Dupla/metabolismo , RNA de Cadeia Dupla/uso terapêutico , Receptor 3 Toll-Like/metabolismo , Receptor 3 Toll-Like/uso terapêutico , Interleucina-6/metabolismo , Modelos Animais de Doenças , Asma/induzido quimicamente , Pulmão/patologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Poli I-C/farmacologia , Poli I-C/uso terapêutico , Líquido da Lavagem Broncoalveolar
3.
Pharmacol Res Perspect ; 10(6): e01038, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36507603

RESUMO

The innate immune system has an emerging role as a mediator of neuro-immune communication and a therapeutic target for itch. Toll-like receptor 3 (TLR3) plays an important role in itch, as shown in TLR3 knock-out mice. In this study, to evaluate effects of TLR3 inhibitors on histamine-independent itch, we used two kinds of isothiocyanate (ITC). Both phenethyl isothiocyanate (PEITC) and sulforaphane (SFN) inhibited Poly I:C (PIC)-induced signaling in the RAW264.7 cell line. We then investigated the anti-pruritic effect of these compounds on PIC- and chloroquine (CQ)-induced scratching behavior. PEITC and SFN both suppressed PIC-evoked scratching behavior in mice, and PEITC also inhibited CQ-induced acute itch. Finally, we examined the oxazolone-induced chronic itch model in mice. Surprisingly, oral dosing of both compounds suppressed scratching behaviors that were observed in mice. Our findings demonstrate that TLR3 is a critical mediator in acute and chronic itch transduction in mice and may be a promising therapeutic target for pruritus in human skin disorders. It is noteworthy that SFN has potential for use as an antipruritic as it is a phytochemical that is used as a supplement.


Assuntos
Antipruriginosos , Receptor 3 Toll-Like , Animais , Humanos , Camundongos , Antipruriginosos/farmacologia , Antipruriginosos/uso terapêutico , Cloroquina , Camundongos Knockout , Prurido/induzido quimicamente , Prurido/tratamento farmacológico , Prurido/metabolismo , Pele/metabolismo , Receptor 3 Toll-Like/uso terapêutico
4.
Biomaterials ; 290: 121831, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36240687

RESUMO

Immunotherapies including immune checkpoint blockade (ICB) have become integral to treatments for immunogenic tumors by reinvigorating host immune functions to attack tumor cells. However, the clinical applications of ICB are limited by relatively low response rates as well as inherent and acquired resistance in many types of cancer. A potential solution is to selectively deliver immune modulators to tumors to activate the tumor microenvironment (TME) and enhance the therapeutic effect of ICB. Here we report the design of polyinosinic: polycytidylic acid (pIC) nanoscale coordination polymer (NCP), pIC@NCP, and its ability to activate tumor-specific immune responses and synergize with ICB for potent antitumor effects. With prolonged blood circulation, facile cell uptake, and triggered release of pIC in acidic TMEs, pIC@NCP shows robust tumor growth inhibition via activation of the endosomal toll-like receptor 3 signaling pathway. The synergistic combination of pIC@NCP and ICB further controls tumor growth in syngeneic mouse models of colorectal cancer and a spontaneous mouse model of prostate cancer. This study highlights the potential of NCP delivery of nucleic acid therapeutics for immune activation and cancer therapy.


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Camundongos , Masculino , Animais , Receptor 3 Toll-Like/uso terapêutico , Polímeros/farmacologia , Imunoterapia , Neoplasias/tratamento farmacológico , Microambiente Tumoral , Fatores Imunológicos/farmacologia
5.
J Am Heart Assoc ; 11(20): e026076, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36216458

RESUMO

Background Spinal cord ischemia (SCI) remains a devastating complication after aortic dissection or repair. A primary hypoxic damage is followed by a secondary damage resulting in further cellular loss via apoptosis. Affected patients have a poor prognosis and limited therapeutic options. Shock wave therapy (SWT) improves functional outcome, neuronal degeneration and survival in murine spinal cord injury. In this first-in-human study we treated 5 patients with spinal cord ischemia with SWT aiming to prove safety and feasibility. Methods and Results Human neurons were subjected to ischemic injury with subsequent SWT. Reactive oxygen species and cellular apoptosis were quantified using flow cytometry. Signaling of the antioxidative transcription factor NRF2 (nuclear factor erythroid 2-related factor 2) and immune receptor Toll-like receptor 3 (TLR3) were analyzed. To assess whether SWT act via a conserved mechanism, transgenic tlr3-/- zebrafish created via CRISPR/Cas9 were subjected to spinal cord injury. To translate our findings into a clinical setting, 5 patients with SCI underwent SWT. Baseline analysis and follow-up (6 months) included assessment of American Spinal Cord Injury Association (ASIA) impairment scale, evaluation of Spinal Cord Independence Measure score and World Health Organization Quality of Life questionnaire. SWT reduced the number of reactive oxygen species positive cells and apoptosis upon ischemia via induction of the antioxidative factor nuclear factor erythroid 2-related factor 2. Inhibition or deletion of tlr3 impaired axonal growth after spinal cord lesion in zebrafish, whereas tlr3 stimulation enhanced spinal regeneration. In a first-in-human study, we treated 5 patients with SCI using SWT (mean age, 65.3 years). Four patients presented with acute aortic dissection (80%), 2 of them exhibited preoperative neurological symptoms (40%). Impairment was ASIA A in 1 patient (20%), ASIA B in 3 patients (60%), and ASIA D in 1 patient (20%) at baseline. At follow-up, 2 patients were graded as ASIA A (40%) and 3 patients as ASIA B (60%). Spinal cord independence measure score showed significant improvement. Examination of World Health Organization Quality of Life questionnaires revealed increased scores at follow-up. Conclusions SWT reduces oxidative damage upon SCI via immune receptor TLR3. The first-in-human application proved safety and feasibility in patients with SCI. SWT could therefore become a powerful regenerative treatment option for this devastating injury.


Assuntos
Dissecção Aórtica , Tratamento por Ondas de Choque Extracorpóreas , Traumatismos da Medula Espinal , Isquemia do Cordão Espinal , Humanos , Camundongos , Animais , Idoso , Receptor 3 Toll-Like/metabolismo , Receptor 3 Toll-Like/uso terapêutico , Fator 2 Relacionado a NF-E2 , Peixe-Zebra , Estudos de Viabilidade , Espécies Reativas de Oxigênio , Qualidade de Vida , Isquemia do Cordão Espinal/etiologia , Isquemia do Cordão Espinal/prevenção & controle , Isquemia do Cordão Espinal/patologia , Traumatismos da Medula Espinal/terapia , Traumatismos da Medula Espinal/patologia , Medula Espinal/metabolismo , Estresse Oxidativo , Isquemia , Dissecção Aórtica/patologia
6.
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
7.
Clin Cancer Res ; 28(10): 2038-2049, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35046055

RESUMO

PURPOSE: Increased prevalence of cytotoxic T lymphocytes (CTL) in the tumor microenvironment (TME) predicts positive outcomes in patients with epithelial ovarian cancer (EOC), whereas the regulatory T cells (Treg) predict poor outcomes. Guided by the synergistic activity of TLR3 ligands, IFNα, and COX-2 blockers in selectively enhancing CTL-attractants but suppressing Treg-attractants, we tested a novel intraperitoneal chemoimmunotherapy combination (CITC), to assess its tolerability and TME-modulatory impact in patients with recurrent EOC. PATIENTS AND METHODS: Twelve patients were enrolled in phase I portion of the trial NCT02432378, and treated with intraperitoneal cisplatin, intraperitoneal rintatolimod (dsRNA, TLR3 ligand), and oral celecoxib (COX-2 blocker). Patients in cohorts 2, 3, and 4 also received intraperitoneal IFNα at 2, 6, and 18 million units (MU), respectively. Primary objectives were to evaluate safety, identify phase 2 recommended dose (P2RD), and characterize changes in the immune TME. Peritoneal resident cells and intraperitoneal wash fluid were profiled via NanoString and Meso Scale Discovery (MSD) multiplex assay, respectively. RESULTS: The P2RD of IFNα was 6 MU. Median progression-free survival and overall survival were 8.4 and 30 months, respectively. Longitudinal sampling of the peritoneal cavity via intraperitoneal washes demonstrated local upregulation of IFN-stimulated genes (ISG), including CTL-attracting chemokines (CXCL-9, -10, -11), MHC I/II, perforin, and granzymes. These changes were present 2 days after chemokine modulation and subsided within 1 week. CONCLUSIONS: The chemokine-modulating intraperitoneal-CITC is safe, tolerable, and associated with ISG changes that favor CTL chemoattraction and function. This combination (plus DC vaccine) will be tested in a phase II trial. See related commentary by Aranda et al., p. 1993.


Assuntos
Neoplasias Ovarianas , Receptor 3 Toll-Like , Carcinoma Epitelial do Ovário/tratamento farmacológico , Quimiocinas , Ciclo-Oxigenase 2 , Feminino , Humanos , Imunoterapia , Ligantes , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Receptores CXCR3 , Receptor 3 Toll-Like/uso terapêutico , Microambiente Tumoral
8.
J Biomol Struct Dyn ; 40(8): 3753-3768, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-33246391

RESUMO

Hepatitis C virus (HCV), which infected 71 million worldwide and about 5%-6% are from Pakistan, is an ssRNA virus, responsible for end-stage liver disease. To date, no effective therapy is available to cure this disease. Hence, it is important to study the most prevalent genotypes infecting human population and design novel vaccine or small molecule inhibitors to control the infections associated with HCV. Therefore, in this study clinical samples (n = 35; HCV-3a) from HCV patients were subjected to Sanger sequencing method. The sequencing of the core gene, which is generally considered as conserved, involved in the detection, quantitation and genotyping of HCV was performed. Multiple mutations, that is, R46C, R70Q, L91C, G60E, N/S105A, P108A, N110I, S116V, G90S, A77G and G145R that could be linked with response to antiviral therapies were detected. Phylogenetic analysis suggests emerging viral isolates are circulating in Pakistan. Using ab initio modelling technique, we predicted the 3D structure of core protein and subjected to molecular dynamics simulation to extract the most stable conformation of the structure for further analysis. Immunoinformatic approaches were used to propose a multi-epitopes vaccine against HCV by using core protein. The vaccine constructs consist of nine CTL and three HTL epitopes joined by different linkers were docked against the two reported Toll-like receptors (TLR-3 and TLR-8). Docking of vaccine construct with TLR-3 and TLR-8 shows proper binding and in silico expression of the vaccine resulted in a CAI value of 0.93. These analyses suggest that specific immune responses may be produced by the proposed vaccine.Communicated by Ramaswamy H. Sarma.


Assuntos
Hepatite C , Vacinas , Substituição de Aminoácidos , Epitopos de Linfócito T , Fluprednisolona/análogos & derivados , Hepacivirus/genética , Hepatite C/prevenção & controle , Humanos , Paquistão , Filogenia , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/uso terapêutico , Receptor 8 Toll-Like/genética , Vacinas/uso terapêutico
9.
Mol Ther ; 30(3): 1163-1170, 2022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-34563676

RESUMO

The adenosine deaminase inhibitor 2'-deoxycoformycin (pentostatin, Nipent) has been used since 1982 to treat leukemia and lymphoma, but its mode of action is still unknown. Pentostatin was reported to decrease methylation of cellular RNA. We discovered that RNA extracted from pentostatin-treated cells or mice has enhanced immunostimulating capacities. Accordingly, we demonstrated in mice that the anticancer activity of pentostatin required Toll-like receptor 3, the type I interferon receptor, and T cells. Upon systemic administration of pentostatin, type I interferon is produced locally in tumors, resulting in immune cell infiltration. We combined pentostatin with immune checkpoint inhibitors and observed synergistic anti-cancer activities. Our work identifies pentostatin as a new class of an anticancer immunostimulating drug that activates innate immunity within tumor tissues and synergizes with systemic T cell therapies.


Assuntos
Antineoplásicos , Linfoma , Animais , Antineoplásicos/uso terapêutico , Camundongos , Pentostatina/farmacologia , Pentostatina/uso terapêutico , RNA , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/uso terapêutico
10.
J Immunol ; 187(10): 5320-7, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21998457

RESUMO

Cytoplasmic and endosomal RNA sensors recognize RNA virus infection and signals to protect host cells by inducing type I IFN. The cytoplasmic RNA sensors, retinoic acid inducible gene I/melanoma differentiation-associated gene 5, actually play pivotal roles in sensing virus replication. IFN-ß promoter stimulator-1 (IPS-1) is their common adaptor for IFN-inducing signaling. Toll/IL-1R homology domain-containing adaptor molecule 1 (TICAM-1), also known as TRIF, is the adaptor for TLR3 that recognizes viral dsRNA in the early endosome in dendritic cells and macrophages. Poliovirus (PV) belongs to the Picornaviridae, and melanoma differentiation-associated gene 5 reportedly detects replication of picornaviruses, leading to the induction of type I IFN. In this study, we present evidence that the TLR3/TICAM-1 pathway governs IFN induction and host protection against PV infection. Using human PVR transgenic (PVRtg) mice, as well as IPS-1(-/-) and TICAM-1(-/-) mice, we found that TICAM-1 is essential for antiviral responses that suppress PV infection. TICAM-1(-/-) mice in the PVRtg background became markedly susceptible to PV, and their survival rates were decreased compared with wild-type or IPS-1(-/-) mice. Similarly, serum and organ IFN levels were markedly reduced in TICAM-1(-/-)/PVRtg mice, particularly in the spleen and spinal cord. The sources of type I IFN were CD8α(+)/CD11c(+) splenic dendritic cells and macrophages, where the TICAM-1 pathway was more crucial for PV-derived IFN induction than was the IPS-1 pathway in ex vivo and in vitro analyses. These data indicate that the TLR3/TICAM-1 pathway functions are dominant in host protection and innate immune responses against PV infection.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Imunidade Inata , Poliomielite/imunologia , Receptor 3 Toll-Like/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/uso terapêutico , Animais , Células Cultivadas , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Inata/genética , Interferon Tipo I/biossíntese , Interferon Tipo I/uso terapêutico , Macrófagos/imunologia , Macrófagos/patologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células NIH 3T3 , Poliomielite/genética , Poliomielite/mortalidade , Poliovirus/imunologia , Receptores Virais/genética , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/uso terapêutico , Células Vero
11.
J Immunol ; 186(4): 2422-9, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21242525

RESUMO

Maturation of dendritic cells (DC) to competent APC is essential for the generation of acquired immunity and is a major function of adjuvants. dsRNA, a molecular signature of viral infection, drives DC maturation by activating TLR3, but the size of dsRNA required to activate DC and the expression patterns of TLR3 protein in DC subsets have not been established. In this article, we show that cross-priming CD8α(+) and CD103(+) DC subsets express much greater levels of TLR3 than other DC. In resting DC, TLR3 is located in early endosomes and other intracellular compartments but migrates to LAMP1(+) endosomes on stimulation with a TLR3 ligand. Using homogeneous dsRNA oligonucleotides (ONs) ranging in length from 25 to 540 bp, we observed that a minimum length of ∼90 bp was sufficient to induce CD86, IL-12p40, IFN-ß, TNF-α, and IL-6 expression, and to mature DC into APC that cross-presented exogenous Ags to CD8(+) T cells. TLR3 was essential for activation of DC by dsRNA ONs, and the potency of activation increased with dsRNA length and varied between DC subsets. In vivo, dsRNA ONs, in a size-dependent manner, served as adjuvants for the generation of Ag-specific CTL and for inducing protection against lethal challenge with influenza virus when given with influenza nucleoprotein as an immunogen. These results provide the basis for the development of TLR3-specific adjuvants capable of inducing immune responses tailored for viral pathogens.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Oligodesoxirribonucleotídeos/uso terapêutico , Infecções por Orthomyxoviridae/prevenção & controle , RNA de Cadeia Dupla/uso terapêutico , Linfócitos T Citotóxicos/imunologia , Receptor 3 Toll-Like/imunologia , Imunidade Adaptativa/genética , Animais , Células Cultivadas , Ilhas de CpG/imunologia , Testes Imunológicos de Citotoxicidade , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Epitopos de Linfócito T/biossíntese , Epitopos de Linfócito T/uso terapêutico , Feminino , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T Citotóxicos/virologia , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...