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.
Vaccine ; 42(9): 2144-2149, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38461047

RESUMO

Intranasal administration of vaccines is an attractive delivery route to fight viral respiratory infections. However, there are only a few intranasal vaccines used in human, emphasizing the critical need to identify novel safe mucosal adjuvants and antigen delivery systems to expand their usage. We recently revealed an immunostimulating nanoparticle based on a fragment (R4R5) of the Curli-specific gene A (CsgA) protein that confers protection against influenza A virus (IAV) when conjugated to three repeats of the highly conserved M2e epitope and administrated intramuscularly. Herein, the efficacy of this 3M2e-R4R5 nanovaccine was investigated upon administration by intranasal instillation. By triggering robust M2e-specific humoral and cellular responses, both systemic and locally in the respiratory tract, and by priming alveolar macrophages, the intranasal vaccine protected mice against a lethal IAV challenge without the use of additional adjuvant. Thus, CsgA-based nanostructures could serve as a safe and self-adjuvanted antigen delivery system for mucosal immunization.


Assuntos
Vírus da Influenza A , Vacinas contra Influenza , Infecções por Orthomyxoviridae , Humanos , Animais , Camundongos , Nanovacinas , Administração Intranasal , Epitopos , Adjuvantes Imunológicos , Anticorpos Antivirais , Camundongos Endogâmicos BALB C
2.
ACS Infect Dis ; 9(6): 1232-1244, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37200051

RESUMO

Peptides with the ability to self-assemble into nanoparticles have emerged as an attractive strategy to design antigen delivery platforms for subunit vaccines. While toll-like receptor (TLR) agonists are promising immunostimulants, their use as soluble agents is limited by their rapid clearance and off-target inflammation. Herein, we harnessed molecular co-assembly to prepare multicomponent cross-ß-sheet peptide nanofilaments exposing an antigenic epitope derived from the influenza A virus and a TLR agonist. The TLR7 agonist imiquimod and the TLR9 agonist CpG were respectively functionalized on the assemblies by means of an orthogonal pre- or post-assembly conjugation strategy. The nanofilaments were readily uptaken by dendritic cells, and the TLR agonists retained their activity. Multicomponent nanovaccines induced a robust epitope-specific immune response and completely protected immunized mice from a lethal influenza A virus inoculation. This versatile bottom-up approach is promising for the preparation of synthetic vaccines with customized magnitude and polarization of the immune responses.


Assuntos
Vírus da Influenza A , Vacinas contra Influenza , Camundongos , Animais , Peptídeos/química , Adjuvantes Imunológicos/farmacologia , Epitopos
3.
Adv Healthc Mater ; 12(21): e2300224, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37031161

RESUMO

Proteinaceous nanoparticles constitute efficient antigen delivery systems in vaccine formulations due to their size and repetitive nature that mimic most invading pathogens and promote immune activation. Nonetheless, the coadministration of an adjuvant with subunit nanovaccines is usually required to induce a robust, long-lasting, and protective immune response. Herein, the protein Curli-specific gene A (CsgA), which is known to self-assemble into nanofilaments contributing to bacterial biofilm, is exploited to engineer an intrinsically immunostimulatory antigen delivery platform. Three repeats of the M2e antigenic sequence from the influenza A virus matrix 2 protein are merged to the N-terminal domain of engineered CsgA proteins. These chimeric 3M2e-CsgA spontaneously self-assemble into antigen-displaying cross-ß-sheet nanofilaments that activate the heterodimeric toll-like receptors 2 and 1. The resulting nanofilaments are avidly internalized by antigen-presenting cells and stimulate the maturation of dendritic cells. Without the need of any additional adjuvants, both assemblies show robust humoral and cellular immune responses, which translate into complete protection against a lethal experimental infection with the H1N1 influenza virus. Notably, these CsgA-based nanovaccines induce neither overt systemic inflammation, nor reactogenicity, upon mice inoculation. These results highlight the potential of engineered CsgA nanostructures as self-adjuvanted, safe, and versatile antigen delivery systems to fight infectious diseases.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Influenza Humana , Animais , Camundongos , Humanos , Autoantígenos , Adjuvantes Imunológicos , Proteínas da Matriz Viral , Anticorpos Antivirais , Camundongos Endogâmicos BALB C
4.
Vaccines (Basel) ; 10(11)2022 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-36423016

RESUMO

Vaccination has saved billions of human lives and has considerably reduced the economic burden associated with pandemic and endemic infectious diseases. Notwithstanding major advancements in recent decades, multitude diseases remain with no available effective vaccine. While subunit-based vaccines have shown great potential to address the safety concerns of live-attenuated vaccines, their limited immunogenicity remains a major drawback that still needs to be addressed for their use fighting infectious illnesses, autoimmune disorders, and/or cancer. Among the adjuvants and delivery systems for antigens, bacterial proteinaceous supramolecular structures have recently received considerable attention. The use of bacterial proteins with self-assembling properties to deliver antigens offers several advantages, including biocompatibility, stability, molecular specificity, symmetrical organization, and multivalency. Bacterial protein nanoassemblies closely simulate most invading pathogens, acting as an alarm signal for the immune system to mount an effective adaptive immune response. Their nanoscale architecture can be precisely controlled at the atomic level to produce a variety of nanostructures, allowing for infinite possibilities of organized antigen display. For the bottom-up design of the proteinaceous antigen delivery scaffolds, it is essential to understand how the structural and physicochemical properties of the nanoassemblies modulate the strength and polarization of the immune responses. The present review first describes the relationships between structure and the generated immune responses, before discussing potential and current clinical applications.

5.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33255601

RESUMO

Erythropoietin (EPO) is an essential hormone for erythropoiesis, protecting differentiating erythroblasts against apoptosis. EPO has been largely studied in stress or pathological conditions but its regulatory role in steady state erythropoiesis has been less documented. Herein, we report production of EPO by bone marrow-derived macrophages (BMDM) in vitro, and its further enhancement in BMDM conditioned with media from apoptotic cells. Confocal microscopy confirmed EPO production in erythroblastic island (EBI)-associated macrophages, and analysis of mice depleted of EBI macrophages by clodronate liposomes revealed drops in EPO levels in bone marrow (BM) cell lysates, and decreased percentages of EPO-responsive erythroblasts in the BM. We hypothesize that EBI macrophages are an in-situ source of EPO and sustain basal erythropoiesis in part through its secretion. To study this hypothesis, mice were injected with clodronate liposomes and were supplied with exogenous EPO (1-10 IU/mouse) to evaluate potential rescue of the deficiency in erythroid cells. Our results show that at doses of 5 and 10 IU, EPO significantly rescues BM steady state erythropoiesis in mice deficient of macrophages. We propose existence of a mechanism by which EBI macrophages secrete EPO in response to apoptotic erythroblasts, which is in turn controlled by the numbers of erythroid precursors generated.


Assuntos
Apoptose/efeitos dos fármacos , Eritropoese/genética , Eritropoetina/genética , Macrófagos/metabolismo , Animais , Diferenciação Celular/genética , Ácido Clodrônico/farmacologia , Eritroblastos/efeitos dos fármacos , Eritroblastos/metabolismo , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Eritropoese/efeitos dos fármacos , Eritropoetina/farmacologia , Lipossomos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...