Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.787
Filtrar
1.
Front Immunol ; 15: 1420304, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39267752

RESUMO

Despite the decrease in mortality and morbidity due to SARS-CoV-2 infection, the incidence of infections due to Omicron subvariants of SARS-CoV-2 remains high. The mutations acquired by these subvariants, mainly concentrated in the receptor-binding domain (RBD), have caused a shift in infectivity and transmissibility, leading to a loss of effectiveness of the first authorized COVID-19 vaccines, among other reasons, by neutralizing antibody evasion. Hence, the generation of new vaccine candidates adapted to Omicron subvariants is of special interest in an effort to overcome this immune evasion. Here, an optimized COVID-19 vaccine candidate, termed MVA-S(3P_BA.1), was developed using a modified vaccinia virus Ankara (MVA) vector expressing a full-length prefusion-stabilized SARS-CoV-2 spike (S) protein from the Omicron BA.1 variant. The immunogenicity and efficacy induced by MVA-S(3P_BA.1) were evaluated in mice in a head-to-head comparison with the previously generated vaccine candidates MVA-S(3P) and MVA-S(3Pbeta), which express prefusion-stabilized S proteins from Wuhan strain and Beta variant, respectively, and with a bivalent vaccine candidate composed of a combination of MVA-S(3P) and MVA-S(3P_BA.1). The results showed that all four vaccine candidates elicited, after a single intramuscular dose, protection of transgenic K18-hACE2 mice challenged with SARS-CoV-2 Omicron BA.1, reducing viral loads, histopathological lesions, and levels of proinflammatory cytokines in the lungs. They also elicited anti-S IgG and neutralizing antibodies against various Omicron subvariants, with MVA-S(3P_BA.1) and the bivalent vaccine candidate inducing higher titers. Additionally, an intranasal immunization in C57BL/6 mice with all four vaccine candidates induced systemic and mucosal S-specific CD4+ and CD8+ T-cell and humoral immune responses, and the bivalent vaccine candidate induced broader immune responses, eliciting antibodies against the ancestral Wuhan strain and different Omicron subvariants. These results highlight the use of MVA as a potent and adaptable vaccine vector against new emerging SARS-CoV-2 variants, as well as the promising feature of combining multivalent MVA vaccine candidates.


Assuntos
Anticorpos Antivirais , Vacinas contra COVID-19 , COVID-19 , Imunidade Celular , Imunidade Humoral , Camundongos Transgênicos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Animais , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , SARS-CoV-2/imunologia , Vacinas contra COVID-19/imunologia , COVID-19/prevenção & controle , COVID-19/imunologia , Camundongos , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Humanos , Enzima de Conversão de Angiotensina 2/imunologia , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Feminino , Vacinas de DNA/imunologia , Vaccinia virus/imunologia , Vaccinia virus/genética , Imunogenicidade da Vacina
2.
Emerg Microbes Infect ; 13(1): 2401931, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39233480

RESUMO

In 2022, the monkeypox virus (mpox virus, MPXV) exhibited global dissemination across six continents, representing a notable challenge owing to the scarcity of targeted antiviral interventions. Passive immunotherapy, such as the use of monoclonal antibodies (mAbs) and bispecific antibodies (bsAbs), has emerged as a promising option for antiviral regimens. Here, we generated several mAbs against M1R and B6R of MPXV, and subsequently characterized the antiviral activity of these antibodies both in vitro and in vivo. Two neutralizing mAbs, M1H11 and M3B2, targeting M1R, and one B6R-specific mAb, B7C9, were identified. They exhibited varying antiviral efficacy against vaccinia virus (VACV) in vitro and in vivo. A cocktail comprising M1H11 and M3B2 demonstrated a superior protective effect in vivo. A bsAb, Bis-M1M3, was engineered by conjugating the fragment crystallizable (Fc) region of the human-mouse chimeric engineered M1H11 with the single-chain fragment variable (scFv) of M3B2. In mice challenged with MPXV, Bis-M1M3 showed a notable protective effects. Analysis of neutralization mechanism showed that these mAbs and Bis-M1M3 exerted virus-neutralizing effects before the virus infects cells. In vivo pharmacokinetic experiments showed that Bis-M1M3 has a long half-life in rhesus macaques. This study provides crucial insights for further research on broad-spectrum antiviral drugs against MPXV and other orthopoxviruses.


Assuntos
Anticorpos Biespecíficos , Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , Monkeypox virus , Animais , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/farmacologia , Camundongos , Anticorpos Antivirais/imunologia , Humanos , Anticorpos Neutralizantes/imunologia , Anticorpos Monoclonais/imunologia , Monkeypox virus/imunologia , Camundongos Endogâmicos BALB C , Feminino , Mpox/imunologia , Mpox/virologia , Vaccinia virus/imunologia , Testes de Neutralização
3.
Cell Mol Life Sci ; 81(1): 400, 2024 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-39264480

RESUMO

Dendritic cells (DCs) play a crucial role in orchestrating immune responses, particularly in promoting IFNγ-producing-CD8 cytotoxic T lymphocytes (CTLs) and IFNγ-producing-CD4 T helper 1 (Th1) cells, which are essential for defending against viral infections. Additionally, the nuclear envelope protein lamin A/C has been implicated in T cell immunity. Nevertheless, the intricate interplay between innate and adaptive immunity in response to viral infections, particularly the role of lamin A/C in DC functions within this context, remains poorly understood. In this study, we demonstrate that mice lacking lamin A/C in myeloid LysM promoter-expressing cells exhibit a reduced capacity to induce Th1 and CD8 CTL responses, leading to impaired clearance of acute primary Vaccinia virus (VACV) infection. Remarkably, in vitro-generated granulocyte macrophage colony-stimulating factor bone marrow-derived DCs (GM-CSF BMDCs) show high levels of lamin A/C. Lamin A/C absence on GM-CSF BMDCs does not affect the expression of costimulatory molecules on the cell membrane but it reduces the cellular ability to form immunological synapses with naïve CD4 T cells. Lamin A/C deletion induces alterations in NFκB nuclear localization, thereby influencing NF-κB-dependent transcription. Furthermore, lamin A/C ablation modifies the gene accessibility of BMDCs, predisposing these cells to mount a less effective antiviral response upon TLR stimulation. This study highlights the critical role of DCs in interacting with CD4 T cells during antiviral responses and proposes some mechanisms through which lamin A/C may modulate DC function via gene accessibility and transcriptional regulation.


Assuntos
Células Dendríticas , Lamina Tipo A , Camundongos Endogâmicos C57BL , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Animais , Lamina Tipo A/metabolismo , Lamina Tipo A/genética , Camundongos , NF-kappa B/metabolismo , Vaccinia virus/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Camundongos Knockout , Vacínia/imunologia , Células Th1/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Sinapses Imunológicas/metabolismo , Sinapses Imunológicas/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
4.
Euro Surveill ; 29(38)2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39301741

RESUMO

In response to the mpox outbreak in 2022 and 2023, widespread vaccination with modified vaccinia Ankara-Bavarian Nordic (MVA-BN, also known as JYNNEOS or Imvanex) was initiated. Here, we demonstrate that orthopoxvirus-specific binding and MVA-neutralising antibodies waned to undetectable levels 1 year post vaccination in at-risk individuals who received two doses of MVA-BN administered subcutaneously with an interval of 4 weeks, without prior smallpox or mpox vaccination. Continuous surveillance is essential to understand the impact of declining antibody levels.


Assuntos
Anticorpos Antivirais , Orthopoxvirus , Vacinação , Humanos , Anticorpos Antivirais/sangue , Orthopoxvirus/imunologia , Países Baixos/epidemiologia , Masculino , Adulto , Feminino , Vacina Antivariólica/administração & dosagem , Vacina Antivariólica/imunologia , Pessoa de Meia-Idade , Anticorpos Neutralizantes/sangue , Surtos de Doenças/prevenção & controle , Varíola/prevenção & controle , Infecções por Poxviridae/prevenção & controle , Mpox/prevenção & controle , Vaccinia virus/imunologia , Adulto Jovem , Adolescente
5.
Nat Commun ; 15(1): 7267, 2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39179564

RESUMO

Targeted antineoplastic immunotherapies have achieved remarkable clinical outcomes. However, resistance to these therapies due to target absence or antigen shedding limits their efficacy and excludes tumours from candidacy. To address this limitation, here we engineer an oncolytic rhabdovirus, vesicular stomatitis virus (VSVΔ51), to express a truncated targeted antigen, which allows for HER2-targeting with trastuzumab. The truncated HER2 (HER2T) lacks signaling capabilities and is efficiently expressed on infected cell surfaces. VSVΔ51-mediated HER2T expression simulates HER2-positive status in tumours, enabling effective treatment with the antibody-drug conjugate trastuzumab emtansine in vitro, ex vivo, and in vivo. Additionally, we combine VSVΔ51-HER2T with an oncolytic vaccinia virus expressing a HER2-targeted T-cell engager. This dual-virus therapeutic strategy demonstrates potent curative efficacy in vivo in female mice using CD3+ infiltrate for anti-tumour immunity. Our findings showcase the ability to tailor the tumour microenvironment using oncolytic viruses, thereby enhancing compatibility with "off-the-shelf" targeted therapies.


Assuntos
Imunoterapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Receptor ErbB-2 , Linfócitos T , Trastuzumab , Vaccinia virus , Animais , Feminino , Humanos , Imunoterapia/métodos , Camundongos , Receptor ErbB-2/metabolismo , Receptor ErbB-2/imunologia , Receptor ErbB-2/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Linfócitos T/imunologia , Linhagem Celular Tumoral , Vaccinia virus/genética , Vaccinia virus/imunologia , Trastuzumab/uso terapêutico , Trastuzumab/farmacologia , Microambiente Tumoral/imunologia , Vesiculovirus/genética , Vesiculovirus/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos Endogâmicos BALB C
6.
Viruses ; 16(8)2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39205158

RESUMO

Mpox (formerly known as monkeypox) is a zoonotic disease caused by monkeypox virus (MPXV), a DNA virus belonging to the Orthopoxvirus genus, in the Poxviridae family. The disease constitutes a moderate risk to public health at the global level. The MPXV A29L protein plays a crucial role in coordinating virion assembly and facilitating important virus-host interactions. This study focused on the expression, purification, and recombinant protein synthesis of the A29L protein of MPXV using prokaryotic systems. Using hybridoma technology, we successfully generated the monoclonal antibodies (mAbs) 1E12 and 4B2, which specifically recognize the A29L protein. These mAbs were found to be suitable for use in indirect immunofluorescence assays (IFA), Western blotting, and immunoprecipitation (IP). Our investigation also revealed that mAbs 1E12 and 4B2 could detect the A27L protein, a homologous protein found in the vaccinia virus Western Reserve (VACV WR) strain, using IFA, Western blotting, and immunoprecipitation (IP). Using mAbs 1E12 and 4B2 as primary immunological probes, A27L protein expression was detected as early as 6 h postinfection with VACV WR, with increasing protein levels being observed throughout the infection. This study enhances our understanding of the protein structure and function of MPXV and contributes to the development of specific MPXV detection methods.


Assuntos
Anticorpos Monoclonais , Anticorpos Antivirais , Monkeypox virus , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/isolamento & purificação , Animais , Camundongos , Anticorpos Antivirais/imunologia , Monkeypox virus/imunologia , Monkeypox virus/genética , Camundongos Endogâmicos BALB C , Proteínas Virais/imunologia , Proteínas Virais/genética , Humanos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/genética , Feminino , Vaccinia virus/imunologia , Vaccinia virus/genética , Hibridomas
7.
Antiviral Res ; 230: 105974, 2024 10.
Artigo em Inglês | MEDLINE | ID: mdl-39089331

RESUMO

The outbreak of 2022 monkeypox virus (MPXV) infection in nonendemic regions is a global public health concern. A highly effective and safe MPXV vaccine that is available to the general public is urgently needed to control the mpox pandemic. Here, we developed a multivalent mRNA vaccine candidate, MPXV-1103, which expresses the full-length B6, A35, A29 and M1 proteins with three flexible linkers (G4S1)3 in a single sequence. Compared with the monovalent MPXV mRNA vaccine candidates or the quadrivalent mRNA vaccine from mixtures of the four monovalent MPXV mRNA vaccines, MPXV-1103 elicits a robust humoral response and an MPXV-specific T-cell response and protects mice from lethal vaccinia virus (VACV) challenge, with no live virus detected in the nasal or lungs even at dosages as low as 1 µg. Furthermore, analysis of complete blood counts and photomicrographs of tissue from the main organs of mice vaccinated with MPXV-1103 at doses of 5 µg and 20 µg revealed that two doses of MPXV-1103 did not cause any observable pathological changes in the mice. Collectively, our results suggest that MPXV-1103, with features of high efficacy, safety and a simplified manufacturing process, is a promising vaccine candidate for defending against MPXV infection.


Assuntos
Anticorpos Antivirais , Camundongos Endogâmicos BALB C , Vaccinia virus , Vacinas de mRNA , Animais , Camundongos , Vaccinia virus/imunologia , Vaccinia virus/genética , Feminino , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Vacínia/prevenção & controle , Vacínia/imunologia , Mpox/prevenção & controle , Mpox/imunologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Monkeypox virus/imunologia , Linfócitos T/imunologia , Imunidade Humoral
8.
Emerg Microbes Infect ; 13(1): 2387906, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39087555

RESUMO

Despite the high efficiency of current SARS-CoV-2 mRNA vaccines in reducing COVID-19 morbidity and mortality, waning immunity and the emergence of resistant variants underscore the need for novel vaccination strategies. This study explores a heterologous mRNA/Modified Vaccinia virus Ankara (MVA) prime/boost regimen employing a trimeric form of the receptor binding domain (RBD) of the SARS-CoV-2 spike (S) protein compared to a homologous MVA/MVA regimen. In C57BL/6 mice, the RBD was delivered during priming via an mRNA vector encapsulated in nanoemulsions (NE) or lipid nanoparticles (LNP), followed by a booster with a replication-deficient MVA-based recombinant virus (MVA-RBD). This heterologous mRNA/MVA regimen elicited strong anti-RBD binding and neutralizing antibodies (BAbs and NAbs) against both the ancestral SARS-CoV-2 strain and different variants of concern (VoCs). Additionally, this protocol induced robust and polyfunctional RBD-specific CD4 and CD8 T cell responses, particularly in animals primed with mLNP-RBD. In K18-hACE2 transgenic mice, the LNP-RBD/MVA combination provided complete protection from morbidity and mortality following a live SARS-CoV-2 challenge compared with the partial protection observed with mNE-RBD/MVA or MVA/MVA regimens. Although the mNE-RBD/MVA regimen only protects half of the animals, it was able to induce antibodies with Fc-mediated effector functions besides NAbs. Moreover, viral replication and viral load in the respiratory tract were markedly reduced and decreased pro-inflammatory cytokine levels were observed. These results support the efficacy of heterologous mRNA/MVA vaccine combinations over homologous MVA/MVA regimen, using alternative nanocarriers that circumvent intellectual property restrictions of current mRNA vaccine formulations.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Vacinas contra COVID-19 , COVID-19 , Camundongos Endogâmicos C57BL , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Vaccinia virus , Animais , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/administração & dosagem , SARS-CoV-2/imunologia , SARS-CoV-2/genética , Camundongos , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , COVID-19/prevenção & controle , COVID-19/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Anticorpos Neutralizantes/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Humanos , Feminino , Nanopartículas/administração & dosagem , Vacinação , Vacinas de mRNA/administração & dosagem , Camundongos Transgênicos , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Linfócitos T CD8-Positivos/imunologia , Enzima de Conversão de Angiotensina 2/imunologia , Enzima de Conversão de Angiotensina 2/genética , Lipossomos
9.
Immun Inflamm Dis ; 12(8): e1360, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39150224

RESUMO

BACKGROUND: Messenger RNA (mRNA) vaccines emerged as a powerful tool in the fight against infections. Unlike traditional vaccines, this unique type of vaccine elicits robust and persistent innate and humoral immune response with a unique host cell-mediated pathogen gene expression and antigen presentation. METHODS: This offers a novel approach to combat poxviridae infections. From the genome of vaccinia and Mpox viruses, three key genes (E8L, E7R, and H3L) responsible for virus attachment and virulence were selected and employed for designing the candidate mRNA vaccine against vaccinia and Mpox viral infection. Various bioinformatics tools were employed to generate (B cell, CTL, and HTL) epitopes, of which 28 antigenic and immunogenic epitopes were selected and are linked to form the mRNA vaccine construct. Additional components, including a 5' cap, 5' UTR, adjuvant, 3' UTR, and poly(A) tail, were incorporated to enhance stability and effectiveness. Safety measures such as testing for human homology and in silico immune simulations were implemented to avoid autoimmunity and to mimics the immune response of human host to the designed mRNA vaccine, respectively. The mRNA vaccine's binding affinity was evaluated by docking it with TLR-2, TLR-3, TLR-4, and TLR-9 receptors which are subsequently followed by molecular dynamics simulations for the highest binding one to predict the stability of the binding complex. RESULTS: With a 73% population coverage, the mRNA vaccine looks promising, boasting a molecular weight of 198 kDa and a molecular formula of C8901H13609N2431O2611S48 and it is said to be antigenic, nontoxic and nonallergic, making it safe and effective in preventing infections with Mpox and vaccinia viruses, in comparison with other insilico-designed vaccine for vaccinia and Mpox viruses. CONCLUSIONS: However, further validation through in vivo and in vitro techniques is underway to fully assess its potential.


Assuntos
Biologia Computacional , Vaccinia virus , Vacinas de mRNA , Humanos , Vaccinia virus/imunologia , Vaccinia virus/genética , Biologia Computacional/métodos , Infecções por Poxviridae/prevenção & controle , Infecções por Poxviridae/imunologia , Vacínia/prevenção & controle , Vacínia/imunologia , Vacinas Sintéticas/imunologia , RNA Mensageiro/imunologia , RNA Mensageiro/genética , Vacinas Virais/imunologia , Epitopos de Linfócito B/imunologia , Desenvolvimento de Vacinas , Epitopos de Linfócito T/imunologia
10.
Front Immunol ; 15: 1440407, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39072326

RESUMO

Introduction: Bluetongue (BT), caused by bluetongue virus (BTV), is an important arthropod-borne livestock disease listed by the World Organization for Animal Health. Live-attenuated and inactivated vaccines have permitted to control BT but they do not simultaneously protect against the myriad of BTV serotypes. Recently, we identified the highly conserved BTV nonstructural protein NS1 and the N-terminal region of NS2 as antigens capable of conferring multiserotype protection against BTV. Methods: Here, we designed Modified Vaccinia Ankara (MVA) viral vectors that expressed BTV-4 proteins VP2 or VP7 along with NS1 and NS2-Nt as well as MVAs that expressed proteins VP2, VP7 or NS1 and NS2-Nt. Results: Immunization of IFNAR(-/-) mice with two doses of MVA-NS1-2A-NS2-Nt protected mice from BTV-4M infection by the induction of an antigen-specific T cell immune response. Despite rMVA expressing VP7 alone were not protective in the IFNAR(-/-) mouse model, inclusion of VP7 in the vaccine formulation amplified the cell-mediated response induced by NS1 and NS2-Nt. Expression of VP2 elicited protective non-cross-reactive neutralizing antibodies (nAbs) in immunized animals and improved the protection observed in the MVA-NS1-2A-NS2-Nt immunized mice when these three BTV antigens were co-expressed. Moreover, vaccines candidates co-expressing VP2 or VP7 along with NS1 and NS2-Nt provided multiserotype protection. We assessed protective efficacy of both vaccine candidates in sheep against virulent challenge with BTV-4M. Discussion: Immunization with MVA-VP7-NS1-2A-NS2-Nt partially dumped viral replication and clinical disease whereas administration of MVA-VP2-NS1-2A-NS2-Nt promoted a complete protection, preventing viraemia and the pathology produced by BTV infection.


Assuntos
Vírus Bluetongue , Bluetongue , Proteínas do Capsídeo , Vetores Genéticos , Receptor de Interferon alfa e beta , Vaccinia virus , Proteínas não Estruturais Virais , Vacinas Virais , Animais , Vírus Bluetongue/imunologia , Vírus Bluetongue/genética , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/genética , Bluetongue/prevenção & controle , Bluetongue/imunologia , Bluetongue/virologia , Camundongos , Vacinas Virais/imunologia , Vacinas Virais/genética , Vaccinia virus/genética , Vaccinia virus/imunologia , Receptor de Interferon alfa e beta/genética , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/genética , Camundongos Knockout , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Feminino
11.
Vaccine ; 42(23): 126055, 2024 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-38880691

RESUMO

Vaccination is the best strategy to control Paratuberculosis (PTB), which is a significant disease in cattle and sheep. Previously we showed the humoral and cellular immune response induced by a novel vaccine candidate against PTB based on the Argentinian Mycobacterium avium subspecies paratuberculosis (Map) 6611 strain. To improve 6611 immunogenicity and efficacy, we evaluated this vaccine candidate in mice with two different adjuvants and a heterologous boost with a recombinant modified vaccinia Ankara virus (MVA) expressing the antigen 85A (MVA85A). We observed that boosting with MVA85A did not improve total IgG or specific isotypes in serum induced by one or two doses of 6611 formulated with incomplete Freund's adjuvant (IFA). However, when 6611 was formulated with ISA201 adjuvant, MVA85A boost enhanced the production of IFNγ, Th1/Th17 cytokines (IL-2, TNF, IL-17A) and IL-6, IL-4 and IL-10. Also, this group showed the highest levels of IgG2b and IgG3 isotypes, both important for better protection against Map infection in the murine model. Finally, the heterologous scheme elicited the highest levels of protection after Map challenge (lowest CFU count and liver lesion score). In conclusion, our results encourage further evaluation of 6611 strain + ISA201 prime and MVA85A boost in bovines.


Assuntos
Adjuvantes Imunológicos , Anticorpos Antibacterianos , Citocinas , Modelos Animais de Doenças , Imunização Secundária , Imunoglobulina G , Mycobacterium avium subsp. paratuberculosis , Paratuberculose , Animais , Mycobacterium avium subsp. paratuberculosis/imunologia , Imunização Secundária/métodos , Camundongos , Paratuberculose/prevenção & controle , Paratuberculose/imunologia , Imunoglobulina G/sangue , Citocinas/metabolismo , Feminino , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Adjuvantes Imunológicos/administração & dosagem , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/administração & dosagem , Camundongos Endogâmicos BALB C , Vaccinia virus/imunologia , Vaccinia virus/genética , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/genética , Imunidade Celular/imunologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Adjuvante de Freund/administração & dosagem , Adjuvante de Freund/imunologia
12.
J Med Virol ; 96(6): e29749, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38888113

RESUMO

Human immunodeficiency virus (HIV) infection is still a global public health issue, and the development of an effective prophylactic vaccine inducing potent neutralizing antibodies remains a significant challenge. This study aims to explore the inflammation-related proteins associated with the neutralizing antibodies induced by the DNA/rTV vaccine. In this study, we employed the Olink chip to analyze the inflammation-related proteins in plasma in healthy individuals receiving HIV candidate vaccine (DNA priming and recombinant vaccinia virus rTV boosting) and compared the differences between neutralizing antibody-positive (nab + ) and -negative(nab-) groups. We identified 25 differentially expressed factors and conducted enrichment and correlation analysis on them. Our results revealed that significant expression differences in artemin (ARTN) and C-C motif chemokine ligand 23 (CCL23) between nab+ and -nab- groups. Notably, the expression of CCL23 was negatively corelated to the ID50 of neutralizing antibodies and the intensity of the CD4+ T cell responses. This study enriches our understanding of the immune picture induced by the DNA/rTV vaccine, and provides insights for future HIV vaccine development.


Assuntos
Vacinas contra a AIDS , Anticorpos Neutralizantes , Anticorpos Anti-HIV , Infecções por HIV , HIV-1 , Proteômica , Vaccinia virus , Humanos , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Vaccinia virus/imunologia , Vaccinia virus/genética , Anticorpos Anti-HIV/sangue , Anticorpos Anti-HIV/imunologia , HIV-1/imunologia , HIV-1/genética , Adulto , Vacinas contra a AIDS/imunologia , Masculino , Infecções por HIV/imunologia , Vacinas de DNA/imunologia , Feminino , Voluntários Saudáveis , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Plasma/imunologia , Adulto Jovem
13.
Sci Immunol ; 9(96): eadk8141, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38848340

RESUMO

Lymphatic transport shapes the homeostatic immune repertoire of lymph nodes (LNs). LN-resident memory T cells (TRMs) play an important role in site-specific immune memory, yet how LN TRMs form de novo after viral infection remains unclear. Here, we tracked the anatomical distribution of antiviral CD8+ T cells as they seeded skin and LN TRMs using a model of vaccinia virus-induced skin infection. LN TRMs localized to the draining LNs (dLNs) of infected skin, and their formation depended on the lymphatic egress of effector CD8+ T cells from the skin, already poised for residence. Effector CD8+ T cell transit through skin was required to populate LN TRMs in dLNs, a process reinforced by antigen encounter in skin. Furthermore, LN TRMs were protective against viral rechallenge in the absence of circulating memory T cells. These data suggest that a subset of tissue-infiltrating CD8+ T cells egress from tissues during viral clearance and establish a layer of regional protection in the dLN basin.


Assuntos
Memória Imunológica , Linfonodos , Vasos Linfáticos , Células T de Memória , Camundongos Endogâmicos C57BL , Pele , Vaccinia virus , Animais , Linfonodos/imunologia , Vasos Linfáticos/imunologia , Pele/imunologia , Células T de Memória/imunologia , Camundongos , Memória Imunológica/imunologia , Vaccinia virus/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Vacínia/imunologia , Camundongos Transgênicos
14.
Viruses ; 16(6)2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38932162

RESUMO

Vaccinia virus is the most successful vaccine in human history and functions as a protective vaccine against smallpox and monkeypox, highlighting the importance of ongoing research into vaccinia due to its genetic similarity to other emergent poxviruses. Moreover, vaccinia's ability to accommodate large genetic insertions makes it promising for vaccine development and potential therapeutic applications, such as oncolytic agents. Thus, understanding how superior immunity is generated by vaccinia is crucial for designing other effective and safe vaccine strategies. During vaccinia inoculation by scarification, the skin serves as a primary site for the virus-host interaction, with various cell types playing distinct roles. During this process, hematopoietic cells undergo abortive infections, while non-hematopoietic cells support the full viral life cycle. This differential permissiveness to viral replication influences subsequent innate and adaptive immune responses. Dendritic cells (DCs), key immune sentinels in peripheral tissues such as skin, are pivotal in generating T cell memory during vaccinia immunization. DCs residing in the skin capture viral antigens and migrate to the draining lymph nodes (dLN), where they undergo maturation and present processed antigens to T cells. Notably, CD8+ T cells are particularly significant in viral clearance and the establishment of long-term protective immunity. Here, we will discuss vaccinia virus, its continued relevance to public health, and viral strategies permissive to immune escape. We will also discuss key events and populations leading to long-term protective immunity and remaining key gaps.


Assuntos
Evasão da Resposta Imune , Vaccinia virus , Vacínia , Vaccinia virus/imunologia , Vaccinia virus/genética , Humanos , Animais , Vacínia/imunologia , Vacínia/virologia , Células Dendríticas/imunologia , Replicação Viral , Imunidade Adaptativa , Linfócitos T CD8-Positivos/imunologia
15.
Int J Mol Sci ; 25(11)2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38891837

RESUMO

The proteasome generates the majority of peptides presented on MHC class I molecules. The cleavage pattern of the proteasome has been shown to be changed via the proteasome activator (PA)28 alpha beta (PA28αß). In particular, several immunogenic peptides have been reported to be PA28αß-dependent. In contrast, we did not observe a major impact of PA28αß on the generation of different major histocompatibility complex (MHC) classI ligands. PA28αß-knockout mice infected with the lymphocytic choriomeningitis virus (LCMV) or vaccinia virus showed a normal cluster of differentiation (CD) 8 response and viral clearance. However, we observed that the adoptive transfer of wild-type cells into PA28αß-knockout mice led to graft rejection, but not vice versa. Depletion experiments showed that the observed rejection was mediated by CD8+ cytotoxic T cells. These data indicate that PA28αß might be involved in the development of the CD8+ T cell repertoire in the thymus. Taken together, our data suggest that PA28αß is a crucial factor determining T cell selection and, therefore, impacts graft acceptance.


Assuntos
Linfócitos T CD8-Positivos , Rejeição de Enxerto , Antígenos de Histocompatibilidade Classe I , Camundongos Knockout , Animais , Rejeição de Enxerto/imunologia , Camundongos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/imunologia , Ligantes , Camundongos Endogâmicos C57BL , Vírus da Coriomeningite Linfocítica/imunologia , Vaccinia virus/imunologia
16.
J Virol ; 98(7): e0052124, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38874361

RESUMO

The reoccurrence of successive waves of SARS-CoV-2 variants suggests the exploration of more vaccine alternatives is imperative. Modified vaccinia virus Ankara (MVA) is a virus vector exhibiting excellent safety as well as efficacy for vaccine development. Here, a series of recombinant MVAs (rMVAs) expressing monomerized or trimerized S proteins from different SARS-CoV-2 variants are engineered. Trimerized S expressed from rMVAs is found predominantly as trimers on the surface of infected cells. Remarkably, immunization of mice with rMVAs demonstrates that S expressed in trimer elicits higher levels of binding IgG and IgA, as well as neutralizing antibodies for matched and mismatched S proteins than S in the monomer. In addition, trimerized S expressed by rMVA induces enhanced cytotoxic T-cell responses than S in the monomer. Importantly, the rMVA vaccines expressing trimerized S exhibit superior protection against a lethal SARS-CoV-2 challenge as the immunized animals all survive without displaying any pathological conditions. This study suggests that opting for trimerized S may represent a more effective approach and highlights that the MVA platform serves as an ideal foundation to continuously advance SARS-CoV-2 vaccine development. IMPORTANCE: MVA is a promising vaccine vector and has been approved as a vaccine for smallpox and mpox. Our analyses suggested that recombinant MVA expressing S in trimer (rMVA-ST) elicited robust cellular and humoral immunity and was more effective than MVA-S-monomer. Importantly, the rMVA-ST vaccine was able to stimulate decent cross-reactive neutralization against pseudoviruses packaged using S from different sublineages, including Wuhan, Delta, and Omicron. Remarkably, mice immunized with rMVA-ST were completely protected from a lethal challenge of SARS-CoV-2 without displaying any pathological conditions. Our results demonstrated that an MVA vectored vaccine expressing trimerized S is a promising vaccine candidate for SARS-CoV-2 and the strategy might be adapted for future vaccine development for coronaviruses.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Vacinas contra COVID-19 , COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Vaccinia virus , Animais , Vaccinia virus/genética , Vaccinia virus/imunologia , Camundongos , Anticorpos Antivirais/imunologia , COVID-19/prevenção & controle , COVID-19/imunologia , Vacinas contra COVID-19/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/genética , Anticorpos Neutralizantes/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Feminino , Humanos , Camundongos Endogâmicos BALB C , Multimerização Proteica , Imunoglobulina G/imunologia , Linfócitos T Citotóxicos/imunologia , Imunoglobulina A/imunologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/genética , Vetores Genéticos
17.
J Med Virol ; 96(6): e29728, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38860589

RESUMO

Since May 2022, several countries outside of Africa experienced multiple clusters of monkeypox virus (MPXV)-associated disease. In the present study, anti-MPXV and anti-vaccinia virus (VACV) neutralizing antibody responses were evaluated in two cohorts of subjects from the general Italian population (one half born before the WHO-recommended end of smallpox vaccination in 1980, the other half born after). Higher titers (either against MPXV or VACV) were observed in the cohort of individuals born before the interruption of VACV vaccination. An association between VACV and MPXV antibody levels was observed, suggesting that the smallpox vaccination may confer some degree of cross-protection against MPXV infection. Results from this study highlight low levels of immunity toward the assessed Orthopoxviruses, especially in young adults, advocating the introduction of a VACV- or MPXV-specific vaccine in case of resurgence of monkeypox disease outbreaks.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Monkeypox virus , Vacina Antivariólica , Vacinação , Vaccinia virus , Humanos , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Masculino , Adulto , Feminino , Vacina Antivariólica/imunologia , Vacina Antivariólica/administração & dosagem , Itália/epidemiologia , Monkeypox virus/imunologia , Adulto Jovem , Estudos Soroepidemiológicos , Pessoa de Meia-Idade , Vaccinia virus/imunologia , Mpox/epidemiologia , Mpox/imunologia , Adolescente , Varíola/prevenção & controle , Varíola/imunologia , Varíola/epidemiologia , Proteção Cruzada/imunologia , Idoso , Estudos de Coortes , Criança
18.
Vox Sang ; 119(8): 878-882, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38738359

RESUMO

BACKGROUND AND OBJECTIVES: In May 2022, the United Kingdom reported the first case of chained transmission of the monkeypox (mpox) virus without any known epidemiological links to west or central Africa. The monthly number of mpox patients currently has passed a peak and is declining globally, and infected patients include both non-vaccinated and vaccinated individuals. Herein, the virus-neutralizing (VN) activity against vaccinia viruses, which are considered to cross-react with the mpox virus, in the intravenous immunoglobulin (IVIG) lots derived from donors, including vaccinated Japanese populations, was evaluated to clarify the status of the Japanese blood donor population. MATERIALS AND METHODS: VN titres against vaccinia and human mpox viruses in IVIG lots derived from donors in Japan and the United States manufactured between 1999 and 2021 and 1995 and 2001, respectively, were evaluated by neutralization testing. RESULTS: VN titres of IVIG derived from donors in Japan and the United States against vaccinia and mpox viruses showed a slowly decreasing trend between 1999 and 2021. CONCLUSION: VN titres are expected to decrease in the future since the percentage of vaccinated donors in the donor population seems to have decreased. Therefore, continuous monitoring of VN titres is required.


Assuntos
Doadores de Sangue , Imunoglobulinas Intravenosas , Monkeypox virus , Humanos , Japão , Estados Unidos , Imunoglobulinas Intravenosas/uso terapêutico , Anticorpos Neutralizantes/sangue , Vaccinia virus/imunologia , Anticorpos Antivirais/sangue , Mpox/prevenção & controle , Mpox/epidemiologia , Testes de Neutralização , Feminino , Masculino
19.
Viral Immunol ; 37(4): 216-219, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38717823

RESUMO

In May 2022, mpox began to spread worldwide, posing a serious threat to human public health. Modified Vaccinia Ankara-Bavaria Nordic (MVA-BN) is a live attenuated orthopoxvirus vaccine that has been authorized by the U.S. Food and Drug Administration as the vaccine of choice for the prevention of mpox. In this study, we conducted a meta-analysis of all currently published literature on the efficacy and safety of the MVA-BN vaccine in the real world, showing that the MVA-BN vaccine is effective and safe, with efficacy of up to 75% with a single dose and up to 80% with a two-dose vaccine. Meanwhile, we found that subcutaneous injection has lower local and systemic adverse events than intradermal injection, regardless of single- or two-dose vaccination, and subcutaneous injection is better tolerated in children, the elderly, or people with underlying medical conditions. These results have important reference value for clinical practice.


Assuntos
Eficácia de Vacinas , Vacinas Atenuadas , Humanos , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Infecções por Poxviridae/prevenção & controle , Infecções por Poxviridae/imunologia , Vaccinia virus/imunologia , Vaccinia virus/genética , Vacinação , Injeções Subcutâneas , Injeções Intradérmicas , Vacinas Virais/efeitos adversos , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Orthopoxvirus/imunologia , Orthopoxvirus/genética , Criança
20.
Cell Rep ; 43(6): 114269, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38787725

RESUMO

The 2022 mpox outbreak led the World Health Organization (WHO) to declare it a public health emergency of international concern (PHEIC). There is a need to develop more effective and safer mpox virus (MPXV)-specific vaccines in response to the mpox epidemic. The mRNA vaccine is a promising platform to protect against MPXV infection. In this study, we construct two bivalent MPXV mRNA vaccines, designated LBA (B6R-A29L) and LAM (A35R-M1R), and a quadrivalent mRNA vaccine, LBAAM (B6R-A35R-A29L-M1R). The immunogenicity and protective efficacy of these vaccines alone or in combination were evaluated in a lethal mouse model. All mRNA vaccine candidates could elicit potential antigen-specific humoral and cellular immune responses and provide protection against vaccinia virus (VACV) infection. The protective effect of the combination of two bivalent mRNA vaccines and the quadrivalent vaccine was superior to that of the individual bivalent mRNA vaccine. Our study provides valuable insights for the development of more efficient and safer mRNA vaccines against mpox.


Assuntos
Vaccinia virus , Vacinas de mRNA , Animais , Vaccinia virus/imunologia , Vaccinia virus/genética , Camundongos , Feminino , Vacinas de mRNA/imunologia , Humanos , Camundongos Endogâmicos BALB C , Mpox/prevenção & controle , Mpox/imunologia , Vacínia/imunologia , Vacínia/prevenção & controle , Anticorpos Antivirais/imunologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Imunidade Humoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...