Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(3): e2213317120, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36634143

RESUMO

There is an urgent need to develop novel drugs to reduce the mortality from severe infectious diseases with the emergence of new pathogens, including Coronavirus disease 2019 (COVID-19). Although current drugs effectively suppress the proliferation of pathogens, immune cell activation, and inflammatory cytokine functions, they cannot completely reduce mortality from severe infections and sepsis. In this study, we focused on the endothelial cell-specific protein, Roundabout 4 (Robo4), which suppresses vascular permeability by stabilizing endothelial cells, and investigated whether enhanced Robo4 expression could be a novel therapeutic strategy against severe infectious diseases. Endothelial-specific overexpression of Robo4 suppresses vascular permeability and reduces mortality in lipopolysaccharide (LPS)-treated mice. Screening of small molecules that regulate Robo4 expression and subsequent analysis revealed that two competitive small mothers against decapentaplegic (SMAD) signaling pathways, activin receptor-like kinase 5 (ALK5)-SMAD2/3 and ALK1-SMAD1/5, positively and negatively regulate Robo4 expression, respectively. An ALK1 inhibitor was found to increase Robo4 expression in mouse lungs, suppress vascular permeability, prevent extravasation of melanoma cells, and decrease mortality in LPS-treated mice. The inhibitor suppressed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced endothelial barrier disruption and decreased mortality in mice infected with SARS-CoV-2. These results indicate that enhancing Robo4 expression is an efficient strategy to suppress vascular permeability and mortality in severe infectious diseases, including COVID-19, and that small molecules that upregulate Robo4 can be potential therapeutic agents against these diseases.


Assuntos
COVID-19 , Endotoxemia , Animais , Camundongos , Receptores de Superfície Celular/metabolismo , Permeabilidade Capilar , Células Endoteliais/metabolismo , Transdução de Sinais , Regulação para Cima , Endotoxemia/metabolismo , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , COVID-19/metabolismo , SARS-CoV-2/metabolismo
2.
Am J Physiol Renal Physiol ; 326(6): F931-F941, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38634132

RESUMO

Coronavirus disease 2019 (COVID-19) induces respiratory dysfunction as well as kidney injury. Although the kidney is considered a target organ of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and affected by the COVID-19-induced cytokine storm, the mechanisms of renal reaction in SARS-CoV-2 infection are unknown. In this study, a murine COVID-19 model was induced by nasal infection with mouse-adapted SARS-CoV-2 (MA10). MA10 infection induced body weight loss along with lung inflammation in mice 4 days after infection. Serum creatinine levels and the urinary albumin/creatinine ratio increased on day 4 after MA10 infection. Measurement of the urinary neutrophil gelatinase-associated lipocalin/creatinine ratio and hematoxylin and eosin staining revealed tubular damage in MA10-infected murine kidneys, indicating kidney injury in the murine COVID-19 model. Interferon (IFN)-γ and interleukin-6 upregulation in the sera of MA10-infected mice, along with the absence of MA10 in the kidneys, implied that the kidneys were affected by the MA10 infection-induced cytokine storm rather than by direct MA10 infection of the kidneys. RNA-sequencing analysis revealed that antiviral genes, such as the IFN/Janus kinase (JAK) pathway, were upregulated in MA10-infected kidneys. Upon administration of the JAK inhibitor baricitinib on days 1-3 after MA10 infection, an antiviral pathway was suppressed, and MA10 was detected more frequently in the kidneys. Notably, JAK inhibition upregulated the hypoxia response and exaggerated kidney injury. These results suggest that endogenous antiviral activity protects against SARS-CoV-2-induced kidney injury in the early phase of infection, providing valuable insights into the pathogenesis of COVID-19-associated nephropathy.NEW & NOTEWORTHY Patients frequently present with acute kidney injury or abnormal urinary findings after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Here, we investigated how the kidneys respond during SARS-CoV-2 infection using a murine coronavirus disease 2019 (COVID-19) model and showed that Janus kinase-mediated endogenous antiviral activity protects against kidney injury in the early phase of SARS-CoV-2 infection. These findings provide valuable insights into the renal pathophysiology of COVID-19.


Assuntos
COVID-19 , Inibidores de Janus Quinases , Purinas , Pirazóis , SARS-CoV-2 , Sulfonamidas , Animais , COVID-19/complicações , Inibidores de Janus Quinases/farmacologia , Inibidores de Janus Quinases/uso terapêutico , Sulfonamidas/farmacologia , Camundongos , Purinas/farmacologia , Pirazóis/farmacologia , Modelos Animais de Doenças , Injúria Renal Aguda/virologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Azetidinas/farmacologia , Azetidinas/uso terapêutico , Janus Quinases/metabolismo , Janus Quinases/antagonistas & inibidores , Rim/patologia , Rim/virologia , Rim/metabolismo , Rim/efeitos dos fármacos , Tratamento Farmacológico da COVID-19 , Antivirais/farmacologia , Antivirais/uso terapêutico , Masculino , Camundongos Endogâmicos C57BL
3.
Biochem Biophys Res Commun ; 711: 149919, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38608435

RESUMO

Subunit vaccines are among the most useful vaccine modalities; however, their low immunogenicity necessitates the addition of adjuvants. Although adjuvants improve immune responses induced by vaccines, they often cause adverse reactions. To address this, we developed an adjuvant-free subunit vaccine platform that uses pre-existing antibodies generated from past infections or vaccinations as carriers for the delivery of vaccine antigens. Although we have confirmed the usefulness of this platform for nasal vaccines, its suitability as a parenterally injectable vaccine remains uncertain. Here, we verified the potential of our vaccine platform to harness pre-existing immunity for parenterally injectable vaccines. We generated RBD-HA by combining the receptor binding domain (RBD) derived from SARS-CoV-2 as a vaccine antigen with hemagglutinin (HA) sourced from influenza viruses to serve as the carrier protein. We revealed that subcutaneous vaccination with RBD-HA effectively triggered strong RBD-specific IgG responses in mice previously infected with the influenza A virus, even in the absence of adjuvants, and conferred protection to mice against SARS-CoV-2 upon challenge. Furthermore, we revealed that vaccination with RBD-HA did not induce an inflammatory response, such as inflammatory cytokine production, swelling, and recruitment of inflammatory immune cells, whereas conventional vaccines combined with adjuvants induced these adverse reactions. In addition, we demonstrated the remarkable versatility of this platform using a vaccine antigen derived from Streptococcus pneumoniae. These findings indicate the potential of this adjuvant-free vaccine platform to enhance the efficacy of parenterally injectable subunit vaccines and reduce adverse reactions.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Imunoglobulina G , Camundongos Endogâmicos BALB C , SARS-CoV-2 , Animais , Imunoglobulina G/imunologia , Imunoglobulina G/sangue , Camundongos , SARS-CoV-2/imunologia , COVID-19/prevenção & controle , COVID-19/imunologia , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/administração & dosagem , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/administração & dosagem , Humanos , Anticorpos Antivirais/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem
4.
Appl Microbiol Biotechnol ; 103(15): 5947-5955, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31175431

RESUMO

Over the past two decades, lactic acid bacteria (LAB) have been intensively studied as potential bacterial carriers for therapeutic materials, such as vaccine antigens, to the mucosal tissues. LAB have several attractive advantages as carriers of mucosal vaccines, and the effectiveness of LAB vaccines has been demonstrated in numerous studies. Research on LAB vaccines to date has focused on whether antigen-specific immunity, particularly antibody responses, can be induced. However, with recent developments in immunology, microbiology, and vaccinology, more detailed analyses of the underlying mechanisms, especially, of the induction of cell-mediated immunity and memory cells, have been required for vaccine development and licensure. In this mini-review, we will discuss the issues, including (i) immune responses other than antibody production, (ii) persistence of LAB vaccine immunity, (iii) comparative evaluation of LAB vaccines with any existing or reference vaccines, (iv) strategies for increasing the effectiveness of LAB vaccines, and (iv) effects of microbiota on the efficacy of LAB vaccines. Although these issues have been rarely studied or discussed to date in relation to LAB vaccine research, further understanding of them is critical for the practical application of LAB vaccine systems.


Assuntos
Portadores de Fármacos , Imunidade nas Mucosas , Lactobacillales/genética , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Administração através da Mucosa , Pesquisa Biomédica/tendências , Tecnologia Farmacêutica/tendências , Vacinas Sintéticas/administração & dosagem
5.
Front Immunol ; 14: 1082273, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36742329

RESUMO

Direct administration of vaccines to mucosal surfaces, such as via oral or nasal vaccination, represents an attractive alternative, or complement, to current parenteral vaccination because it has a potential to induce antigen-specific immunity both at mucosal and systemic tissues. Although bacterium-like particles (BLPs), peptidoglycan structures derived from lactic acid bacteria, have been investigated as a novel adjuvant for oral or nasal vaccines, it remains unclear whether the administration routes differ the adjuvant effect of BLPs. Here, we showed that the adjuvant effect of BLPs from Lactococcus lactis NZ9000 is greater with the nasal administration than with the oral administration. We conjugated BLPs with Tir, a virulence factor of Citrobacter rodentium, as a model adjuvant-antigen complex, and found that nasal, but not oral, immunization of mice with BLP-Tir induced robust antigen-specific IgA responses at the respiratory and intestinal mucosa, IgG2b-skewed systemic responses, and Th17 cellular responses. As one of the underlying mechanisms, we demonstrated that the nasal administration has a greater delivery efficiency (~1,000-fold) of the BLPs-conjugated antigens to mucosal-associated lymphoid tissues than the oral administration. Furthermore, the nasal, but not oral, administration of BLP-Tir elicited robust innate immune responses that were characterized by the expression of various pro-inflammatory cytokines and chemokines in the mucosal-associated lymphoid tissues. Considering these findings together, we anticipate that BLPs can be an attractive novel adjuvant for nasal vaccines targeting not only respiratory but also gastrointestinal infectious diseases.


Assuntos
Adjuvantes Imunológicos , Vacinas , Animais , Camundongos , Imunização , Imunidade nas Mucosas , Antígenos de Bactérias , Adjuvantes Farmacêuticos , Mucosa Intestinal
6.
Front Immunol ; 14: 1224634, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37720231

RESUMO

Introduction: Vaccinations are ideal for reducing the severity of clinical manifestations and secondary complications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); however, SARS-CoV-2 continues to cause morbidity and mortality worldwide. In contrast to parenteral vaccines such as messenger RNA vaccines, nasal vaccines are expected to be more effective in preventing viral infections in the upper respiratory tract, the primary locus for viral infection and transmission. In this study, we examined the prospects of an inactivated whole-virion (WV) vaccine administered intranasally against SARS-CoV-2. Methods: Mice were immunized subcutaneously (subcutaneous vaccine) or intranasally (nasal vaccine) with the inactivated WV of SARS-CoV-2 as the antigen. Results: The spike protein (S)-specific IgA level was found to be higher upon nasal vaccination than after subcutaneous vaccination. The level of S-specific IgG in the serum was also increased by the nasal vaccine, although it was lower than that induced by the subcutaneous vaccine. The nasal vaccine exhibited a stronger defense against viral invasion in the upper respiratory tract than the subcutaneous vaccine and unimmunized control; however, both subcutaneous and nasal vaccines provided protection in the lower respiratory tract. Furthermore, we found that intranasally administered inactivated WV elicited robust production of S-specific IgA in the nasal mucosa and IgG in the blood of mice previously vaccinated with messenger RNA encoding the S protein. Discussion: Overall, these results suggest that a nasal vaccine containing inactivated WV can be a highly effective means of protection against SARS-CoV-2 infection.


Assuntos
COVID-19 , Vacinas , Animais , Camundongos , SARS-CoV-2 , Imunidade nas Mucosas , COVID-19/prevenção & controle , Mucosa Nasal , Imunoglobulina A , Imunoglobulina G
7.
J Clin Invest ; 133(23)2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38038133

RESUMO

Intranasal vaccines are anticipated to be powerful tools for combating many infectious diseases, including SARS-CoV-2, because they induce not only systemic immunity but also mucosal immunity at the site of initial infection. However, they are generally inefficient in inducing an antigen-specific immune response without adjuvants. Here, we developed an adjuvant-free intranasal vaccine platform that utilizes the preexisting immunity induced by previous infection or vaccination to enhance vaccine effectiveness. We made RBD-HA, a fusion of the receptor-binding domain (RBD) of spike derived from SARS-CoV-2 as a vaccine target with HA derived from influenza A virus (IAV) as a carrier protein. Intranasal immunization of previously IAV-infected mice with RBD-HA without an adjuvant elicited robust production of RBD-specific systemic IgG and mucosal IgA by utilizing both HA-specific preexisting IgG and CD4+ T cells. Consequently, the mice were efficiently protected from SARS-CoV-2 infection. Additionally, we demonstrated the high versatility of this intranasal vaccine platform by assessing various vaccine antigens and preexisting immunity associated with a variety of infectious diseases. The results of this study suggest the promising potential of this intranasal vaccine platform to address problems associated with intranasal vaccines.


Assuntos
Doenças Transmissíveis , Vírus da Influenza A , Vacinas contra Influenza , Animais , Camundongos , Hemaglutininas , Anticorpos Antivirais , Imunização , Vacinação , Adjuvantes Imunológicos/farmacologia , Imunidade nas Mucosas , Vírus da Influenza A/genética , Imunoglobulina G
8.
Sci Adv ; 8(38): eabo6783, 2022 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-36129989

RESUMO

In the initial process of coronavirus disease 2019 (COVID-19), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects respiratory epithelial cells and then transfers to other organs the blood vessels. It is believed that SARS-CoV-2 can pass the vascular wall by altering the endothelial barrier using an unknown mechanism. In this study, we investigated the effect of SARS-CoV-2 on the endothelial barrier using an airway-on-a-chip that mimics respiratory organs and found that SARS-CoV-2 produced from infected epithelial cells disrupts the barrier by decreasing Claudin-5 (CLDN5), a tight junction protein, and disrupting vascular endothelial cadherin-mediated adherens junctions. Consistently, the gene and protein expression levels of CLDN5 in the lungs of a patient with COVID-19 were decreased. CLDN5 overexpression or Fluvastatin treatment rescued the SARS-CoV-2-induced respiratory endothelial barrier disruption. We concluded that the down-regulation of CLDN5 expression is a pivotal mechanism for SARS-CoV-2-induced endothelial barrier disruption in respiratory organs and that inducing CLDN5 expression is a therapeutic strategy against COVID-19.


Assuntos
COVID-19 , Claudina-5/metabolismo , SARS-CoV-2 , Claudina-5/genética , Células Endoteliais/metabolismo , Fluvastatina/metabolismo , Fluvastatina/farmacologia , Humanos , Proteínas de Junções Íntimas/metabolismo
9.
Immunohorizons ; 5(10): 870-883, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34702763

RESUMO

Citrobacter rodentium is a murine pathogenic bacterium that adheres to intestinal epithelial cells, resulting in loss of microvilli and pedestal formation, and alters multiple cellular processes, including actin dynamics. Translocated intimin receptor (Tir), one of its virulence factors, functions as receptor for intimin, a bacterial adhesin, thereby mediating bacterial adhesion to epithelial cells. Although robust immune responses are induced to eliminate pathogenic bacteria in the host, they are suppressed against harmless commensal bacteria. The mechanism(s) underlying such a differentiation remains unclear. This study sought to determine the roles of intimate adhesion in the induction of specific immune responses upon C. rodentium infection. To this end, microbiota-depleted mice were infected with the Tir-F strain expressing full-length Tir or mutant strains expressing the C-terminal truncated Tir that is defective in intimin binding and host cell actin polymerization. There were no differences in the colonization kinetics and Abs responses against C. rodentium LPS among the strains, whereas Abs against the virulence factors were only produced on Tir-F infection. Although there were no differences in the virulence factors mRNA expression levels, colonic hyperplasia, and bacterial translocation to the systemic organs irrespective of the strain, adhesion to colonic epithelial cells was reduced in the mutant strain-infected mice. Furthermore, transcriptomic analysis indicated that robust inflammatory and immune responses were only induced in the Tir-F-infected group and were suppressed in the mutant-infected groups. Taken together, these findings suggest that Tir-mediated intimate adhesion induces inflammatory and immune responses, resulting in the induction of virulence factor-specific Abs.


Assuntos
Aderência Bacteriana/imunologia , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Mucosa Intestinal/patologia , Fatores de Virulência/metabolismo , Adesinas Bacterianas/metabolismo , Animais , Aderência Bacteriana/genética , Linhagem Celular Tumoral , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidade , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Feminino , Microbioma Gastrointestinal/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Camundongos , Mutação , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Organismos Livres de Patógenos Específicos
10.
J Microbiol Methods ; 159: 62-68, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30817947

RESUMO

Evaluation of the efficacy of vaccine candidates that prevent enteropathogenic and enterohemorrhagic Escherichia coli (EPEC/EHEC) infection in mouse models is difficult due to their limited pathogenicity in mice. Citrobacter rodentium, a murine pathogenic bacterium that shares its infection strategy and virulence genes with EPEC/EHEC, has been used as a model pathogen to develop novel vaccine strategies or platforms for these bacteria. However, there are few reports on the comparative effectiveness of novel vaccine platforms as no C. rodentium vaccines have yet been prepared by standard methods such as bacteria attenuation or inactivation. In this study, we investigated the protective effect of the oral administration of formalin-inactivated C. rodentium (Fo-CR) on C. rodentium infection in two mouse strains, C57BL/6 and C3H/HeN, as these strains have different degrees of susceptibility to infection. In C57BL/6 mice, administration of Fo-CR induced significant C. rodentium-specific mucosal and systemic antibody responses, promoted bacterial clearance from the gut and inhibited colonic hyperplasia. Furthermore, in C3H/HeN mice, the administration followed by lethal C. rodentium infection induced significantly high avidity serum IgG specific to C. rodentium and inhibited death, body weight loss, and bacterial invasion to visceral organs. In conclusion, the oral administration of Fo-CR resulted in the protection of mice from C. rodentium infection, indicating that it serves as a reference method for evaluating the efficacy of novel oral vaccine candidates or platforms.


Assuntos
Vacinas Bacterianas/administração & dosagem , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/prevenção & controle , Administração Oral , Animais , Anticorpos Antibacterianos/imunologia , Vacinas Bacterianas/imunologia , Citrobacter rodentium/genética , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/microbiologia , Feminino , Humanos , Imunização , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA