Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Infect Immun ; 88(10)2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32690632

RESUMO

Reproductive failure is the hallmark of brucellosis in animals. An uncommon but important complication in pregnant women who become acutely infected with Brucella melitensis is spontaneous pregnancy loss or vertical transmission to the fetus. Unfortunately, the mechanism behind reproductive failure is still obscure, partially due to the lack of a proper study model. Recently, it was demonstrated that intratracheal (IT) inoculation of nonpregnant guinea pigs would replicate features of clinical disease in humans. To determine if IT inoculation would induce reproductive disease, guinea pigs were infected at mid-gestation and monitored daily for fever and abortions. Fever developed between day 14 to 18 postinoculation, and by 3 weeks postinoculation, 75% of pregnant guinea pigs experienced stillbirths or spontaneous abortions mimicking natural disease. Next, to investigate the guinea pig as a model for evaluating vaccine efficacy during pregnancy, nonpregnant guinea pigs were vaccinated with S19, 16MΔvjbR + Quil-A, or 100 µl PBS + Quil-A (as control). Guinea pigs were bred and vaccinated guinea pigs were challenged at mid-gestation with B. melitensis IT inoculation and monitored for fever and abortions. Vaccination with both vaccines prevented fever and protected against abortion. Together, this study indicates that pregnant guinea pigs are an appropriate animal model to study reproductive disease and offer an improved model to evaluate the ability of vaccine candidates to protect against a serious manifestation of disease.


Assuntos
Vacina contra Brucelose/administração & dosagem , Brucella melitensis/imunologia , Brucelose/prevenção & controle , Modelos Animais de Doenças , Complicações Infecciosas na Gravidez/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Brucella melitensis/patogenicidade , Brucelose/microbiologia , Brucelose/patologia , Feminino , Cobaias , Humanos , Glândulas Mamárias Animais/microbiologia , Glândulas Mamárias Animais/patologia , Placenta/microbiologia , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/microbiologia , Complicações Infecciosas na Gravidez/patologia , Vacinação
2.
Methods Mol Biol ; 2775: 29-46, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38758309

RESUMO

Cryptococcus neoformans and Cryptococcus gattii are the predominant etiological agents of cryptococcosis, a particularly problematic disease in immunocompromised individuals. The increased clinical use of immunosuppressive drugs, the inherent ability of Cryptococcus species to suppress and evade host immune responses, and the emergence of drug-resistant yeast support the need for model systems that facilitate the design of novel immunotherapies and antifungals to combat disease progression. The mouse model of cryptococcosis is a widely used system to study Cryptococcus pathogenesis and the efficacy of antifungal drugs in vivo. In this chapter, we describe three commonly used strategies to establish cryptococcosis in mice: intranasal, intratracheal, and intravenous inoculations. Also, we discuss the methodology for delivering drugs to mice via intraperitoneal injection.


Assuntos
Criptococose , Cryptococcus neoformans , Modelos Animais de Doenças , Animais , Criptococose/microbiologia , Criptococose/tratamento farmacológico , Criptococose/imunologia , Camundongos , Cryptococcus neoformans/patogenicidade , Cryptococcus gattii/patogenicidade , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico
3.
Future Microbiol ; 18: 267-286, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36971082

RESUMO

Background: Pneumonic plague is a fatal respiratory disease caused by Yersinia pestis. Time-course transcriptome analysis on the mechanism of pneumonic plague biphasic syndrome is lacking in the literature. Materials & methods: This study documented the disease course through bacterial load, histopathology, cytokine levels and flow cytometry. RNA-sequencing technology was used to investigate the global transcriptome profile of lung tissue in mice infected with Y. pestis. Results: Inflammation-related genes were significantly upregulated at 48 h post-infection, while genes related to cell adhesion and cytoskeletal structure were downregulated. Conclusion: NOD-like receptor and TNF signaling pathways play a plausible role in pneumonic plague biphasic syndrome and lung injury by controlling the activation and inhibition of the NF-κB signaling pathway.


Assuntos
Peste , Yersinia pestis , Camundongos , Animais , Peste/microbiologia , NF-kappa B/genética , NF-kappa B/metabolismo , Pulmão/microbiologia , Yersinia pestis/genética , Yersinia pestis/metabolismo , Transdução de Sinais , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
4.
Vaccine ; 41(30): 4402-4413, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37308364

RESUMO

Influenza A virus (IAV) is a deadly zoonotic pathogen that remains a burden to global health systems despite continuous vaccinations, indicating the need for an improved vaccine strategy. In this work, we constructed a new recombinant influenza vaccine using Bacillus subtilis spores expressing M2e-FP protein (RSM2eFP) and assessed its potency and efficacy in BALB/c mouse immunized via aerosolized intratracheal inoculation (i.t.) or intragastric (i.g.) administration. Immunization via i.t. route conferred 100 % protection against 20 × LD50 A/PR/8/34 (H1N1) virus compared with only 50 % via the i.g. route. Even when challenged with 40 × LD50 virus, the RSM2eFP vaccine immunized via i.t. provided 80 % protection. Consistently, i.t. inoculation of RSM2eFP spore vaccine induced a stronger lung mucosal immune response and a greater cellular immune response than i.g. administration, as indicated by the high production of IgG and SIgA. In addition, the RSM2eFP spore vaccine diminished the yield of infectious virus in the lung of mice immunized via i.t. These results suggest that i.t. immunization of the RSM2eFP spore vaccine may be a promising strategy for the development of mucosal vaccines against IAV infections.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Animais , Camundongos , Humanos , Influenza Humana/prevenção & controle , Bacillus subtilis/genética , Esporos Bacterianos/genética , Vacinas Sintéticas , Camundongos Endogâmicos BALB C , Anticorpos Antivirais
5.
Front Microbiol ; 13: 1029199, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36338077

RESUMO

Brucella species are considered a significant cause of reproductive pathology in male and female animals. Importantly, Brucella melitensis can induce reproductive disease in humans. Reproductive pathogenesis and evaluation of newly developed countermeasures against brucellosis studies have traditionally utilized female animal models. However, any potential, new intervention for use in humans would need to be evaluated in both sexes. Therefore, animal models for male reproductive brucellosis are desperately needed to understand disease progression. Accordingly, we evaluated guinea pigs and mice using B. melitensis 16 M in an intratracheal model of inoculation at different stages of infection (peracute, acute, and chronic) with an emphasis on determining the effect to the male reproductive organs. Aerosol inoculation resulted in colonization of the reproductive organs (testicle, epididymis, prostate) in both species. Infection peaked during the peracute (1-week post-infection [p.i.]) and acute (2-weeks p.i.) stages of infection in the mouse in spleen, epididymis, prostate, and testicle, but colonization was poorly associated with inflammation. In the guinea pig, peak infection was during the acute stage (4-weeks p.i.) and resulted in inflammation that disrupted spermatogenesis chronically. To determine if vaccine efficacy could be evaluated using these models, males were vaccinated using subcutaneous injection with vaccine candidate 16 MΔvjbR at 109 CFU/100 µl followed by intratracheal challenge with 16 M at 107. Interestingly, vaccination efficacy varied between species and reproductive organs demonstrating the value of evaluating vaccine candidates in multiple models and sexes. Vaccination resulted in a significant reduction in colonization in the mouse, but this could not be correlated with a decrease in inflammation. Due to the ability to evaluate for both colonization and inflammation, guinea pigs seemed the better model not only for assessing host-pathogen interactions but also for future vaccine development efforts.

6.
Front Immunol ; 13: 819089, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154137

RESUMO

Anthrax caused by Bacillus anthracis is a fatal zoonotic disease with a high lethality and poor prognosis. Inhalational anthrax is the most severe of the three forms of anthrax. The currently licensed commercial human anthrax vaccines require a complex immunization procedure for efficacy and have side effects that limit its use in emergent situations. Thus, development of a better anthrax vaccine is necessary. In this study, we evaluate the potency and efficacy of aerosolized intratracheal (i.t.) inoculation with recombinant protective antigen (rPA) subunit vaccines against aerosolized B. anthracis Pasteur II spores (an attenuated strain) challenge in a B10.D2-Hc0 mouse (deficient in complement component C5) model. Immunization of rPA in liquid, powder or powder reconstituted formulations via i.t. route conferred 100% protection against a 20× LD50 aerosolized Pasteur II spore challenge in mice, compared with only 50% of subcutaneous (s.c.) injection with liquid rPA. Consistently, i.t. inoculation of rPA vaccines induced a higher lethal toxin (LeTx) neutralizing antibody titer, a stronger lung mucosal immune response and a greater cellular immune response than s.c. injection. Our results demonstrate that immunization with rPA dry powder vaccine via i.t. route may provide a stable and effective strategy to improve currently available anthrax vaccines and B10.D2-Hc0 mice challenged with B. anthracis attenuated strains might be an alternative model for anthrax vaccine candidate screening.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Imunidade nas Mucosas , Vacinação/métodos , Administração Intranasal , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Neutralizantes/sangue , Bacillus anthracis/imunologia , Feminino , Imunoglobulina G/sangue , Camundongos , Pós , Análise de Sobrevida , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/imunologia
7.
Front Immunol ; 13: 793382, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154110

RESUMO

Pneumonic plague, caused by Yersinia pestis, is an infectious disease with high mortality rates unless treated early with antibiotics. Currently, no FDA-approved vaccine against plague is available for human use. The capsular antigen F1, the low-calcium-response V antigen (LcrV), and the recombinant fusion protein (rF1-LcrV) of Y. pestis are leading subunit vaccine candidates under intense investigation; however, the inability of recombinant antigens to provide complete protection against pneumonic plague in animal models remains a significant concern. In this study, we compared immunoprotection against pneumonic plague provided by rF1, rV10 (a truncation of LcrV), and rF1-V10, and vaccinations delivered via aerosolized intratracheal (i.t.) inoculation or subcutaneous (s.c.) injection. We further considered three vaccine formulations: conventional liquid, dry powder produced by spray freeze drying, or dry powder reconstituted in PBS. The main findings are: (i) rF1-V10 immunization with any formulation via i.t. or s.c. routes conferred 100% protection against Y. pestis i.t. infection; (ii) rF1 or rV10 immunization using i.t. delivery provided significantly stronger protection than rF1 or rV10 immunization via s.c. delivery; and (iii) powder formulations of subunit vaccines induced immune responses and provided protection equivalent to those elicited by unprocessed liquid formulations of vaccines. Our data indicate that immunization with a powder formulation of rF1-V10 vaccines via an i.t. route may be a promising vaccination strategy for providing protective immunity against pneumonic plague.


Assuntos
Vacina contra a Peste/imunologia , Peste/prevenção & controle , Vacinas de Subunidades Antigênicas/imunologia , Yersinia pestis/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Modelos Animais de Doenças , Composição de Medicamentos , Ensaio de Imunoadsorção Enzimática , Feminino , Imunidade nas Mucosas , Imunização/métodos , Camundongos , Camundongos Endogâmicos BALB C , Especificidade de Órgãos , Peste/imunologia , Peste/mortalidade , Vacina contra a Peste/administração & dosagem , Vacina contra a Peste/química , Proteínas Recombinantes/imunologia , Aerossóis e Gotículas Respiratórios , Mucosa Respiratória/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/química
8.
Artigo em Inglês | MEDLINE | ID: mdl-33014895

RESUMO

Plague, which is caused by Yersinia pestis, is one of the most dangerous infectious diseases. No FDA-approved vaccine against plague is available for human use at present. To improve the immune safety of Y. pestis EV76 based live attenuated vaccine and to explore the feasibility of aerosolized intratracheal inoculation (i.t.) route for vaccine delivery, a plasminogen activator protease (pla) gene deletion mutant of the attenuated Y. pestis strain EV76-B-SHU was constructed, and its residual virulence and protective efficacy were evaluated in a mouse model via aerosolized intratracheal inoculation (i.t.) or via subcutaneous injection (s.c.). The residual virulence of EV76-B-SHUΔpla was significantly reduced compared to that of the parental strain EV76-B-SHU following i.t. and s.c. infection. The EV76-B-SHUΔpla induced higher levels of mucosal antibody sIgA in the bronchoalveolar lavage fluid of mice immunized by i.t. but not by s.c.. Moreover, after lethal challenge with Y. pestis biovar Microtus strain 201 (avirulent in humans), the protective efficacy and bacterial clearance ability of the EV76-B-SHUΔpla-i.t. group were comparable to those of the EV76-B-SHUΔpla-s.c. and EV76-B-SHU immunized groups. Thus, the EV76-B-SHUΔpla represents an excellent live-attenuated vaccine candidate against pneumonic plague and aerosolized i.t. represents a promising immunization route in mouse model.


Assuntos
Vacina contra a Peste , Peste , Yersinia pestis , Animais , Modelos Animais de Doenças , Camundongos , Peste/prevenção & controle , Vacinas Atenuadas
9.
Vaccine ; 37(41): 6076-6084, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31477436

RESUMO

Human Q fever is recognized as a worldwide public health problem. It often occurs by inhalation of airborne aerosols contaminated with Coxiella burnetii, a gram-negative intracellular bacterium, mainly from domestic livestock. In this study, we analyzed the possibility to establish mucosal and systemic immunity against C. burnetii infection using a pulmonary delivery of chloroform-methanol residue of C. burnetii (CMR) vaccine. Mice were immunized by the intratracheal inoculation of CMR (IT-CMR) or the subcutaneous injection of CMR (SC-CMR), and the immunized mice were challenged with C. burnetii by the intratracheal route. The levels of IFN-γ, IL-12p70, IL-5, and IL-4 in the IT-CMR group in splenic T cells stimulated ex vivo were significantly higher than in the SC-CMR group. Significantly elevated sIgA to C. burnetii was detected in the bronchoalveolar lavage fluid of mice immunized by IT-CMR but not by SC-CMR, which might have contributed to the significant reduction in C. burnetii load and pathological lesions in the lungs of the mice after the challenge of C. burnetii. These results suggest that compared with SC-CMR in mice, IT-CMR was more efficient to elicit cellular and lung mucosal immune responses against aerosol infection of C. burnetii.


Assuntos
Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/imunologia , Coxiella burnetii/imunologia , Imunidade nas Mucosas/imunologia , Febre Q/prevenção & controle , Administração por Inalação , Animais , Carga Bacteriana/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Clorofórmio/química , Modelos Animais de Doenças , Feminino , Imunoglobulina A/sangue , Interferon gama/sangue , Subunidade p35 da Interleucina-12/sangue , Interleucina-4/sangue , Interleucina-5/sangue , Metanol/química , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T/imunologia , Vacinação
10.
J Clin Med ; 7(10)2018 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-30282915

RESUMO

Prodigiosin (PG) belongs to a family of prodiginines isolated from gram-negative bacteria. It is a water insoluble red pigment and a potent proapoptotic compound. This study elucidates the anti-tumor activity and underlying mechanism of PG in doxorubicin-sensitive (Dox-S) and doxorubicin-resistant (Dox-R) lung cancer cells. The cytotoxicity and cell death characteristics of PG in two cells were measured by MTT assay, cell cycle analysis, and apoptosis/autophagic marker analysis. Then, the potential mechanism of PG-induced cell death was evaluated through the phosphatidylinositol-4,5-bisphosphate 3-kinase-p85/Protein kinase B /mammalian target of rapamycin (PI3K-p85/Akt/mTOR) and Beclin-1/phosphatidylinositol-4,5-bisphosphate 3-kinase-Class III (Beclin-1/PI3K-Class III) signaling. Finally, in vivo efficacy was examined by intratracheal inoculation and treatment. There was similar cytotoxicity with PG in both Dox-S and Dox-R cells, where the half maximal inhibitory concentrations (IC50) were all in 10 µM. Based on a non-significant increase in the sub-G1 phase with an increase of microtubule-associated proteins 1A/1B light chain 3B-phosphatidylethanolamine conjugate (LC3-II), the cell death of both cells was categorized to achieve autophagy. Interestingly, an increase in cleaved-poly ADP ribose polymerase (cleaved-PARP) also showed the existence of an apoptosis-sensitive subpopulation. In both Dox-S and Dox-R cells, PI3K-p85/Akt/mTOR signaling pathways were reduced, which inhibited autophagy initiation. However, Beclin-1/PI3K-Class III downregulation implicated non-canonical autophagy pathways were involved in PG-induced autophagy. At completion of the PG regimen, tumors accumulated in the mice trachea and were attenuated by PG treatment, which indicated the efficacy of PG for both Dox-S and Dox-R lung cancer. All the above results concluded that PG is a potential chemotherapeutic agent for lung cancer regimens regardless of doxorubicin resistance.

11.
Artigo em Inglês | MEDLINE | ID: mdl-23267442

RESUMO

Pulmonary melioidosis, a disease manifestation caused by the bacterium Burkholderia pseudomallei, has been studied using aerosols or intranasal (IN) inoculation in small animal models. Both have inherent disadvantages which may not accurately model primary pulmonary melioidosis in humans. Intratracheal inoculation (IT) by direct visualization of the tracheal opening offers an alternative technique for infection that overcomes the disadvantages of aerosol and IN challenge. In this study, we describe a method which requires relatively inexpensive equipment, little training, and is compliant with the operational constraints of a BSL3 laboratory. Results obtained using trypan blue demonstrated that an inoculum can be accurately delivered into the lungs of mice within a biosafety cabinet (BSC). Whole body imaging and histopathology confirmed that mice inoculated intratracheally with B. pseudomallei develop the primary focus of infection in the lungs, and not the nasal passages which can lead to invasion of the central nervous system and potential neurologic complications. Further, based on colony counts and bioluminescent imaging, dissemination to secondary organs occurred as expected. Taken together, this intratracheal method of inoculation fulfills four goals: (1) to accurately deliver B. pseudomallei into the lungs of the animal model, (2) to avoid potentially confounding complications due to primary infections at sites other than the lung, (3) to maintain normal organ dissemination, and (4) to be BSL3 compliant.


Assuntos
Burkholderia pseudomallei/patogenicidade , Modelos Animais de Doenças , Exposição por Inalação , Melioidose/microbiologia , Pneumonia Bacteriana/microbiologia , Animais , Feminino , Histocitoquímica , Pulmão/microbiologia , Pulmão/patologia , Melioidose/patologia , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia Bacteriana/patologia , Imagem Corporal Total
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa