Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(7): e1012345, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38968329

RESUMO

The CRISPR-Cas13 system has been proposed as an alternative treatment of viral infections. However, for this approach to be adopted as an antiviral, it must be optimized until levels of efficacy rival or exceed the performance of conventional approaches. To take steps toward this goal, we evaluated the influenza viral RNA degradation patterns resulting from the binding and enzymatic activity of mRNA-encoded LbuCas13a and two crRNAs from a prior study, targeting PB2 genomic and messenger RNA. We found that the genome targeting guide has the potential for significantly higher potency than originally detected, because degradation of the genomic RNA is not uniform across the PB2 segment, but it is augmented in proximity to the Cas13 binding site. The PB2 genome targeting guide exhibited high levels (>1 log) of RNA degradation when delivered 24 hours post-infection in vitro and maintained that level of degradation over time, with increasing multiplicity of infection (MOI), and across modern influenza H1N1 and H3N2 strains. Chemical modifications to guides with potent LbuCas13a function, resulted in nebulizer delivered efficacy (>1-2 log reduction in viral titer) in a hamster model of influenza (Influenza A/H1N1/California/04/09) infection given prophylactically or as a treatment (post-infection). Maximum efficacy was achieved with two doses, when administered both pre- and post-infection. This work provides evidence that mRNA-encoded Cas13a can effectively mitigate Influenza A infections opening the door to the development of a programmable approach to treating multiple respiratory infections.

2.
Open Biol ; 14(6): 230363, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38889796

RESUMO

We present a novel small molecule antiviral chemotype that was identified by an unconventional cell-free protein synthesis and assembly-based phenotypic screen for modulation of viral capsid assembly. Activity of PAV-431, a representative compound from the series, has been validated against infectious viruses in multiple cell culture models for all six families of viruses causing most respiratory diseases in humans. In animals, this chemotype has been demonstrated efficacious for porcine epidemic diarrhoea virus (a coronavirus) and respiratory syncytial virus (a paramyxovirus). PAV-431 is shown to bind to the protein 14-3-3, a known allosteric modulator. However, it only appears to target the small subset of 14-3-3 which is present in a dynamic multi-protein complex whose components include proteins implicated in viral life cycles and in innate immunity. The composition of this target multi-protein complex appears to be modified upon viral infection and largely restored by PAV-431 treatment. An advanced analog, PAV-104, is shown to be selective for the virally modified target, thereby avoiding host toxicity. Our findings suggest a new paradigm for understanding, and drugging, the host-virus interface, which leads to a new clinical therapeutic strategy for treatment of respiratory viral disease.


Assuntos
Antivirais , Antivirais/farmacologia , Antivirais/química , Humanos , Animais , Proteínas 14-3-3/metabolismo , Complexos Multiproteicos/metabolismo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Linhagem Celular
3.
Cell Stem Cell ; 31(5): 734-753.e8, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38608707

RESUMO

Autonomic parasympathetic neurons (parasymNs) control unconscious body responses, including "rest-and-digest." ParasymN innervation is important for organ development, and parasymN dysfunction is a hallmark of autonomic neuropathy. However, parasymN function and dysfunction in humans are vastly understudied due to the lack of a model system. Human pluripotent stem cell (hPSC)-derived neurons can fill this void as a versatile platform. Here, we developed a differentiation paradigm detailing the derivation of functional human parasymNs from Schwann cell progenitors. We employ these neurons (1) to assess human autonomic nervous system (ANS) development, (2) to model neuropathy in the genetic disorder familial dysautonomia (FD), (3) to show parasymN dysfunction during SARS-CoV-2 infection, (4) to model the autoimmune disease Sjögren's syndrome (SS), and (5) to show that parasymNs innervate white adipocytes (WATs) during development and promote WAT maturation. Our model system could become instrumental for future disease modeling and drug discovery studies, as well as for human developmental studies.


Assuntos
Diferenciação Celular , Disautonomia Familiar , Células-Tronco Pluripotentes , Humanos , Células-Tronco Pluripotentes/citologia , Disautonomia Familiar/patologia , Neurônios , Síndrome de Sjogren/patologia , COVID-19/virologia , COVID-19/patologia , Animais , Sistema Nervoso Parassimpático , Células de Schwann , Camundongos , SARS-CoV-2/fisiologia
4.
Viruses ; 16(1)2024 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-38275962

RESUMO

Avian influenza (AI) viruses cause infection in birds and humans. Several H5N1 and H7N9 variants are highly pathogenic avian influenza (HPAI) viruses. H5N1 is a highly infectious bird virus infecting primarily poultry, but unlike other AIs, H5N1 also infects mammals and transmits to humans with a case fatality rate above 40%. Similarly, H7N9 can infect humans, with a case fatality rate of over 40%. Since 1996, there have been several HPAI outbreaks affecting humans, emphasizing the need for safe and effective antivirals. We show that probenecid potently inhibits H5N1 and H7N9 replication in prophylactically or therapeutically treated A549 cells and normal human broncho-epithelial (NHBE) cells, and H5N1 replication in VeroE6 cells and mice.


Assuntos
Virus da Influenza A Subtipo H5N1 , Subtipo H7N9 do Vírus da Influenza A , Influenza Aviária , Influenza Humana , Animais , Humanos , Camundongos , Influenza Aviária/tratamento farmacológico , Influenza Aviária/prevenção & controle , Influenza Aviária/epidemiologia , Subtipo H7N9 do Vírus da Influenza A/genética , Probenecid , Aves , Mamíferos
5.
Nat Mater ; 22(3): 369-379, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36443576

RESUMO

Messenger RNA has now been used to vaccinate millions of people. However, the diversity of pulmonary pathologies, including infections, genetic disorders, asthma and others, reveals the lung as an important organ to directly target for future RNA therapeutics and preventatives. Here we report the screening of 166 polymeric nanoparticle formulations for functional delivery to the lungs, obtained from a combinatorial synthesis approach combined with a low-dead-volume nose-only inhalation system for mice. We identify P76, a poly-ß-amino-thio-ester polymer, that exhibits increased expression over formulations lacking the thiol component, delivery to different animal species with varying RNA cargos and low toxicity. P76 allows for dose sparing when delivering an mRNA-expressed Cas13a-mediated treatment in a SARS-CoV-2 challenge model, resulting in similar efficacy to a 20-fold higher dose of a neutralizing antibody. Overall, the combinatorial synthesis approach allowed for the discovery of promising polymeric formulations for future RNA pharmaceutical development for the lungs.


Assuntos
COVID-19 , Animais , Camundongos , RNA Mensageiro/genética , SARS-CoV-2/genética , Polímeros/metabolismo , Pulmão , RNA/metabolismo
6.
Adv Sci (Weinh) ; 9(34): e2202771, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36316224

RESUMO

Despite the success of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccines, there remains a clear need for new classes of preventatives for respiratory viral infections due to vaccine hesitancy, lack of sterilizing immunity, and for at-risk patient populations, including the immunocompromised. While many neutralizing antibodies have been identified, and several approved, to treat COVID-19, systemic delivery, large doses, and high costs have the potential to limit their widespread use, especially in low- and middle-income countries. To use these antibodies more efficiently, an inhalable formulation is developed that allows for the expression of mRNA-encoded, membrane-anchored neutralizing antibodies in the lung to mitigate SARS-CoV-2 infections. First, the ability of mRNA-encoded, membrane-anchored, anti-SARS-CoV-2 antibodies to prevent infections in vitro is demonstrated. Next, it is demonstrated that nebulizer-based delivery of these mRNA-expressed neutralizing antibodies potently abrogates disease in the hamster model. Overall, these results support the use of nebulizer-based mRNA expression of neutralizing antibodies as a new paradigm for mitigating respiratory virus infections.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , RNA Mensageiro/genética , Anticorpos Neutralizantes/uso terapêutico
7.
Food Microbiol ; 107: 104084, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35953178

RESUMO

The potential transmission of SARS-CoV-2 via food has been controversial since the beginning of the COVID-19 pandemic. To investigate these concerns, reliable detection methods and data on virus die-off rates in various foods are needed. Here, an FDA-standard method for the detection of enteric viruses' RNA from soft fruits was modified for the recovery of infectious SARS-CoV-2. Then, the survival of SARS-CoV-2 on berries was investigated as well as the effectiveness of washing virus-contaminated berries with water. The modified method did not significantly reduced log infectivity titers of recovered viruses, but berries did. The detection limit of the method for infectious SARS-CoV-2 was ∼2.97 log TCID50/g of berries. On SARS-CoV-2-inoculated berries that were stored at 4 °C for 7 days, significant reductions in SARS-CoV-2 infectivity were observed over time. In contrast, on frozen berries, infectious SARS-CoV-2 was recovered for 28 days without significant reductions. Washing SARS-CoV-2-inoculated berries with water removed >90% of infectious viruses within 10 min; however, infectious viruses were detected in wash water. Therefore, on fresh berries infectious viruses are markedly inactivated over time and can be largely removed by washing with water. However, the prolonged survival of SARS-CoV-2 on frozen berries suggests that the virus can potentially spread through frozen fruits.


Assuntos
COVID-19 , Vírus , Frutas , Humanos , Pandemias , SARS-CoV-2/genética , Água
8.
bioRxiv ; 2022 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-34931190

RESUMO

We present a small molecule chemotype, identified by an orthogonal drug screen, exhibiting nanomolar activity against members of all the six viral families causing most human respiratory viral disease, with a demonstrated barrier to resistance development. Antiviral activity is shown in mammalian cells, including human primary bronchial epithelial cells cultured to an air-liquid interface and infected with SARS-CoV-2. In animals, efficacy of early compounds in the lead series is shown by survival (for a coronavirus) and viral load (for a paramyxovirus). The drug target is shown to include a subset of the protein 14-3-3 within a transient host multi-protein complex containing components implicated in viral lifecycles and in innate immunity. This multi-protein complex is modified upon viral infection and largely restored by drug treatment. Our findings suggest a new clinical therapeutic strategy for early treatment upon upper respiratory viral infection to prevent progression to lower respiratory tract or systemic disease. One Sentence Summary: A host-targeted drug to treat all respiratory viruses without viral resistance development.

9.
Sci Rep ; 11(1): 18085, 2021 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34508172

RESUMO

Effective vaccines are slowing the COVID-19 pandemic, but SARS-CoV-2 will likely remain an issue in the future making it important to have therapeutics to treat patients. There are few options for treating patients with COVID-19. We show probenecid potently blocks SARS-CoV-2 replication in mammalian cells and virus replication in a hamster model. Furthermore, we demonstrate that plasma concentrations up to 50-fold higher than the protein binding adjusted IC90 value are achievable for 24 h following a single oral dose. These data support the potential clinical utility of probenecid to control SARS-CoV-2 infection in humans.


Assuntos
Antivirais/farmacologia , Células Epiteliais/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Probenecid/farmacologia , SARS-CoV-2/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Chlorocebus aethiops , Células Epiteliais/virologia , Humanos , Pulmão/virologia , Células Vero
10.
Nat Biotechnol ; 39(6): 717-726, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33536629

RESUMO

Cas13a has been used to target RNA viruses in cell culture, but efficacy has not been demonstrated in animal models. In this study, we used messenger RNA (mRNA)-encoded Cas13a for mitigating influenza virus A and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in mice and hamsters, respectively. We designed CRISPR RNAs (crRNAs) specific for PB1 and highly conserved regions of PB2 of influenza virus, and against the replicase and nucleocapsid genes of SARS-CoV-2, and selected the crRNAs that reduced viral RNA levels most efficiently in cell culture. We delivered polymer-formulated Cas13a mRNA and the validated guides to the respiratory tract using a nebulizer. In mice, Cas13a degraded influenza RNA in lung tissue efficiently when delivered after infection, whereas in hamsters, Cas13a delivery reduced SARS-CoV-2 replication and reduced symptoms. Our findings suggest that Cas13a-mediated targeting of pathogenic viruses can mitigate respiratory infections.


Assuntos
COVID-19/terapia , Influenza Humana/terapia , RNA Mensageiro/farmacologia , SARS-CoV-2/genética , Animais , COVID-19/genética , COVID-19/virologia , Sistemas CRISPR-Cas/genética , Cricetinae , Modelos Animais de Doenças , Humanos , Influenza Humana/genética , Influenza Humana/virologia , Camundongos , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/genética , Orthomyxoviridae/patogenicidade , RNA Mensageiro/genética , RNA Viral/genética , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/metabolismo , SARS-CoV-2/patogenicidade
11.
J Med Chem ; 63(21): 12725-12747, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-33054210

RESUMO

The novel coronavirus disease COVID-19 that emerged in 2019 is caused by the virus SARS CoV-2 and named for its close genetic similarity to SARS CoV-1 that caused severe acute respiratory syndrome (SARS) in 2002. Both SARS coronavirus genomes encode two overlapping large polyproteins, which are cleaved at specific sites by a 3C-like cysteine protease (3CLpro) in a post-translational processing step that is critical for coronavirus replication. The 3CLpro sequences for CoV-1 and CoV-2 viruses are 100% identical in the catalytic domain that carries out protein cleavage. A research effort that focused on the discovery of reversible and irreversible ketone-based inhibitors of SARS CoV-1 3CLpro employing ligand-protease structures solved by X-ray crystallography led to the identification of 3 and 4. Preclinical experiments reveal 4 (PF-00835231) as a potent inhibitor of CoV-2 3CLpro with suitable pharmaceutical properties to warrant further development as an intravenous treatment for COVID-19.


Assuntos
Antivirais/farmacologia , Proteases 3C de Coronavírus/antagonistas & inibidores , Cetonas/farmacologia , Inibidores de Proteases/farmacologia , SARS-CoV-2/efeitos dos fármacos , Animais , Antivirais/síntese química , Antivirais/metabolismo , Domínio Catalítico , Chlorocebus aethiops , Proteases 3C de Coronavírus/química , Proteases 3C de Coronavírus/metabolismo , Cristalografia por Raios X , Humanos , Cetonas/síntese química , Cetonas/metabolismo , Inibidores de Proteases/síntese química , Inibidores de Proteases/metabolismo , Ligação Proteica , Células Vero , Tratamento Farmacológico da COVID-19
12.
Virulence ; 11(1): 1024-1040, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32799724

RESUMO

BURKHOLDERIA MALLEI: is a highly pathogenic bacterium that causes the fatal zoonosis glanders. The organism specifies multiple membrane proteins, which represent prime targets for the development of countermeasures given their location at the host-pathogen interface. We investigated one of these proteins, Pal, and discovered that it is involved in the ability of B. mallei to resist complement-mediated killing and replicate inside host cells in vitro, is expressed in vivo and induces antibodies during the course of infection, and contributes to virulence in a mouse model of aerosol infection. A mutant in the pal gene of the B. mallei wild-type strain ATCC 23344 was found to be especially attenuated, as BALB/c mice challenged with the equivalent of 5,350 LD50 completely cleared infection. Based on these findings, we tested the hypothesis that a vaccine containing the Pal protein elicits protective immunity against aerosol challenge. To achieve this, the pal gene was cloned in the vaccine vector Parainfluenza Virus 5 (PIV5) and mice immunized with the virus were infected with a lethal dose of B. mallei. These experiments revealed that a single dose of PIV5 expressing Pal provided 80% survival over a period of 40 days post-challenge. In contrast, only 10% of mice vaccinated with a PIV5 control virus construct survived infection. Taken together, our data establish that the Peptidoglycan-associated lipoprotein Pal is a critical virulence determinant of B. mallei and effective target for developing a glanders vaccine.


Assuntos
Vacinas Bacterianas/imunologia , Burkholderia mallei/química , Burkholderia mallei/patogenicidade , Lipoproteínas/imunologia , Melioidose/prevenção & controle , Peptidoglicano/química , Aerossóis , Animais , Vacinas Bacterianas/administração & dosagem , Burkholderia mallei/imunologia , Linhagem Celular , Feminino , Vetores Genéticos , Imunização , Lipoproteínas/administração & dosagem , Macrófagos/microbiologia , Melioidose/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Parainfluenza 5/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Virulência
13.
BMC Med Genomics ; 12(1): 127, 2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31492148

RESUMO

BACKGROUND: Burkholderia mallei (Bm) is a facultative intracellular bacterial pathogen causing highly-fatal glanders in solipeds and humans. The ability of Bm to thrive intracellularly is thought to be related to exploitation of host immune response-related genes and pathways. Relatively little is known of the molecular strategies employed by this pathogen to modulate these pathways and evade intracellular killing. This manuscript seeks to fill gaps in the understanding of the interface between Bm and innate immunity by examining gene expression changes during infection of host monocytes. METHODS: The transcriptome of Bm-infected human Mono Mac-6 (MM6) monocytes was profiled on Affymetrix Human Transcriptome GeneChips 2.0. Gene expression changes in Bm-infected monocytes were compared to those of Burkholderia thailandensis (Bt)-infected monocytes and to uninfected monocytes. The resulting dataset was normalized using Robust Multichip Average and subjected to statistical analyses employing a univariate F test with a random variance model. Differentially expressed genes significant at p < 0.001 were subjected to leave-one-out cross-validation studies and 1st and 3rd nearest neighbor prediction model. Significant probe sets were used to populate human pathways in Ingenuity Pathway Analysis, with statistical significance determined by Fisher's exact test or z-score. RESULTS: The Pattern Recognition Receptor (PRR) pathway was represented among significantly enriched immune response-related human canonical pathways, with evidence of upregulation across both infections. Among members of this pathway, pentraxin-3 was significantly upregulated by Bm- or Bt-infected monocytes. Pentraxin-3 (PTX3) was demonstrated to bind to both Bt and Burkholderia pseudomallei (Bp), but not Bm. Subsequent assays did not identify a role for PTX3 in potentiating complement-mediated lysis of Bt or in enhancing phagocytosis or replication of Bt in human monocytes. CONCLUSION: We report on the novel binding of PTX3 to Bt and Bp, with lack of interaction with Bm, suggesting that a possible evasive mechanism by Bm warrants further exploration. We determined that (1) PTX3 may not play a role in activating the lytic pathway of complement in different bacterial species and that (2) the opsonophagocytic properties of PTX3 should be investigated in different primary or immortalized cell lines representing host phagocytes, given lack of binding of PTX3 to MM6 monocytes.


Assuntos
Burkholderia/imunologia , Proteína C-Reativa/metabolismo , Perfilação da Expressão Gênica , Imunidade Inata , Monócitos/imunologia , Monócitos/microbiologia , Componente Amiloide P Sérico/metabolismo , Anticorpos/metabolismo , Burkholderia/crescimento & desenvolvimento , Linhagem Celular , Proteínas do Sistema Complemento/metabolismo , Humanos , Imunidade Inata/genética , Viabilidade Microbiana , Proteínas Opsonizantes/metabolismo , Fagocitose , Ligação Proteica , Regulação para Cima/genética
14.
Int J Paleopathol ; 24: 266-278, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30743216

RESUMO

OBJECTIVE: Canine distemper virus (CDV), human measles virus (HMV), and rinderpest virus (RPV) of cattle are morbilliviruses that have caused devastating outbreaks for centuries. This paper seeks to reconstruct the evolutionary history of CDV. MATERIALS AND METHODS: An interdisciplinary approach is adopted, synthesizing paleopathological analysis of 96 Pre-Columbian dogs (750-1470 CE) from the Weyanoke Old Town, Virginia site, with historical reports, molecular analysis and morbilliviral epidemiology. RESULTS: Both measles (c.900CE) and rinderpest (c. 376 BCE) were first reported in Eurasia, while canine distemper was initially described in South America much later (1735 CE); there are no paleopathological indications of CDV in Weyanoke Old Town dogs. Molecularly, CDV is closely related to HMV, while viral codon usage indicates CDV may have previously infected humans; South American measles epidemics occurred prior to the emergence of canine distemper and would have facilitated HMV transmission and adaptation to dogs. CONCLUSIONS: The measles epidemics that decimated indigenous South American populations in the 1500-1700 s likely facilitated the establishment of CDV as a canine pathogen, which eventually spread to Europe and beyond. SIGNIFICANCE: Understanding the historical and environmental conditions that have driven morbilliviral evolution provides important insights into potential future threats of animal/human cross-species infections. LIMITATIONS: Interpreting historical disease descriptions is difficult and the archaeological specimens are limited. Molecular sequence data and codon usage analyses rely on modern viruses. SUGGESTIONS FOR FURTHER RESEARCH: Interdisciplinary approaches are increasingly needed to understand diseases of the past and present, as critical information and knowledge is scattered in different disciplines.


Assuntos
Vírus da Cinomose Canina/genética , Cinomose/epidemiologia , Morbillivirus/genética , Animais , Uso do Códon , Cinomose/história , Cinomose/patologia , Cinomose/virologia , Cães , Europa (Continente)/epidemiologia , História do Século XVI , História do Século XVII , História do Século XVIII , História Antiga , Humanos , Pesquisa Interdisciplinar , Vírus do Sarampo/genética , Paleopatologia , Filogenia , Vírus da Peste Bovina/genética , América do Sul/epidemiologia , Virginia/epidemiologia
15.
Sci Rep ; 9(1): 366, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30675003

RESUMO

To address the unmet needs for human polyclonal antibodies both as therapeutics and diagnostic reagents, building upon our previously established transchromosomic (Tc) cattle platform, we report herein the development of a Tc goat system expressing human polyclonal antibodies in their sera. In the Tc goat system, a human artificial chromosome (HAC) comprising the entire human immunoglobulin (Ig) gene repertoire in the germline configuration was introduced into the genetic makeup of the domestic goat. We achieved this by transferring the HAC into goat fetal fibroblast cells followed by somatic cell nuclear transfer for Tc goat production. Gene and protein expression analyses in the peripheral blood mononuclear cells (PBMC) and the sera, respectively, of Tc caprine demonstrated the successful expression of human Ig genes and antibodies. Furthermore, immunization of Tc caprine with inactivated influenza A (H7N9) viruses followed by H7N9 Hemagglutinin 1 (HA1) boosting elicited human antibodies with high neutralizing activities against H7N9 viruses in vitro. As a small ungulate, Tc caprine offers the advantages of low cost and quick establishment of herds, therefore complementing the Tc cattle platform in responses to a range of medical needs and diagnostic applications where small volumes of human antibody products are needed.


Assuntos
Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Subtipo H7N9 do Vírus da Influenza A/imunologia , Animais , Animais Geneticamente Modificados , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Cromossomos Artificiais Humanos , Ensaio de Imunoadsorção Enzimática , Engenharia Genética , Cabras , Humanos , Imunização , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Testes de Neutralização
16.
mSphere ; 4(1)2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30602525

RESUMO

Burkholderia pseudomallei and Burkholderia mallei are the causative agents of melioidosis and glanders, respectively. There is no vaccine to protect against these highly pathogenic bacteria, and there is concern regarding their emergence as global public health (B. pseudomallei) and biosecurity (B. mallei) threats. In this issue of mSphere, an article by Khakhum and colleagues (N. Khakhum, P. Bharaj, J. N. Myers, D. Tapia, et al., mSphere 4:e00570-18, 2019, https://doi.org/10.1128/mSphere.00570-18) describes a novel vaccination platform with excellent potential for cross-protection against both Burkholderia species. The report also highlights the importance of antibodies in immunity against these facultative intracellular organisms.


Assuntos
Burkholderia pseudomallei , Mormo , Aerossóis , Animais , Burkholderia , Burkholderia mallei , Cavalos
17.
Vaccine X ; 1: 100002, 2019 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-33826684

RESUMO

BACKGROUND: Burkholderia mallei and Burkholderia pseudomallei are the causative agents of glanders and melioidosis, respectively. There is no vaccine to protect against these highly-pathogenic and intrinsically antibiotic-resistant bacteria, and there is concern regarding their use as biological warfare agents. For these reasons, B. mallei and B. pseudomallei are classified as Tier 1 organisms by the U.S. Federal Select Agent Program and the availability of effective countermeasures represents a critical unmet need. METHODS: Vaccines (subunit and vectored) containing the surface-exposed passenger domain of the conserved Burkholderia autotransporter protein BatA were administered to BALB/c mice and the vaccinated animals were challenged with lethal doses of wild-type B. mallei and B. pseudomallei strains via the aerosol route. Mice were monitored for signs of illness for a period of up to 40 days post-challenge and tissues from surviving animals were analyzed for bacterial burden at study end-points. RESULTS: A single dose of recombinant Parainfluenza Virus 5 (PIV5) expressing BatA provided 74% and 60% survival in mice infected with B. mallei and B. pseudomallei, respectively. Vaccination with PIV5-BatA also resulted in complete bacterial clearance from the lungs and spleen of 78% and 44% of animals surviving lethal challenge with B. pseudomallei, respectively. In contrast, all control animals vaccinated with a PIV5 construct expressing an irrelevant antigen and infected with B. pseudomallei were colonized in those tissues. CONCLUSION: Our study indicates that the autotransporter BatA is a valuable target for developing countermeasures against B. mallei and B. pseudomallei and demonstrates the utility of the PIV5 viral vaccine delivery platform to elicit cross-protective immunity against the organisms.

18.
Vet Pathol ; 55(2): 258-267, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29145795

RESUMO

Burkholderia mallei causes the highly contagious and debilitating zoonosis glanders, which infects via inhalation or percutaneous inoculation and often culminates in life-threatening pneumonia and sepsis. In humans, glanders is difficult to diagnose and requires prolonged antibiotic therapy with low success rates. No vaccine exists to protect against B. mallei, and there is concern regarding its use as a bioweapon. The authors previously identified the protein BpaB as a potential target for devising therapies due to its role in adherence to host cells and the formation of biofilms in vitro and its contribution to pathogenicity in a mouse model of glanders. In the present study, the authors developed an immunostaining approach to probe tissues of experimentally infected animals and demonstrated that BpaB is produced exclusively in vivo by wild-type B. mallei in target organs from mice and marmosets. They detected the expression of BpaB by B. mallei both extracellularly and within macrophages, neutrophils, and epithelial cells in respiratory tissues (7/10 marmoset; 2/2 mouse). The authors also noted the intracellular expression of BpaB by B. mallei in macrophages in the regional lymph nodes of mice (2/2 tissues) and MALT of marmosets (4/5 tissues). It is interesting that B. mallei bacteria infecting distal organs did not express BpaB (2/2 mice; 3/3 marmosets), suggesting that the protein is not necessary for bacterial fitness in these anatomic locations. These findings underscore the value of BpaB as a target for developing medical countermeasures and provide insight into its role in pathogenesis.


Assuntos
Burkholderia mallei/patogenicidade , Mormo/microbiologia , Fatores de Virulência/metabolismo , Animais , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/imunologia , Burkholderia mallei/imunologia , Burkholderia mallei/metabolismo , Callithrix/microbiologia , Mormo/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Fatores de Virulência/imunologia
19.
Infect Immun ; 85(8)2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28507073

RESUMO

Burkholderia mallei, a facultative intracellular bacterium and tier 1 biothreat, causes the fatal zoonotic disease glanders. The organism possesses multiple genes encoding autotransporter proteins, which represent important virulence factors and targets for developing countermeasures in pathogenic Gram-negative bacteria. In the present study, we investigated one of these autotransporters, BatA, and demonstrate that it displays lipolytic activity, aids in intracellular survival, is expressed in vivo, elicits production of antibodies during infection, and contributes to pathogenicity in a mouse aerosol challenge model. A mutation in the batA gene of wild-type strain ATCC 23344 was found to be particularly attenuating, as BALB/c mice infected with the equivalent of 80 median lethal doses cleared the organism. This finding prompted us to test the hypothesis that vaccination with the batA mutant strain elicits protective immunity against subsequent infection with wild-type bacteria. We discovered that not only does vaccination provide high levels of protection against lethal aerosol challenge with B. mallei ATCC 23344, it also protects against infection with multiple isolates of the closely related organism and causative agent of melioidosis, Burkholderia pseudomallei Passive-transfer experiments also revealed that the protective immunity afforded by vaccination with the batA mutant strain is predominantly mediated by IgG antibodies binding to antigens expressed exclusively in vivo Collectively, our data demonstrate that BatA is a target for developing medical countermeasures and that vaccination with a mutant lacking expression of the protein provides a platform to gain insights regarding mechanisms of protective immunity against B. mallei and B. pseudomallei, including antigen discovery.


Assuntos
Anticorpos Antibacterianos/imunologia , Burkholderia mallei/imunologia , Burkholderia pseudomallei/imunologia , Melioidose/prevenção & controle , Animais , Proteínas de Bactérias/genética , Burkholderia mallei/genética , Burkholderia mallei/crescimento & desenvolvimento , Burkholderia mallei/patogenicidade , Burkholderia pseudomallei/patogenicidade , Modelos Animais de Doenças , Mormo/imunologia , Mormo/microbiologia , Mormo/prevenção & controle , Imunoglobulina G/imunologia , Melioidose/imunologia , Melioidose/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Vacinação , Fatores de Virulência/genética
20.
Expert Rev Vaccines ; 15(9): 1163-81, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27010618

RESUMO

Burkholderia pseudomallei and Burkholderia mallei are pathogenic bacteria causing fatal infections in animals and humans. Both organisms are classified as Tier 1 Select Agents owing to their highly fatal nature, potential/prior use as bioweapons, severity of disease via respiratory exposure, intrinsic resistance to antibiotics, and lack of a current vaccine. Disease manifestations range from acute septicemia to chronic infection, wherein the facultative intracellular lifestyle of these organisms promotes persistence within a broad range of hosts. This ability to thrive intracellularly is thought to be related to exploitation of host immune response signaling pathways. There are currently considerable gaps in our understanding of the molecular strategies employed by these pathogens to modulate these pathways and evade intracellular killing. A better understanding of the specific molecular basis for dysregulation of host immune responses by these organisms will provide a stronger platform to identify novel vaccine targets and develop effective countermeasures.


Assuntos
Vacinas Bacterianas/isolamento & purificação , Burkholderia mallei/patogenicidade , Burkholderia pseudomallei/patogenicidade , Mormo/imunologia , Mormo/prevenção & controle , Melioidose/imunologia , Melioidose/prevenção & controle , Animais , Vacinas Bacterianas/imunologia , Burkholderia mallei/imunologia , Burkholderia pseudomallei/imunologia , Mormo/patologia , Humanos , Evasão da Resposta Imune , Melioidose/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...