Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
PLoS One ; 10(4): e0124181, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25860021

RESUMO

Burkholderia mallei is a host-adapted bacterium that does not persist outside of its equine reservoir. The organism causes the zoonosis glanders, which is endemic in Asia, Africa, the Middle East and South America. Infection by B. mallei typically occurs via the respiratory or percutaneous route, and the most common manifestations are life-threatening pneumonia and bacteremia. Glanders is difficult to diagnose and requires prolonged antibiotic therapy with low success rates. There is no vaccine to protect against B. mallei and there is concern regarding its use as a biothreat agent. Thus, experiments were performed to establish a non-human primate model of intranasal infection to study the organism and develop countermeasures. Groups of marmosets (Callithrix jacchus) were inoculated intranasally with B. mallei strain ATCC 23344 and monitored for clinical signs of illness for up to 13 days. We discovered that 83% of marmosets inoculated with doses of 2.5 X 10(4) to 2.5 X 10(5) bacteria developed acute lethal infection within 3-4 days. Signs of disease were severe and included lethargy, inappetence, conjunctivitis, mucopurulent and hemorrhagic nasal discharges, and increased respiratory effort with abdominal lifts. Burkholderia mallei was cultured from the lungs, spleen and liver of these animals, and pathologic examination of tissues revealed lesions characteristic of glanders. Challenge experiments also revealed that 91% of animals infected with doses ranging from 25 to 2.5 X 10(3) bacteria exhibited mild non-specific signs of illness and were culture negative. One marmoset inoculated with 2.5 X 10(3) organisms developed moderate signs of disease and reached humane end-points 8 days post-infection. The liver and spleen of this animal were colonized with the agent and pathological analysis of tissues showed nasal, splenic and hepatic lesions. Taken together, these data indicate that the marmoset is a suitable model to study respiratory infection by B. mallei.


Assuntos
Burkholderia mallei/patogenicidade , Callithrix/microbiologia , Mormo/etiologia , Administração Intranasal , Animais , Carga Bacteriana , Modelos Animais de Doenças , Feminino , Mormo/patologia , Mormo/transmissão , Cavalos , Humanos , Fígado/microbiologia , Fígado/patologia , Pulmão/microbiologia , Pulmão/patologia , Masculino , Especificidade da Espécie , Baço/microbiologia , Baço/patologia , Zoonoses/etiologia , Zoonoses/patologia , Zoonoses/transmissão
3.
J Neurosci ; 34(12): 4260-72, 2014 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-24647946

RESUMO

Recent findings suggest that tau oligomers, which form before neurofibrillary tangles (NFTs), are the true neurotoxic tau entities in neurodegenerative tauopathies, including Alzheimer's disease (AD). Studies in animal models of tauopathy suggest that tau oligomers play a key role in eliciting behavioral and cognitive impairments. Here, we used a novel tau oligomer-specific monoclonal antibody (TOMA) for passive immunization in mice expressing mutant human tau. A single dose of TOMA administered either intravenously or intracerebroventricularly was sufficient to reverse both locomotor and memory deficits in a mouse model of tauopathy for 60 d, coincident with rapid reduction of tau oligomers but not phosphorylated NFTs or monomeric tau. Our data demonstrate that antibody protection is mediated by extracellular and rapid peripheral clearance. These findings provide the first direct evidence in support of a critical role for tau oligomers in disease progression and validate tau oligomers as a target for the treatment of AD and other neurodegenerative tauopathies.


Assuntos
Doença de Alzheimer/terapia , Imunização Passiva , Emaranhados Neurofibrilares/imunologia , Tauopatias/terapia , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Animais , Modelos Animais de Doenças , Progressão da Doença , Camundongos , Emaranhados Neurofibrilares/genética , Emaranhados Neurofibrilares/metabolismo , Fosforilação , Tauopatias/genética , Tauopatias/imunologia , Tauopatias/metabolismo , Proteínas tau/genética
4.
Pathogens ; 2(2)2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24349761

RESUMO

Burkholderia mallei, the etiologic agent of glanders, are Category B select agents with biothreat potential, and yet effective therapeutic treatments are lacking. In this study, we showed that CpG administration increased survival, demonstrating protection in the murine glanders model. Bacterial recovery from infected lungs, liver and spleen was significantly reduced in CpG-treated animals as compared with non-treated mice. Reciprocally, lungs of CpG-treated infected animals were infiltrated with higher levels of neutrophils and inflammatory monocytes, as compared to control animals. Employing the B. mallei bioluminescent strain CSM001 and the Neutrophil-Specific Fluorescent Imaging Agent, bacterial dissemination and neutrophil trafficking were monitored in real-time using multimodal in vivo whole body imaging techniques. CpG-treatment increased recruitment of neutrophils to the lungs and reduced bioluminescent bacteria, correlating with decreased bacterial burden and increased protection against acute murine glanders. Our results indicate that protection of CpG-treated animals was associated with recruitment of neutrophils prior to infection and demonstrated, for the first time, simultaneous real time in vivo imaging of neutrophils and bacteria. This study provides experimental evidence supporting the importance of incorporating optimized in vivo imaging methods to monitor disease progression and to evaluate the efficacy of therapeutic treatment during bacterial infections.

5.
Virus Res ; 176(1-2): 61-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23769974

RESUMO

Human adenovirus type 41 (HAdV-41) has the potential to be constructed as a gene transfer vector for oral vaccine or gene therapy targeting gastrointestinal tract. Block in release of progeny virus from host cell severely affects the yield during virus amplification. In this study, HAdV-5 adenovirus death protein (ADP) gene was used to replace the open reading frames (ORFs) of the HAdV-41 E3 region to construct a backbone plasmid pAdbone41ADP. Recombinant adenoviral plasmids harboring ADP and GFP genes (pAd41ADP-GFP) were generated. Plaques were formed and HAdV-41-ADP-GFP virus was rescued after transfecting pAd41ADP-GFP into the packaging cell line 293TE32. When amplified on 293TE32 cells, HAdV-41-ADP-GFP virus released to the culture medium was 10-50 times more than control virus HAdV-41-GFP, which did not carry ADP gene. The results demonstrated that incorporation of the ADP gene substantially increased the yield of recombinant HAdV-41 virus through enhancing spread of progeny virus among packaging cells.


Assuntos
Proteínas E3 de Adenovirus/metabolismo , Adenovírus Humanos/crescimento & desenvolvimento , Proteínas E3 de Adenovirus/genética , Adenovírus Humanos/genética , Linhagem Celular , Humanos , Plasmídeos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção , Carga Viral , Ensaio de Placa Viral
6.
PLoS One ; 8(5): e63331, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23691024

RESUMO

Mycobacterium tuberculosis (M.tb) is the second leading infectious cause of death worldwide and the primary cause of death in people living with HIV/AIDS. There are several excellent animal models employed to study tuberculosis (TB), but many have limitations for reproducing human pathology and none are amenable to the direct study of HIV/M.tb co-infection. The humanized mouse has been increasingly employed to explore HIV infection and other pathogens where animal models are limiting. Our goal was to develop a small animal model of M.tb infection using the bone marrow, liver, thymus (BLT) humanized mouse. NOD-SCID/γc(null) mice were engrafted with human fetal liver and thymus tissue, and supplemented with CD34(+) fetal liver cells. Excellent reconstitution, as measured by expression of the human CD45 pan leukocyte marker by peripheral blood populations, was observed at 12 weeks after engraftment. Human T cells (CD3, CD4, CD8), as well as natural killer cells and monocyte/macrophages were all observed within the human leukocyte (CD45(+)) population. Importantly, human T cells were functionally competent as determined by proliferative capacity and effector molecule (e.g. IFN-γ, granulysin, perforin) expression in response to positive stimuli. Animals infected intranasally with M.tb had progressive bacterial infection in the lung and dissemination to spleen and liver from 2-8 weeks post infection. Sites of infection in the lung were characterized by the formation of organized granulomatous lesions, caseous necrosis, bronchial obstruction, and crystallization of cholesterol deposits. Human T cells were distributed throughout the lung, liver, and spleen at sites of inflammation and bacterial growth and were organized to the periphery of granulomas. These preliminary results demonstrate the potential to use the humanized mouse as a model of experimental TB.


Assuntos
Modelos Animais de Doenças , Tuberculose/fisiopatologia , Animais , Transplante de Medula Óssea/métodos , Humanos , Fígado/citologia , Fígado/patologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Linfócitos T/imunologia , Timo/citologia
7.
Vet Microbiol ; 163(3-4): 223-34, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23428380

RESUMO

Venezuelan equine encephalitis virus (VEEV) is an important, naturally emerging zoonotic pathogen. Recent outbreaks in Venezuela and Colombia in 1995 indicate that VEEV still poses a serious public health threat. Astrocytes may be target cells in human and mouse infection and they play an important role in repair through gliosis. In this study, we report that virulent VEEV efficiently infects cultured normal human astrocytes, three different murine astrocyte cell lines and astrocytes in the mouse brain. The attenuation of virus replication positively correlates with the increased levels of production of IL-8, IL-17, IFN-gamma and IP-10. In addition, VEEV infection induces release of basic fibroblast growth factor and production of potent chemokines such as RANTES and MIP-1-beta from cultured human astrocytes. This growth factor and cytokine profile modeled by astrocytes in vitro may contribute to both neuroprotection and repair and may play a role in leukocyte recruitment in vivo.


Assuntos
Astrócitos/imunologia , Astrócitos/virologia , Citocinas/imunologia , Vírus da Encefalite Equina Venezuelana/fisiologia , Encefalomielite Equina Venezuelana/imunologia , Zoonoses/virologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Núcleo Celular/virologia , Células Cultivadas , Quimiocinas/imunologia , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/virologia , Fatores de Crescimento de Fibroblastos/imunologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Camundongos , Nucleocapsídeo/metabolismo
8.
Vaccine ; 30(27): 4095-105, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22525795

RESUMO

TC83 is a human vaccine with investigational new drug status and is used as a prototype Venezuelan equine encephalitis virus for pathogenesis and antiviral research. Differing from other experimental models, the virus causes high titer infection in the brain and 90-100% mortality in the C3H/HeN murine model. To better characterize the susceptibility to disease development in C3H/HeN mice, we have analyzed the gene transcriptomes and cytokine production in the brains of infected mice. Our analysis indicated the potential importance of natural killer cells in the encephalitic disease development. This paper describes for the first time a pathogenic role for natural killer cells in VEEV encephalitis.


Assuntos
Vírus da Encefalite Equina Venezuelana/imunologia , Vírus da Encefalite Equina Venezuelana/patogenicidade , Encefalomielite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/patologia , Células Matadoras Naturais/imunologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite Equina Venezuelana/mortalidade , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C3H , Análise de Sobrevida
9.
PLoS One ; 7(3): e34176, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22448290

RESUMO

Prophylactic administration of CpG oligodeoxynucleotides (CpG ODNs) is known to confer protection against lethal sepsis caused by Burkholderia pseudomallei in the mouse model. The mechanisms whereby CpG regulates the innate immune response to provide protection against B. pseudomallei, however, are poorly characterized. In the present study, we demonstrate that intranasal treatment of mice with Class C CpG, results in recruitment of inflammatory monocytes and neutrophils to the lung at 48 h post-treatment. Mice infected with B. pseudomallei 48 h post-CpG treatment had reduced organ bacterial load and significantly altered cytokine and chemokine profiles concomitant with protection as compared to control animals. CpG administration reduced the robust production of chemokines and pro-inflammatory cytokines in blood, lung and spleen, observed following infection of non-treated animals. Death of control animals coincided with the time of peak cytokine production (day 1-3), while a moderate; sustained cytokine production in CpG-treated animals was associated with survival. In general, CpG treatment resulted in diminished expression of cytokines and chemokines post-infection, though IL-12p40 was released in larger quantities in CpG treated animals. In contrast to CpG-treated animals, the lungs of infected control animals were infiltrated with leukocytes, especially neutrophils, and large numbers of necrotic lesions were observed in lung sections. Therapeutic treatment of B. pseudomallei-infected animals with CpG at 24 h post-infection did not impact survival compared to control animals. In summary, protection of CpG-treated animals was associated with recruitment of inflammatory monocytes and neutrophils into the lungs prior to infection. These responses correspond with early control of bacterial growth, a dampened inflammatory cytokine/chemokine response, reduced lung pathology, and greatly increased survival. In contrast, a delay in recruitment of inflammatory cell populations, despite a robust production of pro-inflammatory cytokines, was associated with poorly controlled bacterial growth, severe lung pathology, and death of control animals.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Burkholderia pseudomallei/patogenicidade , Imunidade Inata , Pulmão/imunologia , Melioidose/imunologia , Melioidose/prevenção & controle , Oligodesoxirribonucleotídeos/administração & dosagem , Animais , Burkholderia pseudomallei/isolamento & purificação , Quimiocinas/metabolismo , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Pulmão/microbiologia , Pulmão/patologia , Melioidose/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/imunologia , Monócitos/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Baço/imunologia , Baço/microbiologia , Baço/patologia , Taxa de Sobrevida
10.
Front Microbiol ; 2: 227, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22125550

RESUMO

Burkholderia are highly evolved Gram-negative bacteria that primarily infect solipeds but are transmitted to humans by ingestion and cutaneous or aerosol exposures. Heightened concern over human infections of Burkholderia mallei and the very closely related species B. pseudomallei is due to the pathogens' proven effectiveness as bioweapons, and to the increased potential for natural opportunistic infections in the growing diabetic and immuno-compromised populations. These Burkholderia species are nearly impervious to antibiotic treatments and no vaccine exists. In this study, the genome of the highly virulent B. mallei ATCC23344 strain was examined by expression library immunization for gene-encoded protective antigens. This protocol for genomic-scale functional screening was customized to accommodate the unusually large complexity of Burkholderia, and yielded 12 new putative vaccine candidates. Five of the candidates were individually tested as protein immunogens and three were found to confer significant partial protection against a lethal pulmonary infection in a murine model of disease. Determinations of peripheral blood cytokine and chemokine profiles following individual protein immunizations show that interleukin-2 (IL-2) and IL-4 are elicited by the three confirmed candidates, but unexpectedly interferon-γ and tumor necrosis factor-α are not. We suggest that these pathogen components, discovered using genetic immunization and confirmed in a conventional protein format, will be useful toward the development of a safe and effective glanders vaccine.

11.
Front Microbiol ; 2: 174, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21904535

RESUMO

Bioluminescent imaging (BLI) technology is a powerful tool for monitoring infectious disease progression and treatment approaches. BLI is particularly useful for tracking fastidious intracellular pathogens that might be difficult to recover from certain organs. Burkholderia mallei, the causative agent of glanders, is a facultative intracellular pathogen and has been classified by the CDC as a Category B select agent due to its highly infectious nature and potential use as a biological weapon. Very little is known regarding pathogenesis or treatment of glanders. We investigated the use of bioluminescent reporter constructs to monitor the dynamics of infection as well as the efficacy of therapeutics for B. mallei in real-time. A stable luminescent reporter B. mallei strain was created using the pUTmini-Tn5::luxKm2 plasmid and used to monitor glanders in the BALB/c murine model. Mice were infected via the intranasal route with 5 × 10(3) bacteria and monitored by BLI at 24, 48, and 72 h. We verified that our reporter construct maintained similar virulence and growth kinetics compared to wild-type B. mallei and confirmed that it maintains luminescent stability in the presence or absence of antibiotic selection. The luminescent signal was initially seen in the lungs, and progressed to the liver and spleen over the course of infection. We demonstrated that antibiotic treatment 24 h post-infection resulted in reduction of bioluminescence that can be attributed to decreased bacterial burden in target organs. These findings suggest that BLI can be used to monitor disease progression and efficacy of therapeutics during glanders infections. Finally, we report an alternative method to mini-Tn5::luxKm2 transposon using mini-Tn7-lux elements that insert site-specifically at known genomic attachment sites and that can also be used to tag bacteria.

12.
J Neuroimmunol ; 234(1-2): 165-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21481948

RESUMO

Autoimmune myasthenia gravis (MG) is characterized by T cell and antibody responses to muscle nicotinic acetylcholine receptor (AChR). It is well known that MG as other autoimmune diseases is more prevalent in women than men and estrogen administration enhances experimental autoimmune MG (EAMG) severity. To determine whether estrogen influences EAMG pathogenesis through estrogen receptor alpha (ERα) activation, ERα knockout (KO) and wild-type (WT) C57BL/6 mice were immunized with AChR. ERα KO mice were equally susceptible to EAMG as WT mice and exhibited comparable antibody and immunopathological responses to AChR, suggesting a lack of involvement of ERα in EAMG pathogenesis.


Assuntos
Receptor alfa de Estrogênio/deficiência , Miastenia Gravis Autoimune Experimental/genética , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/patologia , Animais , Anticorpos/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Predisposição Genética para Doença/genética , Força da Mão/fisiologia , Imunização/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miastenia Gravis Autoimune Experimental/fisiopatologia , Receptores Colinérgicos/imunologia , Fatores de Tempo
13.
Faraday Discuss ; 149: 23-36; discussion 63-77, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21413172

RESUMO

Rapid detection of the category B biothreat agents Burkholderia pseudomallei and Burkholderia mallei in acute infections is critical to ensure that appropriate treatment is administered quickly to reduce an otherwise high probability of mortality (ca. 40% for B. pseudomallei). We are developing assays that can be used in clinical laboratories or security applications for the direct detection of surface-localized and secreted macromolecules produced by these organisms. We present our current medium-throughout approach for target selection and production of Burkholderia macromolecules and describe the generation of a Fab molecule targeted to the B. mallei BimA protein. We also present development of prototype assays for detecting Burkholderia species using anti-lipopolysaccharide antibodies.


Assuntos
Burkholderia mallei/isolamento & purificação , Burkholderia pseudomallei/isolamento & purificação , Ensaio de Imunoadsorção Enzimática/métodos , Mormo/microbiologia , Melioidose/microbiologia , Animais , Burkholderia mallei/metabolismo , Burkholderia pseudomallei/metabolismo , Chaperonina 60/química , Chaperonina 60/metabolismo , Mormo/diagnóstico , Humanos , Melioidose/diagnóstico , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/metabolismo
14.
Clin Dev Immunol ; 2011: 768542, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21197095

RESUMO

Mycobacterium tuberculosis and M. bovis share >99% genetic identity and induce similar host responses and disease profiles upon infection. There is a rich history of codiscovery in the development of control measures applicable to both human and bovine tuberculosis (TB) including skin-testing procedures, M. bovis BCG vaccination, and interferon-γ release assays. The calf TB infection model offers several opportunities to further our understanding of TB immunopathogenesis. Recent observations include correlation of central memory immune responses with TB vaccine efficacy, association of SIRPα(+) cells in ESAT-6:CFP10-elicited multinucleate giant cell formation, early γδ T cell responses to TB, antimycobacterial activity of memory CD4(+) T cells via granulysin production, association of specific antibody with antigen burden, and suppression of innate immune gene expression in infected animals. Partnerships teaming researchers with veterinary and medical perspectives will continue to provide mutual benefit to TB research in man and animals.


Assuntos
Vacinas contra a Tuberculose , Tuberculose Bovina , Tuberculose , Animais , Animais Recém-Nascidos , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Biomarcadores/análise , Bovinos , Perfilação da Expressão Gênica , Humanos , Imunidade/genética , Memória Imunológica , Macaca fascicularis , Masculino , Camundongos , Mycobacterium bovis/genética , Mycobacterium bovis/imunologia , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/imunologia , Receptores Imunológicos/agonistas , Linfócitos T/imunologia , Tuberculose/imunologia , Tuberculose/prevenção & controle , Vacinas contra a Tuberculose/genética , Vacinas contra a Tuberculose/imunologia , Tuberculose Bovina/imunologia , Tuberculose Bovina/prevenção & controle , Vacinação/métodos
15.
J Cent Nerv Syst Dis ; 3: 67-73, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-23861639

RESUMO

Alzheimer's disease (AD) is the most common neurodegenerative disorder. Currently available therapies are symptomatic but do not alter underlying disease progression. Immunotherapeutic approaches such as anti Aß peptide active vaccination trials have had limited success to date. Intravenous immunoblobulin (IVIg) is widely used in immune-mediated neurological disorders such myasthenia gravis and Guillain-Barre syndrome. These preparations have been obtained from the pooled plasma of healthy human donors and contain natural anti-amyloid antibodies and are well tolerated. A small pilot study of passive immunotherapy using IVIg has suggested cognitive improvement. A multicenter phase III trial is ongoing and will determine whether or not this treatment can ameliorate cognitive deficits in mild-to-moderate AD. Here, we briefly review the pathogenic role of amyloid and tau in AD, as well as immunotherapeutic efforts to date. We also summarize what is known about naturally occurring anti-Aß and tau antibodies in IVIg with a view toward explaining potential mechanisms underlying their therapeutic effects.

16.
Vet Immunol Immunopathol ; 138(4): 312-7, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21074276

RESUMO

Secretory IgA (SIgA) constitutes the largest component of the humoral immune system of the body with gram quantities of this isotype produced by mammals on a daily basis. Secretory IgA (SIgA) antibodies function by both blocking pathogen/commensal entry at mucosal surfaces and virus neutralization. Several pathways of induction of IgA responses have been described which depend on T cells (T cell dependent or TD) pathways or are independent of T cells (T-independent or TI) and are mediated by dendritic cells (DCs) and/or epithelial cells. Many elements of IgA regulation readily cross species barriers (adjuvants, soluble and cognate factors) and are highly conserved whereas other pathways may be more specific to any given species and must be evaluated. Regulation of IgA production in cattle is not completely understood and thus we have focused in part on highly conserved factors such as transforming growth factor beta, Type I and Type 2 interferons, neuropeptides which interdigitate mucosal tissues (vasoactive intestinal peptide or VIP), and a small peptide (IgA inducing peptide or IGIP) which can serve as targets for modulation and increasing SIgA virus-specific antibodies. We have evaluated the potential utility of modulating these factors in vitro in regulation of qualitative aspects of antibodies of the IgM, IgG and IgA isotypes at mucosal surfaces and in secretions of the upper and lower respiratory tract to a virus of economic and public health importance, foot and mouth disease virus (FMDV). IgA responses in cattle are essential for host defense in response to various infectious agents. In cattle, IgA is not released into the colostrum, as is the case for other mammals but only IgG1 is selectively transported. In previous studies in cattle, IgA has been shown to be regulated by several cytokines including IFN-gamma, Type I interferons such as IFN-alpha and IFN-tau, transforming growth factor beta, IgA inducing peptide and other potential factors such as APRIL and BlyS which have not yet been fully evaluated in cattle. Many of these factors, namely TGF-beta and Type I interferons block cell cycle progression which is an essential component of Ig class switching and thus these factors require additional regulatory factors such as IL-2 to drive cells through cell cycle resulting in class switch recombination. Among these factors, IgA inducing peptide was originally identified from a bovine gut associated lymphoid tissue expression library and is highly conserved in pigs and humans at >90% at the amino acid level. The factor is regulated differently in various species but is consistently produced by dendritic cells.


Assuntos
Bovinos/imunologia , Imunoglobulina A/biossíntese , Animais , Linfócitos B/imunologia , Citocinas/imunologia , Células Dendríticas/imunologia , Humanos , Imunidade Humoral , Imunoglobulina A Secretora/biossíntese , Interferons/imunologia , Camundongos , Especificidade da Espécie , Peptídeo Intestinal Vasoativo/imunologia
17.
Vaccine ; 28(47): 7551-5, 2010 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-20837078

RESUMO

Burkholderia thailandensis is a less virulent close relative of Burkholderia pseudomallei, a CDC category B biothreat agent. We have previously shown that lipopolysaccharide (LPS) extracted from B. pseudomallei can provide protection against a lethal challenge of B. pseudomallei in a mouse model of melioidosis. Sugar analysis on LPS from B. thailandensis strain E264 confirmed that this polysaccharide has a similar structure to LPS from B. pseudomallei. Mice were immunised with LPS from B. thailandensis or B. pseudomallei and challenged with a lethal dose of B. pseudomallei strain K96243. Similar protection levels were observed when either LPS was used as the immunogen. This data suggests that B. thailandensis LPS has the potential to be used as part of a subunit based vaccine against pathogenic B. pseudomallei.


Assuntos
Burkholderia pseudomallei/patogenicidade , Burkholderia/patogenicidade , Lipopolissacarídeos/imunologia , Melioidose/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/isolamento & purificação , Vacinas Bacterianas/imunologia , Burkholderia/imunologia , Burkholderia pseudomallei/imunologia , Modelos Animais de Doenças , Feminino , Lipopolissacarídeos/isolamento & purificação , Melioidose/imunologia , Camundongos , Camundongos Endogâmicos BALB C
18.
Expert Rev Anti Infect Ther ; 8(3): 325-38, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20192686

RESUMO

Burkholderia pseudomallei and Burkholderia mallei are the causative agents of melioidosis and glanders, respectively. Both Gram-negative pathogens are endemic in many parts of the world. Although natural acquisition of these pathogens is rare in the majority of countries, these bacteria have recently gained much interest because of their potential as bioterrorism agents. In modern times, their potential destructive impact on public health has escalated owing to the ability of these pathogens to cause opportunistic infections in diabetic and perhaps otherwise immunocompromised people, two growing populations worldwide. For both pathogens, severe infection in humans carries a high mortality rate, both species are recalcitrant to antibiotic therapy - B. pseudomallei more so than B. mallei - and no licensed vaccine exists for either prophylactic or therapeutic use. The potential malicious use of these organisms has accelerated the investigation of new ways to prevent and to treat the diseases. The availability of several B. pseudomallei and B. mallei genome sequences has greatly facilitated target identification and development of new therapeutics. This review provides a compilation of literature covering studies in antimelioidosis and antiglanders antimicrobial drug discovery, with a particular focus on potential novel therapeutic approaches to combat these diseases.


Assuntos
Antibacterianos , Burkholderia mallei/efeitos dos fármacos , Burkholderia pseudomallei/efeitos dos fármacos , Mormo/tratamento farmacológico , Melioidose/tratamento farmacológico , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Cricetinae , Modelos Animais de Doenças , Mormo/microbiologia , Humanos , Melioidose/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Ratos
19.
Artigo em Inglês | MEDLINE | ID: mdl-24379895

RESUMO

Burkholderia mallei and B. pseudomallei are Gram-negative pathogenic bacteria, responsible for the diseases glanders and melioidosis, respectively. Furthermore, there is currently no vaccine available against these Burkholderia species. In this study, we aimed to identify protective proteins against these pathogens. Immunization with recombinant B. mallei Hcp1 (type VI secreted/structural protein), BimA (autotransporter protein), BopA (type III secreted protein), and B. pseudomallei LolC (ABC transporter protein) generated significant protection against lethal inhaled B. mallei ATCC23344 and B. pseudomallei 1026b challenge. Immunization with BopA elicited the greatest protective activity, resulting in 100% and 60% survival against B. mallei and B. pseudomallei challenge, respectively. Moreover, sera from recovered mice demonstrated reactivity with the recombinant proteins. Dendritic cells stimulated with each of the different recombinant proteins showed distinct cytokine patterns. In addition, T cells from immunized mice produced IFN-γ following in vitro re-stimulation. These results indicated therefore that it was possible to elicit cross-protective immunity against both B. mallei and B. pseudomallei by vaccinating animals with one or more novel recombinant proteins identified in B. mallei.

20.
Tuberculosis (Edinb) ; 89 Suppl 1: S33-40, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20006302

RESUMO

Artemisia afra [Jacq] (Asteraceae) phytotherapy is widely used for its medicinal properties in traditional practices. In this study we investigated whether extracts of A. afra are capable of controlling mycobacterial replication. For Mycobacterium aurum cultured in the presence of aqueous-, methanol- and dichloromethane (DCM) extracts of A. afra we found that bacterial replication was inhibited by the dichloromethane extract only. Activity of the DCM extract was confirmed in dose-dependent studies against both M. aurum and M. tuberculosis with an IC(50) =270 microg/ml and IC(50) = 290microg/ml, respectively. Fractionation of the DCM extract and evaluation of its efficacy in vitro found that most of the antimycobacterial activity was associated with isolate fraction C8 that contained several sesquiterpene lactones, the most prominent of which are Artemin and Arsubin. Evaluation of the bactericidal efficacy in vitro showed that isolate fraction C8 reduced replication of M. aurum and M. tuberculosis in a dose-dependent manner with IC(50) =1.9 microg/ml and IC(50) = 2.0 microg/ml, respectively, and an MIC = 10 microg/ml. Further, isolate fraction C8 and the DCM extract was administered to M. tuberculosis-infected mice at a tolerated dose of 1000 microg/kg for up to 26 weeks and mycobacterial burdens compared to untreated-, INH/RIF treated- and aqueous-extract-treated animals to assess its bactericidal activity in vivo. Bacterial replication remained unaffected during treatment with either isolate fraction C8 or the DCM extract resulting in pulmonary and splenic bacilli burdens comparable to that of untreated mice. In contrast, INH/RIF treatment cleared M. tuberculosis infection after only 8 weeks to undetectable levels. Interestingly, treatment of M. tuberculosis-infected mice with aqueous extract of A. afra regulated pulmonary inflammation during early infection notwithstanding its inability to inhibit mycobacterial growth. This study clearly demonstrates that A. afra contains in vitro anti-mycobacterial activity, modulates pulmonary inflammation in early mycobacterial infection, and that the mouse experimental tuberculosis model may serve as a useful assay for evaluating the utility of phytotherapy.


Assuntos
Artemisia , Cloreto de Metileno/farmacologia , Extratos Vegetais/farmacologia , Tuberculose/tratamento farmacológico , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Fitoterapia , Tuberculose/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...