Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Sci Rep ; 10(1): 10297, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32581238

ABSTRACT

Patients with beta-thalassaemia increase the risk of bacterial infections, particularly Burkholderia pseudomallei (Bp), the causative agent of melioidosis in Thailand. Impaired immune cell functions may be the cause of this susceptibility, but detailed mechanisms have not been defined. In this study, we observed impaired production of IFN-gamma and IL-10 by whole blood from beta-thalassaemia patients upon stimulation with a range of bacteria-derived stimuli. In contrast, IFN-gamma response via TCR and plasma IgG specific for Bp were still intact. Importantly, mRNA expression of heme oxygenase 1 (HO-1), a potential modulator of immune function, was increased in whole blood from beta-thalassaemia patients, either with or without stimulation with Bp in vitro. Induction of HO-1 by hemin or CoPP in vitro reduced production of IFN-gamma and IL-10 from healthy human PBMCs and decreased bacterial clearance activity of whole blood from healthy controls and beta-thalassaemia, while inhibition of HO-1 by SnPP enhanced both functions in healthy controls. These results were confirmed to some extent in purified human monocytes of healthy controls. Our results suggest a mechanism that excess hemin of beta-thalassaemia patients is a significant cause of immune suppression via HO-1 induction and may underlie the susceptibility of these individuals to severe bacterial infection.


Subject(s)
Burkholderia pseudomallei/immunology , Heme Oxygenase-1/metabolism , Leukocytes, Mononuclear/immunology , Melioidosis/immunology , beta-Thalassemia/immunology , Adult , Aged , Cells, Cultured , Female , Healthy Volunteers , Heme Oxygenase-1/genetics , Humans , Immune Tolerance , Interferon-gamma/metabolism , Interleukin-10/metabolism , Leukocytes, Mononuclear/metabolism , Male , Melioidosis/microbiology , Middle Aged , Primary Cell Culture , RNA, Messenger/isolation & purification , Real-Time Polymerase Chain Reaction , Thailand , Young Adult , beta-Thalassemia/blood , beta-Thalassemia/complications
2.
Tuberculosis (Edinb) ; 123: 101939, 2020 07.
Article in English | MEDLINE | ID: mdl-32452426

ABSTRACT

Type 2 diabetes mellitus (T2DM) is an important risk factor for development of tuberculosis (TB). Our previous study showed glibenclamide, an anti-diabetic drug used to control blood glucose concentration, reduced interleukin (IL)-8 secretion from primary human monocytes challenged with M. tuberculosis (Mtb). In mice infected with Mtb, IL-1ß is essential for host resistance through the enhancement of cyclooxygenase that limits excessive Type I interferon (IFN) production and fosters Mtb containment. We hypothesize that glibenclamide may also interfere with monocyte mediated immune responses against Mtb and alter the balance between IL-1ß and IFNα-mediated immunity. Purified monocytes from non-diabetic and diabetic individuals were infected with Mtb or M. bovis BCG. We demonstrate that monocytes from diabetes patients who were being treated with glibenclamide showed reduced IL-1ß and IL-8 secretion when exposed to Mtb. Additionally, these responses also occurred when monocytes from non-diabetic individuals were pre-treated with glibenclamide in vitro. Moreover, this pre-treatment enhanced IFNa1 expression but was not involved with prostaglandin E2 (PGE2) expression in response to Mtb infection. Taken together, our data show that glibenclamide might exacerbate susceptibility of diabetes patients to Mtb infection by reducing IL-1ß and IL-8 production by monocytes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glyburide/toxicity , Hypoglycemic Agents/toxicity , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Monocytes/drug effects , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/microbiology , Adult , Aged , Case-Control Studies , Cells, Cultured , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/immunology , Dinoprostone/metabolism , Female , Host-Pathogen Interactions , Humans , Interferon-alpha/metabolism , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Mycobacterium bovis/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Risk Assessment , Tuberculosis/immunology
3.
Vaccine ; 38(6): 1416-1423, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31862194

ABSTRACT

Tuberculosis still claims more lives than any other pathogen, and a vaccine better than BCG is urgently needed. One of the challenges for novel TB vaccines is to protect against all Mycobacterium tuberculosis lineages, including the most virulent ones, such as the Beijing lineage. Here we developed a live attenuated M. tuberculosis mutant derived from GC1237, a Beijing strain responsible for tuberculosis outbreaks in the Canary Islands. The mutant strain is inactivated both in the Rv1503c gene, responsible for surface glycolipid synthesis, and in the two-component global regulator PhoPR. This double mutant is as safe as BCG in immunodeficient SCID mice. In immune-competent mice and guinea pigs, the mutant is as protective as BCG against M. tuberculosis strains of common lineage 4 (Euro-American). By contrast, in mice the vaccine is protective against a M. tuberculosis strain of lineage 2 (East-Asian, Beijing), while BCG is not. These results highlight differences in protection efficacy of live attenuated M. tuberculosis-derived vaccine candidates depending on their genetic background, and provide insights for the development of novel live vaccines against TB, especially in East-Asian countries where M. tuberculosis strains of the Beijing family are highly dominant.


Subject(s)
Tuberculosis Vaccines/immunology , Tuberculosis , Animals , BCG Vaccine , Guinea Pigs , Mice , Mice, SCID , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , Tuberculosis/prevention & control , Vaccines, Attenuated/immunology
4.
Nat Commun ; 10(1): 2887, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253760

ABSTRACT

Understanding how immune challenges elicit different responses is critical for diagnosing and deciphering immune regulation. Using a modular strategy to interpret the complex transcriptional host response in mouse models of infection and inflammation, we show a breadth of immune responses in the lung. Lung immune signatures are dominated by either IFN-γ and IFN-inducible, IL-17-induced neutrophil- or allergy-associated gene expression. Type I IFN and IFN-γ-inducible, but not IL-17- or allergy-associated signatures, are preserved in the blood. While IL-17-associated genes identified in lung are detected in blood, the allergy signature is only detectable in blood CD4+ effector cells. Type I IFN-inducible genes are abrogated in the absence of IFN-γ signaling and decrease in the absence of IFNAR signaling, both independently contributing to the regulation of granulocyte responses and pathology during Toxoplasma gondii infection. Our framework provides an ideal tool for comparative analyses of transcriptional signatures contributing to protection or pathogenesis in disease.


Subject(s)
Candidiasis/metabolism , Interferon Type I/metabolism , Interferon-gamma/metabolism , Melioidosis/metabolism , Orthomyxoviridae Infections/metabolism , Respiratory Syncytial Virus Infections/metabolism , Animals , Burkholderia pseudomallei , Candida albicans , Candidiasis/immunology , Candidiasis/microbiology , Gene Expression Regulation/immunology , Influenza A Virus, H3N2 Subtype , Interferon Type I/blood , Interferon Type I/genetics , Interferon-gamma/blood , Interferon-gamma/genetics , Lung , Melioidosis/immunology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Receptor, Interferon alpha-beta , Receptors, Interferon , Respiratory Syncytial Virus Infections/immunology , Interferon gamma Receptor
5.
Article in English | MEDLINE | ID: mdl-30834386

ABSTRACT

We have resequenced the genomes of four Burkholderia pseudomallei K96243 laboratory cultures and compared them to the reported genome sequence that was published in 2004. Compared with the reference genome, these laboratory cultures harbored up to 42 single-nucleotide variants and up to 11 indels, including a 31.7-kb deletion in one culture.

6.
J Microbiol Biotechnol ; 29(3): 489-499, 2019 Mar 28.
Article in English | MEDLINE | ID: mdl-30691253

ABSTRACT

Subunit vaccines are safer and more stable than live vaccines although they have the disadvantage of eliciting poor immune response. To develop a subunit vaccine, an effective delivery system targeting the key elements of the protective immune response is a prerequisite. In this study, oxidized carbon nanospheres (OCNs) were used as a subunit vaccine delivery system and tuberculosis (TB) was chosen as a model disease. TB is among the deadliest infectious diseases worldwide and an effective vaccine is urgently needed. The ability of OCNs to deliver recombinant Mycobacterium tuberculosis (Mtb) proteins, Ag85B and HspX, into bone marrow derived macrophages (BMDMs) and dendritic cells (BMDCs) was investigated. For immunization, OCNs were mixed with the two TB antigens as well as the adjuvant monophosphoryl lipid A (MPL). The protective efficacy was analyzed in vaccinated mice by aerosol Mtb challenge with a virulent strain of Mtb and the bacterial burdens were measured. The results showed that OCNs are highly effective in delivering Mtb proteins into the cytosol of BMDMs and BMDCs. Upon immunization, this vaccine formula induced robust Th1 immune response characterized by cytokine profiles from restimulated splenocytes and specific antibody titer. More importantly, enhanced cytotoxic CD8⁺ T cell activation was observed. However, it did not reduce the bacteria burden in the lung and spleen from the aerosol Mtb challenge. Taken together, OCNs are highly effective in delivering subunit protein vaccine and induce robust Th1 and CD8⁺ T cell response. This vaccine delivery system is suitable for application in settings where cell-mediated immune response is needed.


Subject(s)
Carbon/chemistry , Drug Delivery Systems/methods , Nanospheres/chemistry , T-Lymphocytes, Cytotoxic/immunology , Tuberculosis Vaccines , Tuberculosis/prevention & control , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology , Acyltransferases/genetics , Adjuvants, Immunologic , Administration, Mucosal , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bone Marrow , Cytokines/metabolism , Dendritic Cells/drug effects , Disease Models, Animal , Female , Immunity, Cellular , Immunization , Lung/microbiology , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mycobacterium tuberculosis/pathogenicity , Spleen/microbiology , Th1 Cells/drug effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Synthetic
7.
Front Immunol ; 9: 2109, 2018.
Article in English | MEDLINE | ID: mdl-30283449

ABSTRACT

Tuberculosis (TB) is a global public health problem, which is caused by Mycobacterium tuberculosis (Mtb). Type 2 diabetes mellitus (T2DM) is one of the leading predisposing factors for development of TB after HIV/AIDS. Glibenclamide is a widely used anti-diabetic drug in low and middle-income countries where the incidence of TB is very high. In a human macrophage cell line, glibenclamide, a K+ATP-channel blocker, promoted alternative activation of macrophages by enhancing expression of the M2 marker CD206 during M2 polarization. M2 macrophages are considered poorly microbicidal and associated with TB susceptibility. Here, we investigated the effect of glibenclamide on M1 and M2 phenotypes of primary human monocytes and further determined whether specific drug treatment for T2DM individuals influences the antibacterial function of monocytes in response to mycobacterial infection. We found that glibenclamide significantly reduced M1 (HLA-DR+ and CD86+) surface markers and TNF-α production on primary human monocytes against mycobacterial infection. In contrast, M2 (CD163+ and CD206+) surface markers and IL-10 production were enhanced by pretreatment with glibenclamide. Additionally, reduction of bactericidal activity also occurred when primary human monocytes from T2DM individuals who were being treated with glibenclamide were infected with Mtb in vitro, consistent with the cytokine responses. We conclude that glibenclamide reduces M1 and promotes M2 polarization leading to impaired bactericidal ability of primary human monocytes of T2DM individuals in response to Mtb and may lead to increased susceptibility of T2DM individuals to TB and other bacterial infectious diseases.


Subject(s)
Glyburide/pharmacology , Macrophage Activation/drug effects , Macrophages/immunology , Monocytes/immunology , Tuberculosis/immunology , Adult , Aged , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/immunology , Female , Humans , Hypoglycemic Agents/pharmacology , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Macrophage Activation/immunology , Macrophages/metabolism , Male , Mannose Receptor , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Middle Aged , Monocytes/metabolism , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/physiology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Tuberculosis/complications , Tuberculosis/microbiology
8.
PLoS One ; 13(5): e0196202, 2018.
Article in English | MEDLINE | ID: mdl-29771915

ABSTRACT

Burkholderia pseudomallei, a gram-negative intracellular bacillus, is the causative agent of a tropical infectious disease called melioidosis. Bacterial ATP-binding cassette (ABC) transporters import and export a variety of molecules across bacterial cell membranes. At present, their significance in B. pseudomallei pathogenesis is poorly understood. We report here characterization of the BPSL1039-1040 ABC transporter. B. pseudomallei cultured in M9 medium supplemented with nitrate, demonstrated that BPSL1039-1040 is involved in nitrate transport for B. pseudomallei growth under anaerobic, but not aerobic conditions, suggesting that BPSL1039-1040 is functional under reduced oxygen tension. In addition, a nitrate reduction assay supported the function of BPSL1039-1040 as nitrate importer. A bpsl1039-1040 deficient mutant showed reduced biofilm formation as compared with the wild-type strain (P = 0.027) when cultured in LB medium supplemented with nitrate under anaerobic growth conditions. This reduction was not noticeable under aerobic conditions. This suggests that a gradient in oxygen levels could regulate the function of BPSL1039-1040 in B. pseudomallei nitrate metabolism. Furthermore, the B. pseudomallei bpsl1039-1040 mutant had a pronounced effect on plaque formation (P < 0.001), and was defective in intracellular survival in both non-phagocytic (HeLa) and phagocytic (J774A.1 macrophage) cells, suggesting reduced virulence in the mutant strain. The bpsl1039-1040 mutant was found to be attenuated in a BALB/c mouse intranasal infection model. Complementation of the bpsl1039-1040 deficient mutant with the plasmid-borne bpsl1039 gene could restore the phenotypes observed. We propose that the ability to acquire nitrate for survival under anaerobic conditions may, at least in part, be important for intracellular survival and has a contributory role in the pathogenesis of B. pseudomallei.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Biofilms/growth & development , Burkholderia pseudomallei/physiology , Intracellular Space/microbiology , Macrophages/microbiology , Melioidosis/immunology , ATP-Binding Cassette Transporters/genetics , Anaerobiosis , Animals , Bacterial Proteins/genetics , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/metabolism , Burkholderia pseudomallei/pathogenicity , Cell Survival , Disease Models, Animal , Female , HeLa Cells , Humans , Macrophages/cytology , Mice , Mice, Inbred BALB C , Mutation , Nitrites/metabolism , Phenotype , Virulence
9.
Vaccine ; 35(44): 5981-5989, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28336210

ABSTRACT

B. pseudomallei is the cause of melioidosis, a serious an often fatal disease of humans and animals. The closely related bacterium B. mallei, which cases glanders, is considered to be a clonal derivative of B. pseudomallei. Both B. pseudomallei and B. mallei were evaluated by the United States and the former USSR as potential bioweapons. Much of the effort to devise biodefence vaccines in the past decade has been directed towards the identification and formulation of sub-unit vaccines which could protect against both melioidosis and glanders. A wide range of proteins and polysaccharides have been identified which protective immunity in mice. In this review we highlight the significant progress that has been made in developing glycoconjugates as sub-unit vaccines. We also consider some of the important the criteria for licensing, including the suitability of the "animal rule" for assessing vaccine efficacy, the protection required from a vaccine and the how correlates of protection will be identified. Vaccines developed for biodefence purposes could also be used in regions of the world where naturally occurring disease is endemic.


Subject(s)
Bacterial Vaccines/immunology , Burkholderia mallei/immunology , Burkholderia pseudomallei/immunology , Glanders/immunology , Glanders/prevention & control , Melioidosis/immunology , Melioidosis/prevention & control , Animals , Clinical Trials as Topic , Humans
10.
Microb Pathog ; 107: 175-180, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28323151

ABSTRACT

The NRPS/PKS cluster encodes the enzymes necessary for glidobactin synthesis it is partially conserved in various members of the Burkholderia genus including B. pseudomallei. In this study we have shown that the insertional inactivation or deletion of glbC in this cluster in B. pseudomallei could reduce the ability of the bacterium to survive or grow in murine macrophages or in human neutrophils. Exogenously added proteasome inhibitors were able to chemically complement the mutation. The insertional inactivation or deletion of glbC increased virulence in an acute model of infection in Balb/c or C57BL/6 mice but virulence in a chronic model of infection was similar to that of the wild type. Our findings contrast with the previous finding that inactivation of the glb gene cluster in B. pseudomallei strain 1026b resulted in marked attenuation, and provides evidence of differential roles for some genes in virulence of different strains of B. pseudomallei.


Subject(s)
Burkholderia pseudomallei/growth & development , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/metabolism , Lysine/analogs & derivatives , Proteasome Inhibitors/metabolism , Virulence Factors/genetics , Animals , Bacterial Proteins/genetics , Burkholderia pseudomallei/pathogenicity , Cell Line , DNA, Bacterial/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Bacterial , Genes, Bacterial/genetics , Humans , Lysine/drug effects , Lysine/genetics , Macrophages/microbiology , Melioidosis/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Multigene Family/genetics , Mutagenesis, Insertional/methods , Mutation , Neutrophils/microbiology , Peptide Synthases/genetics , Polyketide Synthases/genetics , Sequence Deletion , Survival , Virulence
11.
Sci Rep ; 7: 42791, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28216665

ABSTRACT

Melioidosis, caused by Burkholderia pseudomallei, is endemic in northeastern Thailand and Northern Australia. Severe septicemic melioidosis is associated with high levels of pro-inflammatory cytokines and is correlated with poor clinical outcomes. IL-10 is an immunoregulatory cytokine, which in other infections can control the expression of pro-inflammatory cytokines, but its role in melioidosis has not been addressed. Here, whole blood of healthy seropositive individuals (n = 75), living in N. E. Thailand was co-cultured with B. pseudomallei and production of IL-10 and IFN-γ detected and the cellular sources identified. CD3- CD14+ monocytes were the main source of IL-10. Neutralization of IL-10 increased IFN-γ, IL-6 and TNF-α production and improved bacteria killing. IFN-γ production and microbicidal activity were impaired in individuals with diabetes mellitus (DM). In contrast, IL-10 production was unimpaired in individuals with DM, resulting in an IL-10 dominant cytokine balance. Neutralization of IL-10 restored the IFN-γ response of individuals with DM to similar levels observed in healthy individuals and improved killing of B. pseudomallei in vitro. These results demonstrate that monocyte derived IL-10 acts to inhibit potentially protective cell mediated immune responses against B. pseudomallei, but may also moderate the pathological effects of excessive cytokine production during sepsis.


Subject(s)
Burkholderia pseudomallei/drug effects , Cytokines/metabolism , Diabetes Mellitus, Type 2/blood , Interleukin-10/pharmacology , Melioidosis/immunology , Adult , Aged , Burkholderia pseudomallei/immunology , Cells, Cultured , Endemic Diseases , Female , Healthy Volunteers , Humans , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Male , Middle Aged , Thailand , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Young Adult
12.
Immunology ; 151(1): 98-109, 2017 05.
Article in English | MEDLINE | ID: mdl-28066900

ABSTRACT

Burkholderia pseudomallei is the causative agent of melioidosis, an infectious disease with high incidence and mortality in South East Asia and northern Australia. To date there is no protective vaccine and antibiotic treatment is prolonged and not always effective. Most people living in endemic areas have been exposed to the bacteria and have developed some immunity, which may have helped to prevent disease. Here, we used a humanized mouse model (hu-PBL-SCID), reconstituted with human peripheral blood mononuclear cells from seropositive donors, to illustrate the potential of three known antigens (FliC, OmpA and N-PilO2) for boosting both T-cell and B-cell immune responses. All three antigens boosted the production of specific antibodies in vivo, and increased the number of antibody and interferon-γ-secreting cells, and induced antibody affinity maturation. Moreover, antigen-specific antibodies isolated from either seropositive individuals or boosted mice, were found to enhance phagocytosis and oxidative burst activities from human polymorphonuclear cells. Our study demonstrates that FliC, OmpA and N-PilO2 can stimulate human memory T and B cells and highlight the potential of the hu-PBL-SCID system for screening and evaluation of novel protein antigens for inclusion in future vaccine trials against melioidosis.


Subject(s)
B-Lymphocytes/immunology , Bacterial Vaccines/immunology , Burkholderia pseudomallei/immunology , Melioidosis/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antibodies, Bacterial/blood , B-Lymphocytes/microbiology , Bacterial Outer Membrane Proteins/immunology , Cells, Cultured , Endemic Diseases , Fimbriae Proteins/immunology , Flagellin/immunology , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Melioidosis/epidemiology , Mice , Mice, SCID , T-Lymphocytes/microbiology , Thailand
13.
Sci Rep ; 6: 34794, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27713554

ABSTRACT

The major risk factor for melioidosis, an infectious disease caused by B. pseudomallei, is diabetes mellitus. More than half of diabetic melioidosis patients in Thailand were prescribed glibenclamide. Recent evidence demonstrates that glibenclamide reduces pro-inflammatory cytokine production by polymorphonuclear neutrophils (PMNs) of diabetic individuals in response to this bacterial infection. However, the mechanisms by which glibenclamide affects cytokine production are unknown. We found that PMNs from glibenclamide-treated diabetic individuals infected with live B. pseudomallei in vitro showed lower free glutathione (GSH) levels compared with those of healthy individuals. Glibenclamide decreased GSH levels and glutathione peroxidase (GPx) of PMNs after exposed to live B. pseudomallei. Moreover, glibenclamide reduced cytokine production and migration capacity of infected PMNs, whereas GSH could restore these functions. Taken together, our data show a link between the effect of glibenclamide on GSH and PMN functions in response to B. pseudomallei that may contribute to the susceptibility of diabetic individuals to B. pseudomallei infection.


Subject(s)
Burkholderia pseudomallei/pathogenicity , Glutathione/metabolism , Glyburide/pharmacology , Host-Pathogen Interactions/drug effects , Neutrophils/drug effects , Acetylcysteine/pharmacology , Adult , Burkholderia pseudomallei/drug effects , Case-Control Studies , Cell Movement/drug effects , Cytokines/blood , Diabetes Mellitus/drug therapy , Female , Glutathione/blood , Glutathione/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Male , Melioidosis/drug therapy , Melioidosis/pathology , Neutrophils/microbiology , Phagocytosis/drug effects
14.
J Immunol ; 197(7): 2838-53, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27549173

ABSTRACT

Pattern recognition receptors detect microbial products and induce cytokines, which shape the immunological response. IL-12, TNF-α, and IL-1ß are proinflammatory cytokines, which are essential for resistance against infection, but when produced at high levels they may contribute to immunopathology. In contrast, IL-10 is an immunosuppressive cytokine, which dampens proinflammatory responses, but it can also lead to defective pathogen clearance. The regulation of these cytokines is therefore central to the generation of an effective but balanced immune response. In this study, we show that macrophages derived from C57BL/6 mice produce low levels of IL-12, TNF-α, and IL-1ß, but high levels of IL-10, in response to TLR4 and TLR2 ligands LPS and Pam3CSK4, as well as Burkholderia pseudomallei, a Gram-negative bacterium that activates TLR2/4. In contrast, macrophages derived from BALB/c mice show a reciprocal pattern of cytokine production. Differential production of IL-10 in B. pseudomallei and LPS-stimulated C57BL/6 and BALB/c macrophages was due to a type I IFN and ERK1/2-dependent, but IL-27-independent, mechanism. Enhanced type I IFN expression in LPS-stimulated C57BL/6 macrophages was accompanied by increased STAT1 and IFN regulatory factor 3 activation. Furthermore, type I IFN contributed to differential IL-1ß and IL-12 production in B. pseudomallei and LPS-stimulated C57BL/6 and BALB/c macrophages via both IL-10-dependent and -independent mechanisms. These findings highlight key pathways responsible for the regulation of pro- and anti-inflammatory cytokines in macrophages and reveal how they may differ according to the genetic background of the host.


Subject(s)
Cytokines/biosynthesis , Inflammation/immunology , Interferon Type I/biosynthesis , Interleukin-10/analysis , Macrophages/metabolism , Animals , Burkholderia pseudomallei/immunology , Cytokines/immunology , Interferon Type I/immunology , Interleukin-10/immunology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
15.
PLoS Pathog ; 12(7): e1005770, 2016 07.
Article in English | MEDLINE | ID: mdl-27467705

ABSTRACT

Mycobacterium tuberculosis (Mtb), possesses at least three type VII secretion systems, ESX-1, -3 and -5 that are actively involved in pathogenesis and host-pathogen interaction. We recently showed that an attenuated Mtb vaccine candidate (Mtb Δppe25-pe19), which lacks the characteristic ESX-5-associated pe/ppe genes, but harbors all other components of the ESX-5 system, induces CD4+ T-cell immune responses against non-esx-5-associated PE/PPE protein homologs. These T cells strongly cross-recognize the missing esx-5-associated PE/PPE proteins. Here, we characterized the fine composition of the functional cross-reactive Th1 effector subsets specific to the shared PE/PPE epitopes in mice immunized with the Mtb Δppe25-pe19 vaccine candidate. We provide evidence that the Mtb Δppe25-pe19 strain, despite its significant attenuation, is comparable to the WT Mtb strain with regard to: (i) its antigenic repertoire related to the different ESX systems, (ii) the induced Th1 effector subset composition, (iii) the differentiation status of the Th1 cells induced, and (iv) its particular features at stimulating the innate immune response. Indeed, we found significant contribution of PE/PPE-specific Th1 effector cells in the protective immunity against pulmonary Mtb infection. These results offer detailed insights into the immune mechanisms underlying the remarkable protective efficacy of the live attenuated Mtb Δppe25-pe19 vaccine candidate, as well as the specific potential of PE/PPE proteins as protective immunogens.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cross Reactions , Disease Models, Animal , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Th1 Cells
16.
Vaccine ; 34(23): 2616-21, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27091689

ABSTRACT

Burkholderia pseudomallei is the causative agent of melioidosis, which is associated with a range of clinical manifestations, including sepsis and fatal pneumonia and is endemic in Southeast Asia and Northern Australia. Treatment can be challenging and control of infection involves prolonged antibiotic therapy, yet there are no approved vaccines available to prevent infection. Our aim was to develop and assess the potential of a prophylactic vaccine candidate targeted against melioidosis. The identified candidate is the 22kDa outer membrane protein, OmpW. We previously demonstrated that this protein was immunoprotective in mouse models of Burkholderia cepacia complex (Bcc) infections. We cloned Bp_ompW in Escherichia coli, expressed and purified the protein. Endotoxin free protein administered with SAS adjuvant protected Balb/C mice (75% survival) relative to controls (25% survival) (p<0.05). A potent serological response was observed with IgG2a to IgG1 ratio of 6.0. Furthermore C57BL/6 mice were protected for up to 80 days against a lethal dose of B. pseudomallei and surpassed the efficacy of the live attenuated 2D2 positive control. BpompW is homologous across thirteen sequenced B. pseudomallei strains, indicating that it should be broadly protective against B. pseudomallei. In conclusion, we have demonstrated that BpOmpW is able to induce protective immunity against melioidosis and is likely to be an effective vaccine antigen, possibly in combination with other subunit antigens.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Melioidosis/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Bacterial/blood , Burkholderia pseudomallei , Female , Immunoglobulin G/blood , Mice, Inbred BALB C , Mice, Inbred C57BL , Vaccines, Subunit/immunology
17.
Vaccine ; 34(14): 1665-71, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26917010

ABSTRACT

There is an urgent need for an effective vaccine against human disease caused by Burkholderia pseudomallei, and although a wide range of candidates have been tested in mice none provide high level protection. We considered this might reflect the inability of these vaccine candidates to protect against chronic disease. Using Q-RT PCR we have identified 6 genes which are expressed in bacteria colonising spleens and lungs of chronically infected mice. Three of the genes (BPSL1897, BPSL3369 and BPSL2287) have been expressed in Escherichia coli and the encoded proteins purified. We have also included BPSL2765, a protein known to induce immune responses associated with a reduced incidence of chronic/recurrent disease in humans. Immunisation of mice with a combination of these antigens resulted in the induction of antibody responses against all of the proteins. Compared with mice immunised with capsular polysaccharide or LolC protein, mice immunised with the combination of chronic stage antigens showed enhanced protection against experimental disease in mice.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Melioidosis/prevention & control , Animals , Antibodies, Bacterial/blood , Burkholderia pseudomallei/genetics , Female , Genes, Bacterial , Immunoglobulin G/blood , Lung/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Spleen/microbiology , Transcriptome
18.
Res Microbiol ; 167(3): 159-67, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26654915

ABSTRACT

Kynurenine formamidase (KynB) forms part of the kynurenine pathway which metabolises tryptophan to anthranilate. This metabolite can be used for downstream production of 2-alkyl-4-quinolone (AQ) signalling molecules that control virulence in Pseudomonas aeruginosa. Here we investigate the role of kynB in the production of AQs and virulence-associated phenotypes of Burkholderia pseudomallei K96243, the causative agent of melioidosis. Deletion of kynB resulted in reduced AQ production, increased biofilm formation, decreased swarming and increased tolerance to ciprofloxacin. Addition of exogenous anthranilic acid restored the biofilm phenotype, but not the persister phenotype. This study suggests the kynurenine pathway is a critical source of anthranilate and signalling molecules that may regulate B. pseudomallei virulence.


Subject(s)
Arylformamidase/metabolism , Biofilms/growth & development , Burkholderia pseudomallei/enzymology , Burkholderia pseudomallei/physiology , Locomotion , Quinolones/metabolism , Signal Transduction , Burkholderia pseudomallei/genetics , Gene Deletion , Tryptophan/metabolism , Virulence , ortho-Aminobenzoates/metabolism
19.
Infect Immun ; 84(3): 701-10, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26712202

ABSTRACT

Burkholderia pseudomallei, the causative agent of melioidosis, has complex and poorly understood extracellular and intracellular lifestyles. We used transposon-directed insertion site sequencing (TraDIS) to retrospectively analyze a transposon library that had previously been screened through a BALB/c mouse model to identify genes important for growth and survival in vivo. This allowed us to identify the insertion sites and phenotypes of negatively selected mutants that were previously overlooked due to technical constraints. All 23 unique genes identified in the original screen were confirmed by TraDIS, and an additional 105 mutants with various degrees of attenuation in vivo were identified. Five of the newly identified genes were chosen for further characterization, and clean, unmarked bpsl2248, tex, rpiR, bpsl1728, and bpss1528 deletion mutants were constructed from the wild-type strain K96243. Each of these mutants was tested in vitro and in vivo to confirm their attenuated phenotypes and investigate the nature of the attenuation. Our results confirm that we have identified new genes important to in vivo virulence with roles in different stages of B. pseudomallei pathogenesis, including extracellular and intracellular survival. Of particular interest, deletion of the transcription accessory protein Tex was shown to be highly attenuating, and the tex mutant was capable of providing protective immunity against challenge with wild-type B. pseudomallei, suggesting that the genes identified in our TraDIS screen have the potential to be investigated as live vaccine candidates.


Subject(s)
Bacterial Proteins/metabolism , Burkholderia pseudomallei/growth & development , Burkholderia pseudomallei/metabolism , Melioidosis/microbiology , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Burkholderia pseudomallei/genetics , Female , Humans , Mice , Mice, Inbred BALB C , Microbial Viability , Virulence Factors/genetics
20.
J Immunol ; 195(7): 3248-3261, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26311902

ABSTRACT

Melioidosis, a severe human disease caused by the bacterium Burkholderia pseudomallei, has a wide spectrum of clinical manifestations ranging from acute septicemia to chronic localized illness or latent infection. Murine models have been widely used to study the pathogenesis of infection and to evaluate novel therapies or vaccines, but how faithfully they recapitulate the biology of human melioidosis at a molecular level is not known. In this study, mice were intranasally infected with either high or low doses of B. pseudomallei to generate either acute, chronic, or latent infection and host blood and tissue transcriptional profiles were generated. Acute infection was accompanied by a homogeneous signature associated with induction of multiple innate immune response pathways, such as IL-10, TREM1, and IFN signaling, largely found in both blood and tissue. The transcriptional profile in blood reflected the heterogeneity of chronic infection and quantitatively reflected the severity of disease. Genes associated with fibrosis and tissue remodeling, including matrix metalloproteases and collagen, were upregulated in chronically infected mice with severe disease. Transcriptional signatures of both acute and chronic melioidosis revealed upregulation of iNOS in tissue, consistent with the expression of IFN-γ, but also Arginase-1, a functional antagonist of the iNOS pathway, and was confirmed by immunohistochemistry. Comparison of these mouse blood datasets by pathway and modular analysis with the blood transcriptional signature of patients with melioidosis showed that many genes were similarly perturbed, including Arginase-1, IL-10, TREM1, and IFN signaling, revealing the common immune response occurring in both mice and humans.


Subject(s)
Burkholderia pseudomallei/immunology , Immunity, Innate/immunology , Melioidosis/immunology , Animals , Arginase/biosynthesis , Arginase/blood , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukin-10/blood , Interleukin-10/genetics , Interleukin-10/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Matrix Metalloproteinase 9/blood , Melioidosis/microbiology , Melioidosis/pathology , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Receptors, Immunologic/blood , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Signal Transduction/immunology , Transcriptome/genetics , Triggering Receptor Expressed on Myeloid Cells-1
SELECTION OF CITATIONS
SEARCH DETAIL
...