Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Med Entomol ; 61(2): 442-453, 2024 03 13.
Article in English | MEDLINE | ID: mdl-38104248

ABSTRACT

With the introduction of siliconized artificial membranes, various artificial feeding systems (AFS) for hard ticks (Ixodidae) have been developed over the last decades. Most AFS utilize similar core components but employ diverse approaches, materials, and experimental conditions. Published work describes different combinations of the core components without experimental optimizations for the artificial feeding of different tick species. Amblyomma americanum L., (Acari: Ixodidae) (lone star tick) is a known vector and reservoir for diverse tick-borne pathogens, such as Rickettsia amblyommatis and Ehrlichia chaffeensis. Ongoing environmental changes have supported the expansion of A. americanum into new habitats, contributing to increased tick-borne diseases in endemic areas. However, a significant knowledge gap exists in understanding the underlying mechanisms involved in A. americanum interactions with tick-borne pathogens. Here, we performed a systematic analysis and developed an optimized AFS for nymphal lone star ticks. Our results demonstrate that Goldbeater's membranes, rabbit hair, hair extract, and adult lone star ticks significantly improved the attachment rate of nymphal ticks, whereas tick frass and frass extract did not. With the optimized conditions, we achieved an attachment rate of 46 ±â€…3% and a success rate of 100% (i.e., one or more attached ticks) in each feeding experiment for nymphal lone star ticks. When fed on sheep blood spiked with R. amblyommatis, both nymphal and adult lone star ticks acquired and maintained R. amblyommatis, demonstrating the feasibility of studying A. americanum-pathogen interactions using AFS. Our study can serve as a roadmap to optimize and improve AFS for other medically relevant tick species.


Subject(s)
Ixodidae , Rickettsia , Rickettsiaceae , Rabbits , Animals , Sheep , Ixodidae/microbiology , Amblyomma , Rickettsiales , Nymph/microbiology
2.
Ticks Tick Borne Dis ; 14(2): 102088, 2023 03.
Article in English | MEDLINE | ID: mdl-36436461

ABSTRACT

Since its discovery in the United States in 2017, the Asian longhorned tick (Haemaphysalis longicornis) has been detected in most eastern states between Rhode Island and Georgia. Long Island, east of New York City, a recognized high-risk area for tick-borne diseases, is geographically close to New Jersey and New York sites where H. longicornis was originally found. However, extensive tick surveys conducted in 2018 did not identify H. longicornis on Long Island. In stark contrast, our 2022 tick survey suggests that H. longicornis has rapidly invaded and expanded in multiple surveying sites on Long Island (12 out of 17 sites). Overall, the relative abundance of H. longicornis was similar to that of lone star ticks, Amblyomma americanum, a previously recognized tick species abundantly present on Long Island. Interestingly, our survey suggests that H. longicornis has expanded within the Appalachian forest ecological zone of Long Island's north shore compared to the Pine Barrens located on the south shore of Long Island. The rapid invasion and expansion of H. longicornis into an insular environment are different from the historical invasion and expansion of two native tick species, Ixodes scapularis (blacklegged tick or deer tick) and A. americanum, in Long Island. The implications of H. longicornis transmitting or introducing tick-borne pathogens of public health importance remain unknown.


Subject(s)
Ixodidae , Tick-Borne Diseases , Ticks , Animals , United States , New York City , Georgia , Amblyomma
5.
6.
Am J Pathol ; 189(1): 4-5, 2019 01.
Article in English | MEDLINE | ID: mdl-30558721

ABSTRACT

This Editorial describes the extension in scope of The American Journal of Pathology.


Subject(s)
Artificial Intelligence , Pathology, Clinical , Humans , Periodicals as Topic
7.
Am J Pathol ; 188(1): 2-3, 2018 01.
Article in English | MEDLINE | ID: mdl-29132929

ABSTRACT

This Editorial describes new enhanced scope of The American Journal of Pathology and introduces its new editorial team.


Subject(s)
Periodicals as Topic , Publishing , Editorial Policies , Humans
8.
Infect Immun ; 81(8): 2828-37, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23716605

ABSTRACT

Francisella tularensis, the bacterial cause of tularemia, infects the liver and replicates in hepatocytes in vivo and in vitro. However, the factors that govern adaptation of F. tularensis to the intrahepatocytic niche have not been identified. Using cDNA microarrays, we determined the transcriptional profile of the live vaccine strain (LVS) of F. tularensis grown in the FL83B murine hepatocytic cell line compared to that of F. tularensis cultured in broth. The fslC gene of the fsl operon was the most highly upregulated. Deletion of fslC eliminated the ability of the LVS to produce siderophore, which is involved in uptake of ferric iron, but it did not impair its growth in hepatocytes, A549 epithelial cells, or macrophages. Therefore, we sought an alternative means by which F. tularensis might obtain iron. Deletion of feoB, which encodes a putative ferrous iron transporter, retarded replication of the LVS in iron-restricted media, reduced its growth in hepatocytic and epithelial cells, and impaired its acquisition of iron. Survival of mice infected intradermally with a lethal dose of the LVS was slightly improved by deletion of fslC but was not altered by loss of feoB. However, the ΔfeoB mutant showed diminished ability to colonize the lungs, liver, and spleen of mice that received sublethal inocula. Thus, FeoB represents a previously unidentified mechanism for uptake of iron by F. tularensis. Moreover, failure to produce a mutant strain lacking both feoB and fslC suggests that FeoB and the proteins of the fsl operon are the only major means by which F. tularensis acquires iron.


Subject(s)
Cation Transport Proteins/metabolism , Francisella tularensis/metabolism , Iron/metabolism , Tularemia/metabolism , Animals , Cell Line , Disease Models, Animal , Hepatocytes/microbiology , Humans , Macrophages/metabolism , Macrophages/microbiology , Mice , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Virulence/physiology
9.
Infect Immun ; 80(10): 3490-500, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22851745

ABSTRACT

Yersinia pestis and many other Gram-negative pathogenic bacteria use the chaperone/usher (CU) pathway to assemble virulence-associated surface fibers termed pili or fimbriae. Y. pestis has two well-characterized CU pathways: the caf genes coding for the F1 capsule and the psa genes coding for the pH 6 antigen. The Y. pestis genome contains additional CU pathways that are capable of assembling pilus fibers, but the roles of these pathways in the pathogenesis of plague are not understood. We constructed deletion mutations in the usher genes for six of the additional Y. pestis CU pathways. The wild-type (WT) and usher deletion strains were compared in the murine bubonic (subcutaneous) and pneumonic (intranasal) plague infection models. Y. pestis strains containing deletions in CU pathways y0348-0352, y1858-1862, and y1869-1873 were attenuated for virulence compared to the WT strain by the intranasal, but not subcutaneous, routes of infection, suggesting specific roles for these pathways during pneumonic plague. We examined binding of the Y. pestis WT and usher deletion strains to A549 human lung epithelial cells, HEp-2 human cervical epithelial cells, and primary human and murine macrophages. Y. pestis CU pathways y0348-0352 and y1858-1862 were found to contribute to adhesion to all host cells tested, whereas pathway y1869-1873 was specific for binding to macrophages. The correlation between the virulence attenuation and host cell binding phenotypes of the usher deletion mutants identifies three of the additional CU pathways of Y. pestis as mediating interactions with host cells that are important for the pathogenesis of plague.


Subject(s)
Bacterial Adhesion/physiology , Molecular Chaperones/metabolism , Plague/microbiology , Yersinia pestis/metabolism , Adenocarcinoma/microbiology , Animals , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Cell Line, Tumor , Epithelial Cells/microbiology , Female , Fimbriae, Bacterial , Gene Deletion , Gene Expression Regulation, Bacterial/physiology , Humans , Lung/cytology , Lung Neoplasms/microbiology , Mice , Mice, Inbred C57BL , Molecular Chaperones/genetics , Plague/metabolism , Yersinia pestis/pathogenicity
10.
Am J Pathol ; 180(6): 2184-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22551843

ABSTRACT

Research articles on atherosclerosis have been well represented in The American Journal of Pathology (AJP), with more than 500 articles published since 1925. An initial focus on descriptive studies led to the proposal that atherosclerosis occurs as a response to vascular injury. With time, this view was modified by a greater understanding of the roles played by lipids and integrity of the vessel wall's constituent cells and matrix. AJP has been a major contributor to the field, publishing numerous seminal research papers and review articles on the latest advances in atherosclerosis. This Centennial Review highlights these myriad contributions.


Subject(s)
Atherosclerosis/history , Periodicals as Topic/history , Animals , Atherosclerosis/etiology , Atherosclerosis/pathology , Biomedical Research/history , Biomedical Research/trends , History, 20th Century , History, 21st Century , Humans , Microscopy, Electron , Models, Biological , Muscle, Smooth, Vascular/ultrastructure , United States
11.
Infect Immun ; 80(7): 2371-81, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22526678

ABSTRACT

Recent studies have linked accumulation of the Gr-1⁺ CD11b⁺ cell phenotype with functional immunosuppression in diverse pathological conditions, including bacterial and parasitic infections and cancer. Gr-1⁺ CD11b⁺ cells were the largest population of cells present in the spleens of mice infected with sublethal doses of the Francisella tularensis live vaccine strain (LVS). In contrast, the number of T cells present in the spleens of these mice did not increase during early infection. There was a significant delay in the kinetics of accumulation of Gr-1⁺ CD11b⁺ cells in the spleens of B-cell-deficient mice, indicating that B cells play a role in recruitment and maintenance of this population in the spleens of mice infected with F. tularensis. The splenic Gr-1⁺ CD11b⁺ cells in tularemia were a heterogeneous population that could be further subdivided into monocytic (mononuclear) and granulocytic (polymorphonuclear) cells using the Ly6C and Ly6G markers and differentiated into antigen-presenting cells following ex vivo culture. Monocytic, CD11b⁺ Ly6C(hi) Ly6G⁻ cells but not granulocytic, CD11b⁺ Ly6C(int) Ly6G⁺ cells purified from the spleens of mice infected with F. tularensis suppressed polyclonal T-cell proliferation via a nitric oxide-dependent pathway. Although the monocytic, CD11b⁺ Ly6C(hi) Ly6G⁻ cells were able to suppress the proliferation of T cells, the large presence of Gr-1⁺ CD11b⁺ cells in mice that survived F. tularensis infection also suggests a potential role for these cells in the protective host response to tularemia.


Subject(s)
Francisella tularensis/pathogenicity , Myeloid Cells/cytology , Myeloid Cells/physiology , Spleen/immunology , Spleen/pathology , Tularemia/immunology , Tularemia/pathology , Animals , B-Lymphocytes/immunology , CD11b Antigen/analysis , Disease Models, Animal , Female , Immunophenotyping , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Receptors, Chemokine/analysis , T-Lymphocytes/immunology
12.
Am J Pathol ; 179(4): 1917-28, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21820995

ABSTRACT

Interferon (IFN)-γ is present in lesions of patients with Lyme disease and positively correlates with the severity of manifestations. To investigate the role of IFNγ in the development of Lyme carditis, wild-type and IFNγ-deficient C57BL/6 mice were infected with the causative bacterium, Borrelia burgdorferi. Histological analysis revealed no change in the severity of carditis between wild-type and IFNγ-deficient mice at 14, 21, 25, and 28 days after infection. However, a distinct shift in the types of leukocytes within the hearts of IFNγ-deficient mice was observed at 25 days. In the absence of IFNγ, the number of neutrophils in the heart was increased, whereas the number of T lymphocytes was decreased. Bacterial loads within hearts were the same as in wild-type mice. Macrophages secrete chemokines that recruit immune cells, which could contribute to the accumulation of leukocytes in murine Lyme carditis. The ability of IFNγ and B. burgdorferi to activate murine macrophages was examined, and the two stimuli synergistically induced chemoattractants for mononuclear cells (ie, CXCL9, CXCL10, CXCL11, CXCL16, and CCL12) and decreased those for neutrophils (ie, CXCL1, CXCL2, and CXCL3). IFNγ and B. burgdorferi also synergistically enhanced secretion of CXCL9 and CXCL10 by murine cardiac endothelial cells. These results indicate that IFNγ influences the composition of inflammatory infiltrates in Lyme carditis by promoting the accumulation of leukocytes associated with chronic inflammation and suppressing that of cells that typify acute inflammation.


Subject(s)
Cell Movement , Interferon-gamma/metabolism , Leukocytes/pathology , Lyme Disease/immunology , Lyme Disease/pathology , Myocarditis/immunology , Myocarditis/pathology , Animals , Borrelia burgdorferi/drug effects , Borrelia burgdorferi/immunology , Cell Movement/drug effects , Chemokines/genetics , Chemokines/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Gene Expression Regulation/drug effects , Interferon-gamma/deficiency , Interferon-gamma/pharmacology , Leukocytes/drug effects , Lyme Disease/complications , Lyme Disease/microbiology , Mice , Mice, Inbred C57BL , Myocarditis/complications , Myocarditis/microbiology , Neutrophils/drug effects , Neutrophils/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
J Immunol ; 186(6): 3517-26, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21300822

ABSTRACT

Dysregulation of apoptosis is associated with the development of human cancer and resistance to anticancer therapy. We have previously shown in tumor xenografts that DNA alkylating agents induce sporadic cell necrosis and regression of apoptosis-deficient tumors. Sporadic tumor cell necrosis is associated with extracellular release of cellular content such as the high mobility group box 1 (HMGB1) protein and subsequent recruitment of innate immune cells into the tumor tissue. It remained unclear whether HMGB1 and the activation of innate immunity played a role in tumor response to chemotherapy. In this study, we show that whereas DNA alkylating therapy leads to a complete tumor regression in an athymic mouse tumor xenograft model, it fails to do so in tumors deficient in HMGB1. The HMGB1-deficient tumors have an impaired ability to recruit innate immune cells including macrophages, neutrophils, and NK cells into the treated tumor tissue. Cytokine array analysis reveals that whereas DNA alkylating treatment leads to suppression of protumor cytokines such as IL-4, IL-10, and IL-13, loss of HMGB1 leads to elevated levels of these cytokines upon treatment. Suppression of innate immunity and HMGB1 using depleting Abs leads to a failure in tumor regression. Taken together, these results indicate that HMGB1 plays an essential role in activation of innate immunity and tumor clearance in response to DNA alkylating agents.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Drug Resistance, Neoplasm/immunology , HMGB1 Protein/physiology , Immunity, Innate/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , HEK293 Cells , HMGB1 Protein/deficiency , HMGB1 Protein/metabolism , Humans , Male , Mice , Mice, Nude , Necrosis , Neoplasms, Experimental/pathology , Random Allocation
14.
J Immunol ; 185(2): 1124-31, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20543103

ABSTRACT

Various bacterial pathogens activate the endothelium to secrete proinflammatory cytokines and recruit circulating leukocytes. In contrast, there is a distinct lack of activation of these cells by Francisella tularensis, the causative agent of tularemia. Given the importance of endothelial cells in facilitating innate immunity, we investigated the ability of the attenuated live vaccine strain and virulent Schu S4 strain of F. tularensis to inhibit the proinflammatory response of HUVECs. Living F. tularensis live vaccine strain and Schu S4 did not stimulate secretion of the chemokine CCL2 by HUVECs, whereas material released from heat-killed bacteria did. Furthermore, the living bacteria suppressed secretion in response to heat-killed F. tularensis. This phenomenon was dose and contact dependent, and it occurred rapidly upon infection. The living bacteria did not inhibit the activation of HUVECs by Escherichia coli LPS, highlighting the specificity of this suppression. The endothelial protein C receptor (EPCR) confers anti-inflammatory properties when bound by activated protein C. When the EPCR was blocked, F. tularensis lost the ability to suppress activation of HUVECs. To our knowledge, this is the first report that a bacterial pathogen inhibits the host immune response via the EPCR. Endothelial cells are a critical component of the innate immune response to infection, and suppression of their activation by F. tularensis is likely a mechanism that aids in bacterial dissemination and evasion of host defenses.


Subject(s)
Antigens, CD/immunology , Endothelial Cells/immunology , Francisella tularensis/immunology , Receptors, Cell Surface/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD/metabolism , Cells, Cultured , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelial Protein C Receptor , Enzyme-Linked Immunosorbent Assay , Francisella tularensis/physiology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-8/immunology , Interleukin-8/metabolism , Receptors, Cell Surface/metabolism , Time Factors , Vaccines, Attenuated/immunology
15.
Infect Immun ; 78(4): 1797-806, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20123721

ABSTRACT

Francisella tularensis, the causative agent of tularemia, interacts with host cells of innate immunity in an atypical manner. For most Gram-negative bacteria, the release of lipopolysaccharide (LPS) from their outer membranes stimulates an inflammatory response. When LPS from the attenuated live vaccine strain (LVS) or the highly virulent Schu S4 strain of F. tularensis was incubated with human umbilical vein endothelial cells, neither species of LPS induced expression of the adhesion molecule E-selectin or secretion of the chemokine CCL2. Moreover, a high concentration (10 microg/ml) of LVS or Schu S4 LPS was required to stimulate production of CCL2 by human monocyte-derived macrophages (huMDM). A screen for alternative proinflammatory factors of F. tularensis LVS identified the heat shock protein GroEL as a potential candidate. Recombinant LVS GroEL at a concentration of 10 microg/ml elicited secretion of CXCL8 and CCL2 by huMDM through a TLR4-dependent mechanism. When 1 microg of LVS GroEL/ml was added to an equivalent amount of LVS LPS, the two components synergistically activated the huMDM to produce CXCL8. Schu S4 GroEL was less stimulatory than LVS GroEL and showed a lesser degree of synergy when combined with Schu S4 LPS. These findings suggest that the intrinsically low proinflammatory activity of F. tularensis LPS may be increased in the infected human host through interactions with other components of the bacterium.


Subject(s)
Chaperonin 60/immunology , Francisella tularensis/immunology , Lipopolysaccharides/immunology , Macrophage Activation , Macrophages/immunology , Animals , Bacterial Vaccines/immunology , Chemokine CCL2/metabolism , Female , Humans , Interleukin-8/metabolism , Mice , Mice, Inbred C57BL
16.
Infect Immun ; 78(3): 1022-31, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20028804

ABSTRACT

The highly infectious bacterium Francisella tularensis is a facultative intracellular pathogen and the causative agent of tularemia. TolC, which is an outer membrane protein involved in drug efflux and type I protein secretion, is required for the virulence of the F. tularensis live vaccine strain (LVS) in mice. Here, we show that an LVS DeltatolC mutant colonizes livers, spleens, and lungs of mice infected intradermally or intranasally, but it is present at lower numbers in these organs than in those infected with the parental LVS. For both routes of infection, colonization by the DeltatolC mutant is most severely affected in the lungs, suggesting that TolC function is particularly important in this organ. The DeltatolC mutant is hypercytotoxic to murine and human macrophages compared to the wild-type LVS, and it elicits the increased secretion of proinflammatory chemokines from human macrophages and endothelial cells. Taken together, these data suggest that TolC function is required for F. tularensis to inhibit host cell death and dampen host immune responses. We propose that, in the absence of TolC, F. tularensis induces excessive host cell death, causing the bacterium to lose its intracellular replicative niche. This results in lower bacterial numbers, which then are cleared by the increased innate immune response of the host.


Subject(s)
Bacterial Outer Membrane Proteins/physiology , Francisella tularensis/immunology , Francisella tularensis/pathogenicity , Inflammation , Tularemia/microbiology , Tularemia/pathology , Virulence Factors/physiology , Animals , Bacterial Outer Membrane Proteins/genetics , Cell Line , Colony Count, Microbial , Endothelial Cells/immunology , Endothelial Cells/microbiology , Gene Deletion , Humans , Inflammation Mediators/metabolism , Liver/microbiology , Lung/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Spleen/microbiology , Virulence Factors/deficiency
17.
Vaccine ; 26(41): 5276-88, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18692537

ABSTRACT

Vaccination of mice with Francisella tularensis live vaccine strain (LVS) mutants described so far have failed to induce protection in C57BL/6 mice against challenge with the virulent strain F. tularensis SchuS4. We have previously reported that a mutant of F. tularensis LVS deficient in iron superoxide dismutase (sodB(Ft)) is hypersensitive to oxidative stress and attenuated for virulence in mice. Herein, we evaluated the efficacy of this mutant as a vaccine candidate against respiratory tularemia caused by F. tularensis SchuS4. C57BL/6 mice were vaccinated intranasally (i.n.) with the sodB(Ft) mutant and challenged i.n. with lethal doses of F. tularensis SchuS4. The level of protection against SchuS4 challenge was higher in sodB(Ft) vaccinated group as compared to the LVS vaccinated mice. sodB(Ft) vaccinated mice following SchuS4 challenge exhibited significantly reduced bacterial burden in lungs, liver and spleen, regulated production of pro-inflammatory cytokines and less severe histopathological lesions compared to the LVS vaccinated mice. The sodB(Ft) vaccination induced a potent humoral immune response and protection against SchuS4 required both CD4 and CD8 T cells in the vaccinated mice. sodB(Ft) mutants revealed upregulated levels of chaperonine proteins DnaK, GroEL and Bfr that have been shown to be important for generation of a potent immune response against Francisella infection. Collectively, this study describes an improved live vaccine candidate against respiratory tularemia that has an attenuated virulence and enhanced protective efficacy than the LVS.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , Respiratory Tract Infections/prevention & control , Tularemia/prevention & control , Animals , Antibodies, Bacterial/blood , Bacterial Proteins/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Female , Francisella tularensis/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/immunology , Male , Mice , Mice, Inbred C57BL , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/pathology , Superoxide Dismutase/genetics , Survival Analysis , Tularemia/immunology , Tularemia/pathology , Up-Regulation , Vaccines, Inactivated/immunology
18.
Microb Pathog ; 44(6): 512-23, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18304778

ABSTRACT

Francisella tularensis is a highly virulent bacterium that causes tularemia, a disease that is often fatal if untreated. A live vaccine strain (LVS) of this bacterium is attenuated for virulence in humans but produces lethal disease in mice. F. tularensis has been classified as a Category A agent of bioterrorism. Despite this categorization, little is known about the components of the organism that are responsible for causing disease in its hosts. Here, we report the deletion of a well-characterized lipoprotein of F. tularensis, designated LpnA (also known as Tul4), in the LVS. An LpnA deletion mutant was comparable to the wild-type strain in its ability to grow intracellularly and cause lethal disease in mice. Additionally, mice inoculated with a sublethal dose of the mutant strain were afforded the same protection against a subsequent lethal challenge with the LVS as were mice initially administered a sublethal dose of the wild-type bacterium. The LpnA-deficient strain showed an equivalent ability to promote secretion of chemokines by human monocyte-derived macrophages as its wild-type counterpart. However, recombinant LpnA potently stimulated primary cultures of human macrophages in a Toll-like receptor 2-dependent manner. Although human endothelial cells were also activated by recombinant LpnA, their response was relatively modest. LpnA is clearly unnecessary for multiple functions of the LVS, but its inflammatory capacity implicates it and other Francisella lipoproteins as potentially important to the pathogenesis of tularemia.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Francisella tularensis/immunology , Francisella tularensis/pathogenicity , Inflammation Mediators/immunology , Lipoproteins/immunology , Tularemia/immunology , Animals , Bacterial Proteins/genetics , Bacterial Vaccines/genetics , Chemokines/immunology , Conjugation, Genetic , Endothelial Cells/immunology , Endothelial Cells/microbiology , Female , Francisella tularensis/genetics , Francisella tularensis/metabolism , Humans , Lipoproteins/genetics , Macrophage Activation , Macrophages/immunology , Macrophages/microbiology , Male , Mice , Mice, Inbred C3H , Sequence Deletion , Tularemia/microbiology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Virulence
19.
J Infect Dis ; 196(1): 134-7, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17538893

ABSTRACT

The ability of Francisella tularensis to replicate in macrophages has led many investigators to assume that it resides primarily intracellularly in the blood of mammalian hosts. We have found this supposition to be untrue. In almost all cases, the majority of F. tularensis recovered from the blood of infected mice was in plasma rather than leukocytes. This distribution was observed irrespective of size of inoculum, route of inoculation, time after inoculation, or virulence of the infecting strain. Our findings yield new insight into the pathogenesis of tularemia and may have important ramifications in the search for anti-Francisella therapies.


Subject(s)
Francisella tularensis/physiology , Tularemia/microbiology , Animals , Bacteremia/microbiology , Colony Count, Microbial , Disease Models, Animal , Francisella tularensis/growth & development , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Leukocytes/microbiology , Mice , Mice, Inbred C3H , Microscopy, Fluorescence , Plasma/microbiology
20.
J Immunol ; 178(2): 1172-9, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17202382

ABSTRACT

Borrelia burgdorferi, the agent of Lyme disease, promotes proinflammatory changes in the endothelium that lead to the recruitment of leukocytes. The host immune response to infection results in increased levels of IFN-gamma in the serum and lesions of Lyme disease patients that correlate with greater severity of disease. Therefore, the effect of IFN-gamma on the gene expression profile of primary human endothelial cells exposed to B. burgdorferi was determined. B. burgdorferi and IFN-gamma synergistically augmented the expression of 34 genes, 7 of which encode chemokines. Six of these (CCL7, CCL8, CX3CL1, CXCL9, CXCL10, and CXCL11) attract T lymphocytes, and one (CXCL2) is specific for neutrophils. Synergistic production of the attractants for T cells was confirmed at the protein level. IL-1beta, TNF-alpha, and LPS also cooperated with IFN-gamma to induce synergistic production of CXCL10 by the endothelium, indicating that IFN-gamma potentiates inflammation in concert with a variety of mediators. An in vitro model of the blood vessel wall revealed that an increased number of human T lymphocytes traversed the endothelium exposed to B. burgdorferi and IFN-gamma, as compared with unstimulated endothelial monolayers. In contrast, addition of IFN-gamma diminished the migration of neutrophils across the B. burgdorferi-activated endothelium. IFN-gamma thus alters gene expression by endothelia exposed to B. burgdorferi in a manner that promotes recruitment of T cells and suppresses that of neutrophils. This modulation may facilitate the development of chronic inflammatory lesions in Lyme disease.


Subject(s)
Borrelia burgdorferi/physiology , Endothelium/drug effects , Endothelium/immunology , Interferon-gamma/pharmacology , Borrelia burgdorferi/immunology , Cell Movement , Cells, Cultured , Chemokines/biosynthesis , Chronic Disease , Coculture Techniques , Gene Expression Regulation/drug effects , Humans , Inflammation/immunology , Inflammation/microbiology , Neutrophils/cytology , Neutrophils/drug effects , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...