Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Am J Reprod Immunol ; 81(6): e13111, 2019 06.
Article in English | MEDLINE | ID: mdl-30903720

ABSTRACT

PROBLEM: Neisseria gonorrhoeae (NG) infection has been shown to increase sexual transmission of HIV-1. However, the mechanism of NG-induced enhanced HIV-1 transmission is unknown. METHODS: (a) The cervical tissues were exposed to NG, and cytokine induction was monitored by measuring cytokine proteins in culture supernatants and cytokine mRNAs in tissues. (b) Transcription and replication of HIV-1 in TZM-bl, U1, and ACH2 cells were measured by Beta-Gal activity and p24 proteins in the supernatant, respectively. (c) HIV-1 transmission was assayed in an organ culture system by measuring transmitted HIV-1 in supernatant and HIV-1 gag mRNA in the tissues. (d) Transcriptome analysis was done using second generation sequencing. RESULTS: (a) NG induced membrane ruffling of epithelial layer, caused migration of CD3+ cells to the intraepithelial region, and induced high levels of inflammatory cytokines IL-1ß and TNF-α. (b) NG-induced supernatants (NGIS) increased HIV-1 transcription, induced HIV-1 from latently infected cells, and increased transmission of HIV-1 across cervical mucosa. (c) Transcriptome analysis of the epithelial layer of the tissues exposed to NG, and HIV-1 showed significant upregulation of CXCL10 and IL8. IL-1ß increased the induction of CXCL10 and IL-8 expression in cervical mucosa with a concomitant increase in HIV-1 transmission. CONCLUSION: We present a model in which IL-1ß produced from cervical epithelium during NG exposure increases CXCL10 and IL8 in epithelia. This in turn causes upon HIV-1 infection, the migration of HIV-1 target cells toward the subepithelium, resulting in increased HIV-1 transcription in the sub-mucosa and subsequent enhancement of transmission across cervical mucosa.


Subject(s)
Chemokine CXCL10/immunology , HIV Infections/transmission , Interleukin-1beta/immunology , Interleukin-8/immunology , Neisseria gonorrhoeae , Cells, Cultured , Cervix Uteri/immunology , Epithelium/immunology , Female , Gonorrhea/immunology , HIV Infections/immunology , Humans , Leukocytes, Mononuclear , Organ Culture Techniques
2.
PLoS One ; 12(12): e0189780, 2017.
Article in English | MEDLINE | ID: mdl-29267331

ABSTRACT

Recent avian and swine-origin influenza virus outbreaks illustrate the ongoing threat of influenza pandemics. We investigated immunogenicity and protective efficacy of a multi-antigen (MA) universal influenza DNA vaccine consisting of HA, M2, and NP antigens in cynomolgus macaques. Following challenge with a heterologous pandemic H1N1 strain, vaccinated animals exhibited significantly lower viral loads and more rapid viral clearance when compared to unvaccinated controls. The MA DNA vaccine induced robust serum and mucosal antibody responses but these high antibody titers were not broadly neutralizing. In contrast, the vaccine induced broadly-reactive NP specific T cell responses that cross-reacted with the challenge virus and inversely correlated with lower viral loads and inflammation. These results demonstrate that a MA DNA vaccine that induces strong cross-reactive T cell responses can, independent of neutralizing antibody, mediate significant cross-protection in a nonhuman primate model and further supports development as an effective approach to induce broad protection against circulating and emerging influenza strains.


Subject(s)
Cross Reactions , Influenza Vaccines/immunology , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Neutralizing/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/genetics , Macaca fascicularis , Vaccines, DNA/genetics
3.
JCI Insight ; 2(19)2017 10 05.
Article in English | MEDLINE | ID: mdl-28978810

ABSTRACT

Mycobacterium tuberculosis (Mtb) is a global health threat, compounded by the emergence of drug-resistant strains. A hallmark of pulmonary tuberculosis (TB) is the formation of hypoxic necrotic granulomas, which upon disintegration, release infectious Mtb. Furthermore, hypoxic necrotic granulomas are associated with increased disease severity and provide a niche for drug-resistant Mtb. However, the host immune responses that promote the development of hypoxic TB granulomas are not well described. Using a necrotic Mtb mouse model, we show that loss of Mtb virulence factors, such as phenolic glycolipids, decreases the production of the proinflammatory cytokine IL-17 (also referred to as IL-17A). IL-17 production negatively regulates the development of hypoxic TB granulomas by limiting the expression of the transcription factor hypoxia-inducible factor 1α (HIF1α). In human TB patients, HIF1α mRNA expression is increased. Through genotyping and association analyses in human samples, we identified a link between the single nucleotide polymorphism rs2275913 in the IL-17 promoter (-197G/G), which is associated with decreased IL-17 production upon stimulation with Mtb cell wall. Together, our data highlight a potentially novel role for IL-17 in limiting the development of hypoxic necrotic granulomas and reducing disease severity in TB.


Subject(s)
Granuloma/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Interleukin-17/immunology , Tuberculosis, Pulmonary/immunology , Adult , Aged , Animals , Cell Hypoxia/immunology , Female , Gene Expression Regulation/immunology , Granuloma/microbiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation Mediators/metabolism , Interleukin-17/biosynthesis , Male , Mice, Inbred Strains , Middle Aged , RNA, Messenger/genetics , Tuberculosis, Pulmonary/complications , Young Adult
5.
PLoS One ; 10(8): e0134219, 2015.
Article in English | MEDLINE | ID: mdl-26261989

ABSTRACT

Asthma is a heterogeneous disease whose etiology is poorly understood but is likely to involve innate responses to inhaled microbial components that are found in allergens. The influence of these components on pulmonary inflammation has been largely studied in the context of individual agonists, despite knowledge that they can have synergistic effects when used in combination. Here we have explored the effects of LPS and ß-glucan, two commonly-encountered microbial agonists, on the pathogenesis of allergic and non-allergic respiratory responses to house dust mite allergen. Notably, sensitization with these microbial components in combination acted synergistically to promote robust neutrophilic inflammation, which involved both Dectin-1 and TLR-4. This pulmonary neutrophilic inflammation was corticosteroid-refractory, resembling that found in patients with severe asthma. Thus our results provide key new insights into how microbial components influence the development of respiratory pathology.


Subject(s)
Asthma/etiology , Lipopolysaccharides/immunology , Neutrophils/immunology , beta-Glucans/immunology , Animals , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , Disease Models, Animal , Drug Resistance , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Mice, Knockout , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Pyroglyphidae/immunology , Steroids/administration & dosage , Steroids/pharmacology , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
6.
PLoS Pathog ; 11(1): e1004589, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25621893

ABSTRACT

Aspergillus fumigatus forms ubiquitous airborne conidia that humans inhale on a daily basis. Although respiratory fungal infection activates the adaptor proteins CARD9 and MyD88 via C-type lectin, Toll-like, and interleukin-1 family receptor signals, defining the temporal and spatial pattern of MyD88- and CARD9-coupled signals in immune activation and fungal clearance has been difficult to achieve. Herein, we demonstrate that MyD88 and CARD9 act in two discrete phases and in two cellular compartments to direct chemokine- and neutrophil-dependent host defense. The first phase depends on MyD88 signaling because genetic deletion of MyD88 leads to delayed induction of the neutrophil chemokines CXCL1 and CXCL5, delayed neutrophil lung trafficking, and fatal pulmonary damage at the onset of respiratory fungal infection. MyD88 expression in lung epithelial cells restores rapid chemokine induction and neutrophil recruitment via interleukin-1 receptor signaling. Exogenous CXCL1 administration reverses murine mortality in MyD88-deficient mice. The second phase depends predominately on CARD9 signaling because genetic deletion of CARD9 in radiosensitive hematopoietic cells interrupts CXCL1 and CXCL2 production and lung neutrophil recruitment beyond the initial MyD88-dependent phase. Using a CXCL2 reporter mouse, we show that lung-infiltrating neutrophils represent the major cellular source of CXCL2 during CARD9-dependent recruitment. Although neutrophil-intrinsic MyD88 and CARD9 function are dispensable for neutrophil conidial uptake and killing in the lung, global deletion of both adaptor proteins triggers rapidly progressive invasive disease when mice are challenged with an inoculum that is sub-lethal for single adapter protein knockout mice. Our findings demonstrate that distinct signal transduction pathways in the respiratory epithelium and hematopoietic compartment partially overlap to ensure optimal chemokine induction, neutrophil recruitment, and fungal clearance within the respiratory tract.


Subject(s)
Aspergillus fumigatus/physiology , CARD Signaling Adaptor Proteins/metabolism , Chemokines/metabolism , Myeloid Differentiation Factor 88/metabolism , Pulmonary Aspergillosis/immunology , Signal Transduction , Animals , Humans , Immunity, Innate , Lung/immunology , Mice , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Pulmonary Aspergillosis/microbiology , Receptors, Interleukin-1/metabolism
7.
Vet Immunol Immunopathol ; 163(3-4): 134-45, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25540877

ABSTRACT

The lymphatic endothelium (LE) serves as a conduit for transport of immune cells and soluble antigens from peripheral tissues to draining lymph nodes (LNs), contributing to development of host immune responses and possibly dissemination of microbes. Lymphatic endothelial cells (LECs) are major constituents of the lymphatic endothelium. These specialized cells could play important roles in initiation of host innate immune responses through sensing of pathogen-associated molecular patterns (PAMPs) by pattern recognition receptors (PRRs), including toll-like receptors (TLRs). LECs secrete pro-inflammatory cytokines and chemokines to create local inflammatory conditions for recruitment of naïve antigen presenting cells (APCs) such as dendritic cells (DCs) to sites of infection and/or vaccine administration. In this study, we examined the innate immune potential of primary LEC populations derived from multiple tissues of an animal model for human infectious diseases - the ferret. We generated a total of six primary LEC populations from lung, tracheal, and mesenteric LN tissues from three different ferrets. Standard RT-PCR characterization of these primary LECs showed that they varied in their expression of LEC markers. The ferret LECs were examined for their ability to respond to poly I:C (TLR3 and RIG-I ligand) and other known TLR ligands as measured by production of proinflammatory cytokine (IFNα, IL6, IL10, Mx1, and TNFα) and chemokine (CCL5, CCL20, and CXCL10) mRNAs using real time RT-PCR. Poly I:C exposure induced robust proinflammatory responses by all of the primary ferret LECs. Chemotaxis was performed to determine the functional activity of CCL20 produced by the primary lung LECs and showed that the LEC-derived CCL20 was abundant and functional. Taken together, our results continue to reveal the innate immune potential of primary LECs during pathogen-host interactions and expand our understanding of the roles LECs might play in health and disease in animal models.


Subject(s)
Endothelial Cells/cytology , Ferrets/physiology , Animals , Biomarkers , Cell Culture Techniques , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Endothelial Cells/physiology , Female , Gene Expression Regulation/immunology , Gene Expression Regulation/physiology , Lung , Phylogeny , Toll-Like Receptors/metabolism
8.
mBio ; 5(5): e01277-14, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25182322

ABSTRACT

UNLABELLED: Kaposi's sarcoma (KS) is an unusual neoplasia wherein the tumor consists primarily of endothelial cells infected with human herpesvirus 8 (HHV-8; Kaposi's sarcoma-associated herpesvirus) that are not fully transformed but are instead driven to excess proliferation by inflammatory and angiogenic factors. This oncogenic process has been postulated but unproven to depend on a paracrine effect of an abnormal excess of host cytokines and chemokines produced by HHV-8-infected B lymphocytes. Using newly developed measures for intracellular detection of lytic cycle proteins and expression of cytokines and chemokines, we show that HHV-8 targets a range of naive B cell, IgM memory B cell, and plasma cell-like populations for infection and induction of interleukin-6, tumor necrosis factor alpha, macrophage inhibitory protein 1α, macrophage inhibitory protein 1ß, and interleukin-8 in vitro and in the blood of HHV-8/HIV-1-coinfected subjects with KS. These B cell lineage subsets that support HHV-8 infection are highly polyfunctional, producing combinations of 2 to 5 of these cytokines and chemokines, with greater numbers in the blood of subjects with KS than in those without KS. Our study provides a new paradigm of B cell polyfunctionality and supports a key role for B cell-derived cytokines and chemokines produced during HHV-8 infection in the development of KS. IMPORTANCE: Kaposi's sarcoma (KS) is the most common cancer in HIV-1-infected persons and is caused by one of only 7 human cancer viruses, i.e., human herpesvirus 8 (HHV-8). It is unclear how this virus causes neoplastic transformation. Development and outgrowth of endothelial cell lesions characteristic of KS are hypothesized to be dependent on virus replication and multiple immune mediators produced by the KS cells and inflammatory cells, yet the roles of these viral and cell factors have not been defined. The present study advances our understanding of KS in that it supports a central role for HHV-8 infection of B cells inducing multiple cytokines and chemokines that can drive development of the cancer. Notably, HIV-1-infected individuals who developed KS had greater numbers of such HHV-8-infected, polyfunctional B cells across a range of B cell phenotypic lineages than did HHV-8-infected persons without KS. This intriguing production of polyfunctional immune mediators by B cells serves as a new paradigm for B cell function and classification.


Subject(s)
B-Lymphocytes/virology , Herpesvirus 8, Human/physiology , Sarcoma, Kaposi/virology , B-Lymphocytes/cytology , Cell Line , Cell Proliferation/physiology , Chemokines/metabolism , Cytokines/metabolism , DNA, Viral/genetics , Humans , Immunoglobulin M/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Microarray Analysis , Tumor Necrosis Factor-alpha/metabolism , Virus Replication
9.
mBio ; 5(3): e01031-13, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24781743

ABSTRACT

ABSTRACT HIV-1-infected nonprogressors (NP) inhibit disease progression for years without antiretroviral therapy. Defining the mechanisms for this resistance to disease progression could be important in determining strategies for controlling HIV-1 infection. Here we show that two types of professional antigen-presenting cells (APC), i.e., dendritic cells (DC) and B lymphocytes, from NP lacked the ability to mediate HIV-1 trans infection of CD4(+) T cells. In contrast, APC from HIV-1-infected progressors (PR) and HIV-1-seronegative donors (SN) were highly effective in mediating HIV-1 trans infection. Direct cis infection of T cells with HIV-1 was comparably efficient among NP, PR, and SN. Lack of HIV-1 trans infection in NP was linked to lower cholesterol levels and an increase in the levels of the reverse cholesterol transporter ABCA1 (ATP-binding cassette transporter A1) in APC but not in T cells. Moreover, trans infection mediated by APC from NP could be restored by reconstitution of cholesterol and by inhibiting ABCA1 by mRNA interference. Importantly, this appears to be an inherited trait, as it was evident in APC obtained from NP prior to their primary HIV-1 infection. The present study demonstrates a new mechanism wherein enhanced lipid metabolism in APC results in remarkable control of HIV-1 trans infection that directly relates to lack of HIV-1 disease progression. IMPORTANCE HIV-1 can be captured by antigen-presenting cells (APC) such as dendritic cells and transferred to CD4 helper T cells, which results in greatly enhanced viral replication by a mechanism termed trans infection. A small percentage of HIV-1-infected persons are able to control disease progression for many years without antiretroviral therapy. In our study, we linked this lack of disease progression to a profound inability of APC from these individuals to trans infect T cells. This effect was due to altered lipid metabolism in their APC, which appears to be an inherited trait. These results provide a basis for therapeutic interventions to control of HIV-1 infection through modulation of cholesterol metabolism.


Subject(s)
Cholesterol/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Case-Control Studies , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Disease Progression , Genotype , HIV Infections/genetics , HIV Infections/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Mutation , Receptors, CCR5/genetics , Viral Load
10.
PLoS Pathog ; 10(5): e1004099, 2014 May.
Article in English | MEDLINE | ID: mdl-24831696

ABSTRACT

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), infects one third of the world's population. Among these infections, clinical isolates belonging to the W-Beijing appear to be emerging, representing about 50% of Mtb isolates in East Asia, and about 13% of all Mtb isolates worldwide. In animal models, infection with W-Beijing strain, Mtb HN878, is considered "hypervirulent" as it results in increased mortality and causes exacerbated immunopathology in infected animals. We had previously shown the Interleukin (IL) -17 pathway is dispensable for primary immunity against infection with the lab adapted Mtb H37Rv strain. However, it is not known whether IL-17 has any role to play in protective immunity against infection with clinical Mtb isolates. We report here that lab adapted Mtb strains, such as H37Rv, or less virulent Mtb clinical isolates, such as Mtb CDC1551, do not require IL-17 for protective immunity against infection while infection with Mtb HN878 requires IL-17 for early protective immunity. Unexpectedly, Mtb HN878 induces robust production of IL-1ß through a TLR-2-dependent mechanism, which supports potent IL-17 responses. We also show that the role for IL-17 in mediating protective immunity against Mtb HN878 is through IL-17 Receptor signaling in non-hematopoietic cells, mediating the induction of the chemokine, CXCL-13, which is required for localization of T cells within lung lymphoid follicles. Correct T cell localization within lymphoid follicles in the lung is required for maximal macrophage activation and Mtb control. Since IL-17 has a critical role in vaccine-induced immunity against TB, our results have far reaching implications for the design of vaccines and therapies to prevent and treat emerging Mtb strains. In addition, our data changes the existing paradigm that IL-17 is dispensable for primary immunity against Mtb infection, and instead suggests a differential role for IL-17 in early protective immunity against emerging Mtb strains.


Subject(s)
Immunity, Innate/genetics , Interleukin-17/physiology , Mycobacterium tuberculosis/immunology , Animals , Cells, Cultured , Communicable Diseases, Emerging/genetics , Communicable Diseases, Emerging/immunology , Cytoprotection/genetics , Cytoprotection/immunology , Female , Interleukin-17/genetics , Interleukin-1beta/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/pathogenicity , Receptors, Interleukin-1 Type I/genetics , Toll-Like Receptor 2/physiology , Tuberculosis/genetics , Tuberculosis/immunology
11.
Cell Host Microbe ; 15(2): 153-63, 2014 Feb 12.
Article in English | MEDLINE | ID: mdl-24528862

ABSTRACT

Chronic tuberculosis in an immunocompetent host is a consequence of the delicately balanced growth of Mycobacterium tuberculosis (Mtb) in the face of host defense mechanisms. We identify an Mtb enzyme (TdmhMtb) that hydrolyzes the mycobacterial glycolipid trehalose dimycolate and plays a critical role in balancing the intracellular growth of the pathogen. TdmhMtb is induced under nutrient-limiting conditions and remodels the Mtb envelope to increase nutrient influx but concomitantly sensitizes Mtb to stresses encountered in the host. Consistent with this, a ΔtdmhMtb mutant is more resilient to stress and grows to levels higher than those of wild-type in immunocompetent mice. By contrast, mutant growth is retarded in MyD88(-/-) mice, indicating that TdmhMtb provides a growth advantage to intracellular Mtb in an immunocompromised host. Thus, the effects and countereffects of TdmhMtb play an important role in balancing intracellular growth of Mtb in a manner that is directly responsive to host innate immunity.


Subject(s)
Cord Factors/metabolism , Hydrolases/metabolism , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/physiology , Animals , Cytosol/microbiology , Gene Deletion , Hydrolases/genetics , Hydrolysis , Mice , Mice, Knockout , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development
12.
J Infect Dis ; 209(12): 2012-6, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24403559

ABSTRACT

Highly pathogenic avian influenza virus infection is characterized by a marked inflammatory response, but the impact of infection on dendritic cells (DCs) is unknown. We show that influenza A virus subtype H5N1 infection rapidly and profoundly impacts DCs in cynomolgus macaques, increasing the number of blood myeloid and plasmacytoid DCs by 16- and 60-fold, respectively. Infection was associated with recruitment, activation, and apoptosis of DCs in lung-draining lymph nodes; granulocyte and macrophage infiltration in lungs was also detected, together with expression of CXCL10. This degree of DC mobilization is unprecedented in viral infection and suggests a potential role for DCs in the pathogenesis of highly pathogenic avian influenza virus.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Influenza A Virus, H5N1 Subtype , Orthomyxoviridae Infections/immunology , Animals , Cell Proliferation , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Lung/pathology , Lung/virology , Lymph Nodes/virology , Macaca fascicularis/virology , Macrophages/metabolism , Male , Orthomyxoviridae Infections/pathology
13.
Am J Pathol ; 184(1): 55-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24183780

ABSTRACT

Mucosal vaccines are thought to confer superior protection against mucosal infectious diseases. In addition, mucosal routes of vaccine delivery preferentially induce the generation of T helper 17 (Th17) cells, which produce the cytokine IL-17. Th17 cells are critical in mediating vaccine-induced immunity against several mucosal infectious diseases. However, IL-17 is also a potent proinflammatory cytokine, and we recently showed that IL-17 mediates immunopathology and lung injury after influenza infection in mice. In the present study, we tested the hypothesis that mucosal pre-exposure to Th17-inducing adjuvants can promote disease exacerbation upon subsequent infection with influenza virus. Mice mucosally pre-exposed to Th17-inducing adjuvants, such as type II heat-labile enterotoxin or cholera toxin, resulted in increased morbidity and exacerbated lung inflammation upon subsequent infection with influenza virus. Furthermore, the increased morbidity was accompanied by increased expression of inflammatory chemokines and increased accumulation of neutrophils. Importantly, blockade of the IL-17 pathway in mice pre-exposed to Th17-inducing adjuvants resulted in attenuation of the inflammatory phenotype seen in influenza-infected mice. Our findings indicate that, before mucosal Th17-inducing adjuvants can be used in vaccine strategies, the short- and long-term detrimental effects of such adjuvants on disease exacerbation and lung injury in response to infections, such as influenza, should be carefully studied.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Orthomyxoviridae Infections/immunology , Th17 Cells/immunology , Animals , Female , Flow Cytometry , Immunohistochemistry , In Situ Hybridization , Influenza A virus , Influenza Vaccines/immunology , Interleukin-17/immunology , Male , Mice , Mice, Inbred C57BL , Mucous Membrane/immunology , Orthomyxoviridae Infections/pathology , Polymerase Chain Reaction
14.
Am J Respir Crit Care Med ; 188(9): 1137-46, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24047412

ABSTRACT

RATIONALE: A hallmark of pulmonary tuberculosis (TB) is the formation of granulomas. However, the immune factors that drive the formation of a protective granuloma during latent TB, and the factors that drive the formation of inflammatory granulomas during active TB, are not well defined. OBJECTIVES: The objective of this study was to identify the underlying immune mechanisms involved in formation of inflammatory granulomas seen during active TB. METHODS: The immune mediators involved in inflammatory granuloma formation during TB were assessed using human samples and experimental models of Mycobacterium tuberculosis infection, using molecular and immunologic techniques. MEASUREMENTS AND MAIN RESULTS: We demonstrate that in human patients with active TB and in nonhuman primate models of M. tuberculosis infection, neutrophils producing S100 proteins are dominant within the inflammatory lung granulomas seen during active TB. Using the mouse model of TB, we demonstrate that the exacerbated lung inflammation seen as a result of neutrophilic accumulation is dependent on S100A8/A9 proteins. S100A8/A9 proteins promote neutrophil accumulation by inducing production of proinflammatory chemokines and cytokines, and influencing leukocyte trafficking. Importantly, serum levels of S100A8/A9 proteins along with neutrophil-associated chemokines, such as keratinocyte chemoattractant, can be used as potential surrogate biomarkers to assess lung inflammation and disease severity in human TB. CONCLUSIONS: Our results thus show a major pathologic role for S100A8/A9 proteins in mediating neutrophil accumulation and inflammation associated with TB. Thus, targeting specific molecules, such as S100A8/A9 proteins, has the potential to decrease lung tissue damage without impacting protective immunity against TB.


Subject(s)
Calgranulin A/immunology , Calgranulin B/immunology , Granuloma, Respiratory Tract/immunology , Inflammation Mediators/immunology , Neutrophils/immunology , Tuberculosis, Pulmonary/immunology , Animals , Chemokines/immunology , Chemotactic Factors/immunology , Cytokines/immunology , Disease Models, Animal , Humans , Macaca mulatta , Mice , Mice, Inbred C57BL
15.
Clin Vaccine Immunol ; 20(8): 1320-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23803902

ABSTRACT

CXCL13 is a constitutively expressed chemokine that controls migration of immune cells to lymphoid follicles. Previously, we found CXCL13 mRNA levels increased in rhesus macaque spleen tissues during AIDS. This led us to examine the levels and locations of CXCL13 by detailed in situ methods in cynomolgus macaque lymphoid and intestinal tissues. Our results revealed that there were distinct localization patterns of CXCL13 mRNA compared to protein in germinal centers. These patterns shifted during the course of simian immunodeficiency virus (SIV) infection, with increased mRNA expression within and around follicles during AIDS compared to uninfected or acutely infected animals. Unexpectedly, CXCL13 expression was also found in abundance in Paneth cells in crypts throughout the small intestine. Therefore, we expanded our analyses to include chemokines and antimicrobial peptides (AMPs) not previously demonstrated to be expressed by Paneth cells in intestinal tissues. We examined the expression patterns of multiple chemokines, including CCL25, as well as α-defensin 6 (DEFA6), ß-defensin 2 (BDEF2), rhesus θ-defensin 1 (RTD-1), and Reg3γ in situ in intestinal tissues. Of the 10 chemokines examined, CXCL13 was unique in its expression by Paneth cells. BDEF2, RTD-1, and Reg3γ were also expressed by Paneth cells. BDEF2 and RTD-1 previously have not been shown to be expressed by Paneth cells. These findings expand our understanding of mucosal immunology, innate antimicrobial defenses, homeostatic chemokine function, and host protective mechanisms against microbial translocation.


Subject(s)
Antimicrobial Cationic Peptides/biosynthesis , Chemokine CXCL13/biosynthesis , Intestinal Mucosa/immunology , Paneth Cells/immunology , Simian Immunodeficiency Virus/immunology , Animals , Gene Expression Profiling , Intestine, Small/immunology , Lymph Nodes/immunology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/immunology , alpha-Defensins/biosynthesis , beta-Defensins/biosynthesis
16.
Lymphat Res Biol ; 11(1): 26-34, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23531182

ABSTRACT

Abstract LYVE-1 is a marker expressed by lymphatic endothelial cells (LECs) that line the lymphatic endothelium. Through studies designed to examine potential changes in expression of LYVE-1 in cynomolgus macaque colon tissues during the course of simian immunodeficiency virus (SIV) infection, we discovered that LYVE-1 was expressed by heterogenous populations of cells. As revealed by in situ hybridization (ISH), LYVE-1 mRNA levels in colon were decreased in macaques with AIDS compared with acutely infected or uninfected macaques. In the submucosal layer of the colon, approximately half of the LYVE-1-expressing cells co-expressed the dendritic cell (DC) marker, DC-SIGN/CD209, and this percentage did not change appreciably during infection. Subsets of cells expressing LYVE-1 also co-expressed macrophage markers, such as CD68 and the macrophage mannose receptor (MMR)/CD206, in both the colon and lymph nodes. LECs, DCs, and macrophages that co-expressed LYVE-1 were observed in colon and lymph node from uninfected, healthy animals as well as in tissues with SIV-driven inflammation. These findings provide further definition of the phenotypic overlap between LECs and antigen presenting cells, reveal the heterogeneity within the population of cells expressing the lymphatic marker LYVE-1, and show that SIV modulates this population of cells in a mucosal surface across which the virus is acquired.


Subject(s)
Cell Adhesion Molecules/immunology , Intestine, Large/immunology , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Vesicular Transport Proteins/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/immunology , Antigens, Differentiation, Myelomonocytic/metabolism , Biomarkers/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Colon/immunology , Colon/metabolism , Colon/virology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/immunology , Endothelium, Lymphatic/metabolism , Fluorescent Antibody Technique , Gene Expression/immunology , Host-Pathogen Interactions/immunology , In Situ Hybridization , Intestine, Large/metabolism , Intestine, Large/virology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/virology , Macaca fascicularis , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mannose Receptor , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Microscopy, Confocal , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
17.
J Exp Med ; 210(3): 551-61, 2013 Mar 11.
Article in English | MEDLINE | ID: mdl-23401489

ABSTRACT

Pulmonary Staphylococcus aureus (SA) infections are a public health concern and a major complication of hyper-IgE syndrome, caused by mutations in STAT3. In contrast to previous findings of skin infection, we observed that clearance of SA from the lung did not require T, B, or NK cells but did require Stat3 activation. Immunohistochemistry showed robust Stat3 phosphorylation in the lung epithelium. We identified that a critical Stat3 target gene in lung epithelium is Reg3g (regenerating islet-derived 3 γ), a gene which is highly expressed in gastrointestinal epithelium but whose role in pulmonary host defense is uncharacterized. Stat3 regulated Reg3g transcription through direct binding at the Reg3g promoter region. Recombinant Reg3γ bound to SA and had both bacteriostatic and bactericidal activity in a dose-dependent fashion. Stat3 inhibition in vivo reduced Reg3g transcripts in the lung, and more importantly, recombinant Reg3γ rescued mice from defective SA clearance. These findings reveal an antibacterial function for lung epithelium through Stat3-mediated induction of Reg3γ.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/immunology , Pneumonia, Staphylococcal/immunology , Proteins/physiology , STAT3 Transcription Factor/physiology , Animals , Cytokine Receptor gp130/physiology , Immunity, Innate , Interleukin-6/biosynthesis , Leukemia Inhibitory Factor/biosynthesis , Lung/immunology , Lymphocytes/immunology , Male , Mice , Mice, Inbred C57BL , Pancreatitis-Associated Proteins
18.
J Acquir Immune Defic Syndr ; 63(4): 428-37, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23429503

ABSTRACT

BACKGROUND: Chemokines provide critical immune cell homing and activation signals that if altered could affect the inflammatory milieu and cellular composition of lymphoid tissues. During HIV-1 and simian immunodeficiency virus (SIV)-infection, the virus triggers an increase in inflammation or activation, leading to immunodeficiency and development of opportunistic infections, such as in the lungs-a massive interface between the host and the environment. METHODS: Chemokine, cytokine, and chemokine receptor expression profiles were determined using real-time reverse transcriptase-polymerase chain reaction and in situ hybridization in hilar lymph nodes (HiLNs) from cynomolgus macaques at different stages after infection with SIV/DeltaB670. Immunostaining of tissue sections and flow cytometric analysis of cryopreserved cells were used to examine cellular compositions of lymph nodes. RESULTS: Interferon-gamma, type 1 chemokine, and cognate chemokine receptor mRNAs were upregulated, whereas type 2 and homeostatic chemokine and chemokine receptor mRNAs were down-regulated in HiLNs after SIV infection. Local SIV and interferon-gamma levels were positively correlated with type 1 chemokine levels but negatively correlated with type 2 and homeostatic chemokine levels. Using in situ hybridization, Pneumocystis carinii rRNA was detected in lung-draining lymph nodes from animals with P. carinii pneumonia. Changes in the cellular composition of HiLNs included decreased proportions of CD4 cells and dendritic cells and increased proportions of CD8, CXCR3, and CCR5 cells. CONCLUSIONS: SIV infection of cynomolgus macaques dramatically alters the cellular homing signals of lung-draining lymph nodes, which correlated with changes in the immune cellular composition. These changes could contribute to the loss of immune function that defines AIDS.


Subject(s)
Chemokines/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Receptors, Chemokine/metabolism , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Forkhead Transcription Factors/metabolism , Interferon-gamma/metabolism , Lung , Macaca fascicularis , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Pneumocystis carinii/genetics , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , RNA, Ribosomal/metabolism , T-Lymphocyte Subsets/immunology
19.
J Clin Invest ; 123(2): 712-26, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23281399

ABSTRACT

One third of the world's population is infected with Mycobacterium tuberculosis (Mtb). Although most infected people remain asymptomatic, they have a 10% lifetime risk of developing active tuberculosis (TB). Thus, the current challenge is to identify immune parameters that distinguish individuals with latent TB from those with active TB. Using human and experimental models of Mtb infection, we demonstrated that organized ectopic lymphoid structures containing CXCR5+ T cells were present in Mtb-infected lungs. In addition, we found that in experimental Mtb infection models, the presence of CXCR5+ T cells within ectopic lymphoid structures was associated with immune control. Furthermore, in a mouse model of Mtb infection, we showed that activated CD4+CXCR5+ T cells accumulated in Mtb-infected lungs and produced proinflammatory cytokines. Mice deficient in Cxcr5 had increased susceptibility to TB due to defective T cell localization within the lung parenchyma. We demonstrated that CXCR5 expression in T cells mediated correct T cell localization within TB granulomas, promoted efficient macrophage activation, protected against Mtb infection, and facilitated lymphoid follicle formation. These data demonstrate that CD4+CXCR5+ T cells play a protective role in the immune response against TB and highlight their potential use for future TB vaccine design and therapy.


Subject(s)
Latent Tuberculosis/immunology , Receptors, CXCR5/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Tuberculosis, Pulmonary/immunology , Animals , Cytokines/biosynthesis , Disease Models, Animal , Female , Granuloma, Respiratory Tract/immunology , Humans , Inflammation Mediators/metabolism , Lung/immunology , Lung/microbiology , Lymphocyte Activation , Lymphoid Tissue/immunology , Macrophage Activation , Male , Mice , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/immunology
20.
Cytokine ; 61(3): 924-32, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23360828

ABSTRACT

CCL20 is currently the only known chemokine ligand for the receptor CCR6, and is a mucosal chemokine involved in normal and pathological immune responses. Although nucleotide sequence data are available for ccl20 and ccr6 sequences from multiple species, the ferret ccl20 and ccr6 sequences have not been determined. To increase our understanding of immune function in ferret models of infection and vaccination, we have used RT-PCR to obtain the ferret ccl20 and ccr6 cDNA sequences and functionally characterize the encoded proteins. The open reading frames of both genes were highly conserved across species and mostly closely related to canine sequences. For functional analyses, single cell clones expressing ferret CCR6 were generated, a ferret CCL20/mouse IgG(2a) fusion protein (fCCL20-mIgG(2a)) was produced, and fCCL20 was chemically synthesized. Cell clones expressing ferret CCR6 responded chemotactically to fCCL20-mIgG2a fusion protein and synthetic ferret CCL20. Chemotaxis inhibition studies identified the polyphenol epigallocatechin-3-gallate and the murine γ-herpesvirus 68 M3 protein as inhibitors of fCCL20. Surface plasmon resonance studies revealed that EGCG bound directly to fCCL20. These results provide molecular characterization of previously unreported ferret immune gene sequences and for the first time identify a broad-spectrum small molecule inhibitor of CCL20 and reveal CCL20 as a target for the herpesviral M3 protein.


Subject(s)
Chemokine CCL20/metabolism , Chemotaxis , Ferrets/metabolism , Receptors, CCR6/metabolism , Amino Acid Sequence , Animals , Catechin/analogs & derivatives , Catechin/pharmacology , Chemokine CCL20/chemistry , Chemotaxis/drug effects , Cloning, Molecular , DNA, Complementary/genetics , Dogs , Mice , Molecular Sequence Data , Open Reading Frames/genetics , Phylogeny , Protein Binding/drug effects , Receptors, CCR6/chemistry , Sequence Alignment , Sequence Analysis, DNA , Viral Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL