Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Proc Natl Acad Sci U S A ; 116(13): 6371-6378, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30850538

ABSTRACT

Tuberculosis (TB) remains a leading killer among infectious diseases, and a better TB vaccine is urgently needed. The critical components and mechanisms of vaccine-induced protection against Mycobacterium tuberculosis (Mtb) remain incompletely defined. Our previous studies demonstrate that Vγ2Vδ2 T cells specific for (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP) phosphoantigen are unique in primates as multifunctional effectors of immune protection against TB infection. Here, we selectively immunized Vγ2Vδ2 T cells and assessed the effect on infection in a rhesus TB model. A single respiratory vaccination of macaques with an HMBPP-producing attenuated Listeria monocytogenes (Lm ΔactA prfA*) caused prolonged expansion of HMBPP-specific Vγ2Vδ2 T cells in circulating and pulmonary compartments. This did not occur in animals similarly immunized with an Lm ΔgcpE strain, which did not produce HMBPP. Lm ΔactA prfA* vaccination elicited increases in Th1-like Vγ2Vδ2 T cells in the airway, and induced containment of TB infection after pulmonary challenge. The selective immunization of Vγ2Vδ2 T cells reduced lung pathology and mycobacterial dissemination to extrapulmonary organs. Vaccine effects coincided with the fast-acting memory-like response of Th1-like Vγ2Vδ2 T cells and tissue-resident Vγ2Vδ2 effector T cells that produced both IFN-γ and perforin and inhibited intracellular Mtb growth. Furthermore, selective immunization of Vγ2Vδ2 T cells enabled CD4+ and CD8+ T cells to mount earlier pulmonary Th1 responses to TB challenge. Our findings show that selective immunization of Vγ2Vδ2 T cells can elicit fast-acting and durable memory-like responses that amplify responses of other T cell subsets, and provide an approach to creating more effective TB vaccines.


Subject(s)
Immunization , Lymphocyte Activation/drug effects , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Immunologic Memory/immunology , Interferon-gamma/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Lung/immunology , Lung/pathology , Macaca mulatta/immunology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mycobacterium tuberculosis/drug effects , Organophosphates , Peptide Termination Factors/genetics , Peptide Termination Factors/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Tuberculosis/pathology , Tuberculosis Vaccines/pharmacology , Vaccines, Attenuated/immunology
2.
J Immunol ; 198(12): 4753-4763, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28526681

ABSTRACT

The dominant Vγ2Vδ2 T cell subset recognizes phosphoantigen and exists only in humans and nonhuman primates. Despite the discovery of γδ T cells >30 y ago, a proof-of-concept study has not been done to prove the principle that the Vγ2Vδ2 T cell subset is protective against Mycobacterium tuberculosis and other infections. In this study, we used an adoptive cell-transfer strategy to define the protective role of Vγ2Vδ2 T cells in a primate tuberculosis (TB) model. Vγ2Vδ2 T cells for adoptive transfer displayed central/effector memory and mounted effector functions, including the production of anti-M. tuberculosis cytokines and inhibition of intracellular mycobacteria. They also expressed CXCR3/CCR5/LFA-1 trafficking/tissue-resident phenotypes and consistently trafficked to the airway, where they remained detectable from 6 h through 7 d after adoptive transfer. Interestingly, the test group of macaques receiving transfer of Vγ2Vδ2 T cells at weeks 1 and 3 after high-dose (500 CFU) M. tuberculosis infection exhibited significantly lower levels of M. tuberculosis infection burdens in lung lobes and extrapulmonary organs than did the control groups receiving PBLs or saline. Consistently, adoptive transfer of Vγ2Vδ2 T cells attenuated TB pathology and contained lesions primarily in the infection site of the right caudal lung lobe, with no or reduced TB dissemination to other lobes, spleen, or liver/kidney; in contrast, the controls showed widespread TB dissemination. The proof-of-concept finding supports the view that the dominant Vγ2Vδ2 T cell subset may be included in the rational design of a TB vaccine or host-directed therapy.


Subject(s)
Adoptive Transfer , Mycobacterium tuberculosis/immunology , Phosphoproteins/therapeutic use , Receptors, Antigen, T-Cell, gamma-delta/therapeutic use , T-Lymphocyte Subsets/immunology , Tuberculosis/immunology , Tuberculosis/therapy , Animals , Bacterial Load , Cytokines/biosynthesis , Cytokines/immunology , Immunologic Memory , Lung/immunology , Lung/microbiology , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Macaca fascicularis , Phosphoproteins/administration & dosage , Phosphoproteins/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, CCR5/genetics , Receptors, CCR5/immunology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Tuberculosis/microbiology
3.
J Virol ; 91(14)2017 07 15.
Article in English | MEDLINE | ID: mdl-28356537

ABSTRACT

Despite a great deal of prior research, the early pathogenic events in natural oral poliovirus infection remain poorly defined. To establish a model for study, we infected 39 macaques by feeding them single high doses of the virulent Mahoney strain of wild type 1 poliovirus. Doses ranging from 107 to 109 50% tissue culture infective doses (TCID50) consistently infected all the animals, and many monkeys receiving 108 or 109 TCID50 developed paralysis. There was no apparent difference in the susceptibilities of the three macaque species (rhesus, cynomolgus, and bonnet) used. Virus excretion in stool and nasopharynges was consistently observed, with occasional viremia, and virus was isolated from tonsils, gut mucosa, and draining lymph nodes. Viral replication proteins were detected in both epithelial and lymphoid cell populations expressing CD155 in the tonsil and intestine, as well as in spinal cord neurons. Necrosis was observed in these three cell types, and viral replication in the tonsil/gut was associated with histopathologic destruction and inflammation. The sustained response of neutralizing antibody correlated temporally with resolution of viremia and termination of virus shedding in oropharynges and feces. For the first time, this model demonstrates that early in the infectious process, poliovirus replication occurs in both epithelial cells (explaining virus shedding in the gastrointestinal tract) and lymphoid/monocytic cells in tonsils and Peyer's patches (explaining viremia), extending previous studies of poliovirus pathogenesis in humans. Because the model recapitulates human poliovirus infection and poliomyelitis, it can be used to study polio pathogenesis and to assess the efficacy of candidate antiviral drugs and new vaccines.IMPORTANCE Early pathogenic events of poliovirus infection remain largely undefined, and there is a lack of animal models mimicking natural oral human infection leading to paralytic poliomyelitis. All 39 macaques fed with single high doses ranging from 107 to 109 TCID50 Mahoney type 1 virus were infected, and many of the monkeys developed paralysis. Virus excretion in stool and nasopharynges was consistently observed, with occasional viremia; tonsil, mesentery lymph nodes, and intestinal mucosa served as major target sites of viral replication. For the first time, this model demonstrates that early in the infectious process, poliovirus replication occurs in both epithelial cells (explaining virus shedding in the gastrointestinal tract) and lymphoid/monocytic cells in tonsils and Peyer's patches (explaining viremia), thereby supplementing historical reconstructions of poliovirus pathogenesis. Because the model recapitulates human poliovirus infection and poliomyelitis, it can be used to study polio pathogenesis, candidate antiviral drugs, and the efficacy of new vaccines.


Subject(s)
Macaca , Poliomyelitis/pathology , Poliovirus/growth & development , Poliovirus/pathogenicity , Animal Structures/virology , Animals , Disease Models, Animal , Epithelial Cells/virology , Feces/virology , Leukocytes/virology , Nasopharynx/virology , Virus Shedding
4.
Proc Natl Acad Sci U S A ; 112(29): E3883-92, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26150504

ABSTRACT

Molecular mechanisms for T-cell immune responses modulated by T cell-inhibitory molecules during tuberculosis (TB) infection remain unclear. Here, we show that active human TB infection up-regulates CD244 and CD244 signaling-associated molecules in CD8(+) T cells and that blockade of CD244 signaling enhances production of IFN-γ and TNF-α. CD244 expression/signaling in TB correlates with high levels of a long noncoding RNA (lncRNA)-BC050410 [named as lncRNA-AS-GSTT1(1-72) or lncRNA-CD244] in the CD244(+)CD8(+) T-cell subpopulation. CD244 signaling drives lncRNA-CD244 expression via sustaining a permissive chromatin state in the lncRNA-CD244 locus. By recruiting polycomb protein enhancer of zeste homolog 2 (EZH2) to infg/tnfa promoters, lncRNA-CD244 mediates H3K27 trimethylation at infg/tnfa loci toward repressive chromatin states and inhibits IFN-γ/TNF-α expression in CD8(+) T cells. Such inhibition can be reversed by knock down of lncRNA-CD244. Interestingly, adoptive transfer of lncRNA-CD244-depressed CD8(+) T cells to Mycobacterium tuberculosis (MTB)-infected mice reduced MTB infection and TB pathology compared with lncRNA-CD244-expressed controls. Thus, this work uncovers previously unidentified mechanisms in which T cell-inhibitory signaling and lncRNAs regulate T-cell responses and host defense against TB infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epigenesis, Genetic , Immunity , RNA, Long Noncoding/genetics , Signal Transduction , Tuberculosis/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD , Apoptosis/drug effects , Apoptosis/genetics , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Chromatin/metabolism , Computational Biology , Conserved Sequence , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic/drug effects , Evolution, Molecular , Gene Knockdown Techniques , Genome, Human , HEK293 Cells , Humans , Immunity/drug effects , Immunity/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Models, Biological , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Receptors, Immunologic , Signal Transduction/drug effects , Signal Transduction/genetics , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/drug effects
5.
Eur J Immunol ; 45(2): 442-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25141829

ABSTRACT

Whether cytokines can influence the adaptive immune response by antigen-specific γδ T cells during infections or vaccinations remains unknown. We previously demonstrated that, during BCG/Mycobacterium tuberculosis (Mtb) infections, Th17-related cytokines markedly upregulated when phosphoantigen-specific Vγ2Vδ2 T cells expanded. In this study, we examined the involvement of Th17-related cytokines in the recall-like responses of Vγ2Vδ2 T cells following Mtb infection or vaccination against TB. Treatment with IL-17A/IL-17F or IL-22 expanded phosphoantigen 4-hydroxy-3-methyl-but-enyl pyrophosphate (HMBPP)-stimulated Vγ2Vδ2 T cells from BCG-vaccinated macaques but not from naïve animals, and IL-23 induced greater expansion than the other Th17-related cytokines. Consistently, Mtb infection of macaques also enhanced the ability of IL-17/IL-22 or IL-23 to expand HMBPP-stimulated Vγ2Vδ2 T cells. When evaluating IL-23 signaling as a prototype, we found that HMBPP/IL-23-expanded Vγ2Vδ2 T cells from macaques infected with Mtb or vaccinated with BCG or Listeria ΔactA prfA*-ESAT6/Ag85B produced IL-17, IL-22, IL-2, and IFN-γ. Interestingly, HMBPP/IL-23-induced production of IFN-γ in turn facilitated IL-23-induced expansion of HMBPP-activated Vγ2Vδ2 T cells. Furthermore, HMBPP/IL-23-induced proliferation of Vγ2Vδ2 T cells appeared to require APC contact and involve the conventional and novel protein kinase C signaling pathways. These findings suggest that Th17-related cytokines can contribute to recall-like expansion and effector function of Ag-specific γδ T cells after infection or vaccination.


Subject(s)
Interleukin-17/immunology , Mycobacterium tuberculosis/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Th17 Cells/immunology , Tuberculosis/prevention & control , Animals , Bacterial Vaccines/administration & dosage , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression , Interferon-gamma/biosynthesis , Interleukin-17/genetics , Interleukin-17/pharmacology , Interleukin-2/biosynthesis , Interleukin-23/pharmacology , Interleukins/pharmacology , Listeria/immunology , Macaca fascicularis , Mycobacterium bovis/immunology , Organophosphates/immunology , Organophosphates/pharmacology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Recombinant Proteins/pharmacology , Th17 Cells/cytology , Th17 Cells/drug effects , Tuberculosis/immunology , Tuberculosis/microbiology , Vaccination , Interleukin-22
6.
J Immunol ; 192(5): 2120-32, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24489088

ABSTRACT

The possibility that CD4(+) T cells can act as "innate-like" cells to contain very early Mycobacterium tuberculosis dissemination and function as master helpers to sustain multiple effector functions of CD8(+) T cells and CD3(-) lymphocytes during development of adaptive immunity against primary tuberculosis (TB) has not been demonstrated. We showed that pulmonary M. tuberculosis infection of CD4-depleted macaques surprisingly led to very early extrapulmonary M. tuberculosis dissemination, whereas CD4 deficiency clearly resulted in rapid TB progression. CD4 depletion during M. tuberculosis infection revealed the ability of CD8(+) T cells to compensate and rapidly differentiate to Th17-like/Th1-like and cytotoxic-like effectors, but these effector functions were subsequently unsustainable due to CD4 deficiency. Whereas CD3(-) non-T lymphocytes in the presence of CD4(+) T cells developed predominant Th22-like and NK-like (perforin production) responses to M. tuberculosis infection, CD4 depletion abrogated these Th22-/NK-like effector functions and favored IL-17 production by CD3(-) lymphocytes. CD4-depleted macaques exhibited no or few pulmonary T effector cells constitutively producing IFN-γ, TNF-α, IL-17, IL-22, and perforin at the endpoint of more severe TB, but they presented pulmonary IL-4(+) T effectors. TB granulomas in CD4-depleted macaques contained fewer IL-22(+) and perforin(+) cells despite the presence of IL-17(+) and IL-4(+) cells. These results implicate a previously unknown innate-like ability of CD4(+) T cells to contain extrapulmonary M. tuberculosis dissemination at very early stage. Data also suggest that CD4(+) T cells are required to sustain multiple effector functions of CD8(+) T cells and CD3(-) lymphocytes and to prevent rapid TB progression during M. tuberculosis infection of nonhuman primates.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Mycobacterium tuberculosis/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Tuberculosis, Pulmonary/immunology , Animals , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/pathology , Macaca fascicularis , Th1 Cells/microbiology , Th1 Cells/pathology , Th17 Cells/microbiology , Th17 Cells/pathology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology
7.
PLoS Pathog ; 9(8): e1003501, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23966854

ABSTRACT

Dominant Vγ2Vδ2 T-cell subset exist only in primates, and recognize phosphoantigen from selected pathogens including M. tuberculosis(Mtb). In vivo function of Vγ2Vδ2 T cells in tuberculosis remains unknown. We conducted mechanistic studies to determine whether earlier expansion/differentiation of Vγ2Vδ2 T cells during Mtb infection could increase immune resistance to tuberculosis in macaques. Phosphoantigen/IL-2 administration specifically induced major expansion and pulmonary trafficking/accumulation of phosphoantigen-specific Vγ2Vδ2 T cells, significantly reduced Mtb burdens and attenuated tuberculosis lesions in lung tissues compared to saline/BSA or IL-2 controls. Expanded Vγ2Vδ2 T cells differentiated into multifunctional effector subpopulations capable of producing anti-TB cytokines IFNγ, perforin and granulysin, and co-producing perforin/granulysin in lung tissue. Mechanistically, perforin/granulysin-producing Vγ2Vδ2 T cells limited intracellular Mtb growth, and macaque granulysin had Mtb-bactericidal effect, and inhibited intracellular Mtb in presence of perforin. Furthermore, phosphoantigen/IL2-expanded Vγ2Vδ2 T effector cells produced IL-12, and their expansion/differentiation led to enhanced pulmonary responses of peptide-specific CD4+/CD8+ Th1-like cells. These results provide first in vivo evidence implicating that early expansion/differentiation of Vγ2Vδ2 T effector cells during Mtb infection increases resistance to tuberculosis. Thus, data support a rationale for conducting further studies of the γδ T-cell-targeted treatment of established TB, which might ultimately help explore single or adjunctive phosphoantigen expansion of Vγ2Vδ2 T-cell subset as intervention of MDR-tuberculosis or HIV-related tuberculosis.


Subject(s)
Interleukin-2/administration & dosage , Lung/immunology , Macaca fascicularis/microbiology , Mycobacterium tuberculosis/physiology , Phosphoproteins/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/cytology , Tuberculosis/prevention & control , Animals , Bronchoalveolar Lavage , Cell Differentiation/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Interleukin-2/pharmacology , Lung/metabolism , Lung/microbiology , Macaca fascicularis/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Tuberculosis/immunology , Tuberculosis/microbiology
8.
PLoS Pathog ; 8(11): e1002984, 2012.
Article in English | MEDLINE | ID: mdl-23144609

ABSTRACT

T-cell immune responses modulated by T-cell immunoglobulin and mucin domain-containing molecule 3 (Tim-3) during Mycobacterium tuberculosis (Mtb) infection in humans remain poorly understood. Here, we found that active TB patients exhibited increases in numbers of Tim-3-expressing CD4(+) and CD8(+) T cells, which preferentially displayed polarized effector memory phenotypes. Consistent with effector phenotypes, Tim-3(+)CD4(+) and Tim-3(+)CD8(+) T-cell subsets showed greater effector functions for producing Th1/Th22 cytokines and CTL effector molecules than Tim-3(-) counterparts, and Tim-3-expressing T cells more apparently limited intracellular Mtb replication in macrophages. The increased effector functions for Tim-3-expressing T cells consisted with cellular activation signaling as Tim-3(+)CD4(+) and Tim-3(+)CD8(+) T-cell subsets expressed much higher levels of phosphorylated signaling molecules p38, stat3, stat5, and Erk1/2 than Tim-3- controls. Mechanistic experiments showed that siRNA silencing of Tim-3 or soluble Tim-3 treatment interfering with membrane Tim-3-ligand interaction reduced de novo production of IFN-γ and TNF-α by Tim-3-expressing T cells. Furthermore, stimulation of Tim-3 signaling pathways by antibody cross-linking of membrane Tim-3 augmented effector function of IFN-γ production by CD4(+) and CD8(+) T cells, suggesting that Tim-3 signaling helped to drive stronger effector functions in active TB patients. This study therefore uncovered a previously unknown mechanism for T-cell immune responses regulated by Tim-3, and findings may have implications for potential immune intervention in TB.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Immunologic Memory , Membrane Proteins/immunology , Th1 Cells/immunology , Tuberculosis/immunology , Female , Hepatitis A Virus Cellular Receptor 2 , Humans , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Male , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/immunology
9.
J Immunol ; 189(3): 1285-93, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22745375

ABSTRACT

Although Listeria monocytogenes can induce systemic infection causing spontaneous abortion, septicemia, and meningitis, studies have not been performed to investigate human anti-L. monocytogenes immune responses, including those of Ag-specific Vγ2Vδ2 T cells, a dominant human γδ T cell subset. L. monocytogenes is the only pathogen known to possess both the mevalonate and non-mevalonate isoprenoid biosynthesis pathways that produce metabolic phosphates or phosphoantigens activating human Vγ2Vδ2 T cells, making it interesting to explore in vivo anti-L. monocytogenes immune responses of Vγ2Vδ2 T cells. In this study, we demonstrated that subclinical systemic L. monocytogenes infection of rhesus macaques via parenteral inoculation or vaccination with an attenuated Listeria strain induced multieffector-functional immune responses of phosphoantigen-specific Vγ2Vδ2 T cells. Subclinical systemic infection and reinfection with attenuated L. monocytogenes uncovered the ability of Vγ2Vδ2 T cells to mount expansion and adaptive or recall-like expansion. Expanded Vγ2Vδ2 T cells could traffic to and accumulate in the pulmonary compartment and intestinal mucosa. Expanded Vγ2Vδ2 T cells could evolve into effector cells producing IFN-γ, TNF-α, IL-4, IL-17, or perforin after L. monocytogenes infection, and some effector Vγ2Vδ2 T cells could coproduce IL-17 and IFN-γ, IL-4 and IFN-γ, or TNF-α and perforin. Surprisingly, in vivo-expanded Vγ2Vδ2 T effector cells in subclinical L. monocytogenes infection could directly lyse L. monocytogenes-infected target cells and inhibit intracellular L. monocytogenes bacteria. Thus, we present the first demonstration, to our knowledge, of multieffector-functional Vγ2Vδ2 T cell responses against L. monocytogenes.


Subject(s)
Bacterial Proteins/immunology , Epitopes, T-Lymphocyte/immunology , Membrane Proteins/immunology , Organophosphates/immunology , Peptide Termination Factors/immunology , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , T-Lymphocyte Subsets/immunology , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Cells, Cultured , Cytotoxicity Tests, Immunologic , Epitopes, T-Lymphocyte/administration & dosage , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/pathology , Listeriosis/prevention & control , Macaca mulatta , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Organophosphates/administration & dosage , Peptide Termination Factors/administration & dosage , Peptide Termination Factors/genetics , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/pathology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
10.
J Immunol ; 188(9): 4278-88, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22474020

ABSTRACT

The possibility that simultaneous expansion of T regulatory cells (Treg) and T effector cells early postinfection can confer some immunological benefits has not been studied. In this study, we tested the hypothesis that early, simultaneous cytokine expansion of Treg and T effector cells in a tissue infection site can allow these T cell populations to act in concert to control tissue inflammation/damage while containing infection. IL-2 treatments early after Mycobacterium tuberculosis infection of macaques induced simultaneous expansion of CD4(+)CD25(+)Foxp3(+) Treg, CD8(+)CD25(+)Foxp3(+) T cells, and CD4(+) T effector/CD8(+) T effector/Vγ2Vδ2 T effector populations producing anti-M. tuberculosis cytokines IFN-γ and perforin, and conferred resistance to severe TB inflammation and lesions. IL-2-expanded Foxp3(+) Treg readily accumulated in pulmonary compartment, but despite this, rapid pulmonary trafficking/accumulation of IL-2-activated T effector populations still occurred. Such simultaneous recruitments of IL-2-expanded Treg and T effector populations to pulmonary compartment during M. tuberculosis infection correlated with IL-2-induced resistance to TB lesions without causing Treg-associated increases in M. tuberculosis burdens. In vivo depletion of IL-2-expanded CD4(+)Foxp3(+) Treg and CD4(+) T effectors during IL-2 treatment of M. tuberculosis-infected macaques significantly reduced IL-2-induced resistance to TB lesions, suggesting that IL-2-expanded CD4(+) T effector cells and Treg contributed to anti-TB immunity. Thus, IL-2 can simultaneously activate and expand T effector cells and Foxp3(+) Treg populations and confer resistance to severe TB without enhancing M. tuberculosis infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Interleukin-2/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes, Regulatory/immunology , Tuberculosis, Pulmonary/immunology , Animals , Forkhead Transcription Factors/immunology , Interferon-gamma/immunology , Macaca fascicularis , Perforin/immunology
11.
J Infect Dis ; 208(2): 260-70, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23568175

ABSTRACT

Whether vaccination against a virus can protect against more virulent coinfection with the virus and additional pathogen(s) remains poorly characterized. Overlapping endemicity of human immunodeficiency virus (HIV) and malaria suggests that HIV/malaria coinfection frequently complicates acute and chronic HIV infection. Here we showed that vaccination of macaques with recombinant Listeria ΔactA prfA* expressing simian/human immunodeficiency virus (SHIV) gag and env elicited Gag- and Env-specific T-cell responses, and protected against life-threatening SHIV-related malaria after SHIV/Plasmodium fragile coinfection. SHIV antigen immunization reduced peak viremia, resisted SHIV/malaria-induced lymphoid destruction, and blunted coinfection-accelerated decline of CD4(+) T-cell counts after SHIV/malaria coinfection. SHIV antigen immunization also weakened coinfection-driven overreactive proinflammatory interferon-γ (IFNγ) responses and led to developing T helper cell 17/22 (Th17/Th22) responses after SHIV/malaria coinfection. The findings suggest that vaccination against AIDS virus can alter patterns of immune responses to the SHIV/malaria coinfection and protect against life-threatening SHIV-related malaria.


Subject(s)
Antigens, Viral/immunology , Coinfection/immunology , HIV Infections/immunology , Malaria/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Coinfection/microbiology , Coinfection/parasitology , Coinfection/prevention & control , Gene Products, env/immunology , Gene Products, gag/immunology , HIV Infections/parasitology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/immunology , Macaca mulatta/immunology , Macaca mulatta/microbiology , Macaca mulatta/virology , Malaria/microbiology , Malaria/prevention & control , Plasmodium/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , T-Lymphocytes, Helper-Inducer/immunology , Vaccination/methods , Vaccines, Synthetic/immunology
12.
J Immunol ; 187(1): 190-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21632708

ABSTRACT

The role of IL-22-producing CD4(+) T cells in intracellular pathogen infections is poorly characterized. IL-22-producing CD4(+) T cells may express some effector molecules on the membrane, and therefore synergize or contribute to antimicrobial effector function. This hypothesis cannot be tested by conventional approaches manipulating a single IL-22 cytokine at genetic and protein levels, and IL-22(+) T cells cannot be purified for evaluation due to secretion nature of cytokines. In this study, we surprisingly found that upon activation, CD4(+) T cells in Mycobacterium tuberculosis-infected macaques or humans could evolve into T effector cells bearing membrane-bound IL-22 after de novo IL-22 production. Membrane-bound IL-22(+) CD4(+) T effector cells appeared to mature in vivo and sustain membrane distribution in highly inflammatory environments during active M. tuberculosis infection. Near-field scanning optical microscopy/quantum dot-based nanoscale molecular imaging revealed that membrane-bound IL-22, like CD3, distributed in membrane and engaged as ∼100-200 nm nanoclusters or ∼300-600 nm nanodomains for potential interaction with IL-22R. Importantly, purified membrane-bound IL-22(+) CD4(+) T cells inhibited intracellular M. tuberculosis replication in macrophages. Our findings suggest that IL-22-producing T cells can evolve to retain IL-22 on membrane for prolonged IL-22 t(1/2) and to exert efficient cell-cell interaction for anti-M. tuberculosis effector function.


Subject(s)
Anti-Bacterial Agents/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Interleukins/metabolism , Membrane Proteins/metabolism , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Cell Differentiation/immunology , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interleukins/biosynthesis , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Intracellular Fluid/microbiology , Lymphocyte Activation/immunology , Macaca mulatta , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Membrane Proteins/biosynthesis , Mycobacterium tuberculosis/growth & development , Protein Binding/immunology , Protein Transport/immunology , Tuberculosis, Pulmonary/pathology , Interleukin-22
13.
PLoS Pathog ; 6(2): e1000789, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20195465

ABSTRACT

Differentiation, distribution and immune regulation of human IL-22-producing T cells in infections remain unknown. Here, we demonstrated in a nonhuman primate model that M. tuberculosis infection resulted in apparent increases in numbers of T cells capable of producing IL-22 de novo without in vitro Ag stimulation, and drove distribution of these cells more dramatically in lungs than in blood and lymphoid tissues. Consistently, IL-22-producing T cells were visualized in situ in lung tuberculosis (TB) granulomas by confocal microscopy and immunohistochemistry, indicating that mature IL-22-producing T cells were present in TB granuloma. Surprisingly, phosphoantigen HMBPP activation of Vgamma2Vdelta2 T cells down-regulated the capability of T cells to produce IL-22 de novo in lymphocytes from blood, lung/BAL fluid, spleen and lymph node. Up-regulation of IFNgamma-producing Vgamma2Vdelta2 T effector cells after HMBPP stimulation coincided with the down-regulated capacity of these T cells to produce IL-22 de novo. Importantly, anti-IFNgamma neutralizing Ab treatment reversed the HMBPP-mediated down-regulation effect on IL-22-producing T cells, suggesting that Vgamma2Vdelta2 T-cell-driven IFNgamma-networking function was the mechanism underlying the HMBPP-mediated down-regulation of the capability of T cells to produce IL-22. These novel findings raise the possibility to ultimately investigate the function of IL-22 producing T cells and to target Vgamma2Vdelta2 T cells for balancing potentially hyper-activating IL-22-producing T cells in severe TB.


Subject(s)
Interleukins/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Tuberculosis/immunology , Animals , Antigens, Bacterial/immunology , Cell Differentiation/immunology , Cell Separation , Flow Cytometry , Immunohistochemistry , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukins/biosynthesis , Lymphocyte Activation/immunology , Macaca , Microscopy, Confocal , Mycobacterium tuberculosis/immunology , Organophosphates/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Tuberculosis/metabolism , Interleukin-22
14.
J Immunol ; 185(7): 3940-7, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20805423

ABSTRACT

Clonal responses of Mycobacterium tuberculosis-specific CD4(+) or CD8(+) T effector cells producing antituberculosis cytokine IFN-γ in the context of immune protection against tuberculosis remain poorly characterized in humans. Utilizing decade-long TCR expertise, we previously developed a useful method to isolate clonotypic TCR sequences from Ag-specific IFN-γ-producing T cells and to specifically measure clonotypic TCR frequencies in the T cell pool. In this study, we investigated TCR Vß repertoires/CDR3 usage, clonal expansion or dominance, and pulmonary trafficking or accumulation for purified protein deritative (PPD)-specific T effector cells producing IFN-γ during bacillus Calmette-Guérin (BCG) vaccination and subsequent M. tuberculosis challenge of macaques. We found that while PPD-specific CD4(+) and CD8(+) T effector clones employed diverse TCR Vß repertoires, 30-33% of IFN-γ(+)CD4(+) T cell clones from three M. tuberculosis-infected macaques expressed TCR bearing a conserved residue leucine in CDR3. Many Ag-specific IFN-γ(+) CD4(+) and few CD8(+) T effector cells emerged as dominant clones during mycobacterial infections and underwent major recall expansion after pulmonary M. tuberculosis infection of BCG-vaccinated macaques. PPD-specific T cell clones readily trafficked to the airway or lung after BCG vaccination or M. tuberculosis infection, and some of them continuously accumulated in lungs during M. tuberculosis infection even after they became undetectable in the circulation. Importantly, remarkable recall expansion and pulmonary accumulation of T effector cells coincided with BCG-induced protection against tuberculosis. Thus, rapid clonal expansion and pulmonary accumulation of Ag-specific T effector cells appear to be one of the immune mechanisms underlying immunity against tuberculosis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte/immunology , Genes, T-Cell Receptor , Receptors, Antigen, T-Cell/genetics , Tuberculosis, Pulmonary/immunology , Amino Acid Sequence , Animals , BCG Vaccine/immunology , Cell Separation , Flow Cytometry , Macaca mulatta , Molecular Sequence Data , Mycobacterium tuberculosis/immunology , Reverse Transcriptase Polymerase Chain Reaction/methods , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/prevention & control
15.
Proc Natl Acad Sci U S A ; 106(18): 7553-8, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19383786

ABSTRACT

The possibility that Vgamma2Vdelta2 T effector cells can confer protection against pulmonary infectious diseases has not been tested. We have recently demonstrated that single-dose (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP) plus IL-2 treatment can induce prolonged accumulation of Vgamma2Vdelta2 T effector cells in lungs. Here, we show that a delayed HMBPP/IL-2 administration after inhalational Yersinia pestis infection induced marked expansion of Vgamma2Vdelta2 T cells but failed to control extracellular plague bacterial replication/infection. Surprisingly, despite the absence of infection control, expansion of Vgamma2Vdelta2 T cells after HMBPP/IL-2 treatment led to the attenuation of inhalation plague lesions in lungs. Consistently, HMBPP-activated Vgamma2Vdelta2 T cells accumulated and localized in pulmonary interstitials surrounding small blood vessels and airway mucosa in the lung tissues with no or mild plague lesions. These infiltrating Vgamma2Vdelta2 T cells produced FGF-7, a homeostatic mediator against tissue damages. In contrast, control macaques treated with glucose plus IL-2 or glucose alone exhibited severe hemorrhages and necrosis in most lung lobes, with no or very few Vgamma2Vdelta2 T cells detectable in lung tissues. The findings are consist with the paradigm that circulating Vgamma2Vdelta2 T cells can traffic to lungs for homeostatic protection against tissue damages in infection.


Subject(s)
Interleukin-2/administration & dosage , Lung/immunology , Organophosphates/administration & dosage , Plague/immunology , Pneumonia, Bacterial/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/drug effects , Yersinia pestis , Animals , Cell Movement , Disease Models, Animal , Fibroblast Growth Factor 7/biosynthesis , Homeostasis , Lung/microbiology , Lung/pathology , Macaca , Plague/pathology , Pneumonia, Bacterial/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , T-Lymphocytes/immunology
16.
J Infect Dis ; 204(9): 1450-62, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21921207

ABSTRACT

BACKGROUND: Malaria and AIDS represent 2 leading causes of death from infectious diseases worldwide, and their high geographic overlap means coinfection is prevalent. It remains unknown whether distinct immune responses during coinfection with malaria and human immunodeficiency virus (HIV) affect clinical outcomes. METHODS: We tested this hypothesis by employing macaque models of coinfection with malaria and simian-human immunodeficiency virus (SHIV). RESULTS: Plasmodium fragile malaria coinfection of acutely SHIV-infected macaques induced hyperimmune activation and remarkable expansion of CD4+ and CD8+ T effector cells de novo producing interferon γ or tumor necrosis factor α. Malaria-driven cellular hyperactivation/expansion and high-level Th1-cytokines enhanced SHIV disease characterized by increasing CD4+ T-cell depletion, profound lymphoid depletion or destruction, and even necrosis in lymph nodes and spleens. Importantly, malaria/SHIV-mediated depletion, destruction, and necrosis in lymphoid tissues led to bursting parasite replication and fatal virus-associated malaria. Surprisingly, chronically SHIV-infected macaques without AIDS employed different defense mechanisms during malaria coinfection, and mounted unique ∼200-fold expansion of interleukin 17+/interleukin 22+ T effectors with profound Th1 suppression. Such remarkable expansion of Th17/Th22 cells and inhibition of Th1 response coincided with development of immunity against fatal virus-associated malaria without accelerating SHIV disease. CONCLUSIONS: These novel findings suggest that virus infection status and selected Th1 or Th17/Th22 responses after malaria/AIDS-virus coinfection correlate with distinct outcomes of virus infection and malaria.


Subject(s)
Malaria/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Disease Models, Animal , Macaca mulatta , Plasmodium/immunology , Plasmodium/pathogenicity , Primate Diseases/immunology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Treatment Outcome
17.
PLoS Pathog ; 5(4): e1000392, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19381260

ABSTRACT

The role of CD8 T cells in anti-tuberculosis immunity in humans remains unknown, and studies of CD8 T cell-mediated protection against tuberculosis in mice have yielded controversial results. Unlike mice, humans and nonhuman primates share a number of important features of the immune system that relate directly to the specificity and functions of CD8 T cells, such as the expression of group 1 CD1 proteins that are capable of presenting Mycobacterium tuberculosis lipids antigens and the cytotoxic/bactericidal protein granulysin. Employing a more relevant nonhuman primate model of human tuberculosis, we examined the contribution of BCG- or M. tuberculosis-elicited CD8 T cells to vaccine-induced immunity against tuberculosis. CD8 depletion compromised BCG vaccine-induced immune control of M. tuberculosis replication in the vaccinated rhesus macaques. Depletion of CD8 T cells in BCG-vaccinated rhesus macaques led to a significant decrease in the vaccine-induced immunity against tuberculosis. Consistently, depletion of CD8 T cells in rhesus macaques that had been previously infected with M. tuberculosis and cured by antibiotic therapy also resulted in a loss of anti-tuberculosis immunity upon M. tuberculosis re-infection. The current study demonstrates a major role for CD8 T cells in anti-tuberculosis immunity, and supports the view that CD8 T cells should be included in strategies for development of new tuberculosis vaccines and immunotherapeutics.


Subject(s)
BCG Vaccine/immunology , CD8-Positive T-Lymphocytes/immunology , Tuberculosis/immunology , Animals , CD8 Antigens/immunology , Disease Models, Animal , Humans , Macaca mulatta , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/physiology , Tuberculosis/prevention & control , Vaccination/veterinary
18.
Blood ; 113(4): 837-45, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-18981295

ABSTRACT

Although Foxp3(+) T regulatory cells (Tregs) are well documented for their ability to suppress various immune cells, T-cell subsets capable of counteracting Tregs have not been demonstrated. Here, we assessed phosphoantigen-activated Vgamma2Vdelta2 T cells for the ability to interplay with Tregs in the context of mycobacterial infection. A short-term IL-2 treatment regimen induced marked expansion of CD4(+)CD25(+)Foxp3(+) T cells and subsequent suppression of mycobacterium-driven increases in numbers of Vgamma2Vdelta2 T cells. Surprisingly, activation of Vgamma2Vdelta2 T cells by adding phosphoantigen Picostim to the IL-2 treatment regimen down-regulated IL-2-induced expansion of CD4(+)CD25(+)Foxp3(+) T cells. Consistently, in vitro activation of Vgamma2Vdelta2 T cells by phosphoantigen plus IL-2 down-regulated IL-2-induced expansion of CD4(+)CD25(+)Foxp3(+) T cells. Interestingly, anti-IFN-gamma-neutralizing antibody, not anti-TGF-beta or anti-IL-4, reduced the ability of activated Vgamma2Vdelta2 T cells to down-regulate Tregs, suggesting that autocrine IFN-gamma and its network contributed to Vgamma2Vdelta2 T cells' antagonizing effects. Furthermore, activation of Vgamma2Vdelta2 T cells by Picostim plus IL-2 treatment appeared to reverse Treg-driven suppression of immune responses of phosphoantigen-specific IFNgamma(+) or perforin(+) Vgamma2Vdelta2 T cells and PPD-specific IFNgamma(+)alphabeta T cells. Thus, phos-phoantigen activation of Vgamma2Vdelta2 T cells antagonizes IL-2-induced expansion of Tregs and subsequent suppression of Ag-specific antimicrobial T-cell responses in mycobacterial infection.


Subject(s)
Forkhead Transcription Factors/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2/pharmacology , Mycobacterium Infections/immunology , Phosphoproteins/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , Bacterial Proteins/immunology , CD4 Antigens/immunology , Cell Proliferation/drug effects , Cell Separation , Cells, Cultured , Down-Regulation/drug effects , Interferon-gamma/immunology , Interleukin-4/immunology , Macaca fascicularis , Mycobacterium bovis/immunology , Peptide Fragments/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes, Regulatory/immunology , Time Factors , Transforming Growth Factor beta/immunology
19.
J Immunol ; 182(2): 811-9, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19124724

ABSTRACT

The possibility that mycobacterial infections induce variant cytokine mRNA encoding a functionally distinct protein for immune regulation has not been addressed. In this study, we reported that Mycobacterium tuberculosis and bacillus Calmette-Guérin infections of macaques induced expression of variant IL-4 (VIL-4) mRNA encoding a protein comprised of N-terminal 97 aa identical with IL-4, and unique C-terminal 96 aa including a signaling-related proline-rich motif. While VIL-4 could be stably produced as intact protein, the purified VIL-4 induced apparent expansion of phosphoantigen (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP)-specific Vgamma2Vdelta2 T cells in dose- and time-dependent manners. The unique C-terminal 96 aa bearing the proline-rich motif (PPPCPP) of VIL-4 appeared to confer the ability to expand Vgamma2Vdelta2 T cells, since simultaneously produced IL-4 had only a subtle effect on these gammadelta T cells. Moreover, VIL-4 seemed to use IL-4R alpha for signaling and activation, as the VIL-4-induced expansion of Vgamma2Vdelta2 T cells was blocked by anti-IL-4R alpha mAb but not anti-IL-4 mAb. Surprisingly, VIL-4-expanded Vgamma2Vdelta2 T cells after HMBPP stimulation appeared to be heterologous effector cells capable of producing IL-4, IFN-gamma, and TNF-alpha. Thus, mycobacterial infections of macaques induced variant mRNA encoding VIL-4 that functions as growth factor promoting expansion of HMBPP-specific Vgamma2Vdelta2 T effector cells.


Subject(s)
Diphosphates/immunology , Intercellular Signaling Peptides and Proteins/genetics , Interleukin-4/genetics , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Proliferation , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/physiology , Interleukin-4/biosynthesis , Interleukin-4/isolation & purification , Interleukin-4/physiology , Lymphocyte Activation/immunology , Macaca fascicularis , Macaca mulatta , Molecular Sequence Data , RNA Splicing/immunology , RNA, Messenger/biosynthesis , T-Lymphocyte Subsets/microbiology
20.
J Virol ; 83(22): 11959-65, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19740988

ABSTRACT

Little is known about the in vivo kinetics of T-cell responses in smallpox/monkeypox. We showed that macaque Vgamma2Vdelta2 T cells underwent 3-week-long expansion after smallpox vaccine immunization and displayed simple reexpansion in association with sterile anti-monkeypox virus (anti-MPV) immunity after MPV challenge. Virus-activated Vgamma2Vdelta2 T cells exhibited gamma interferon-producing effector function after phosphoantigen stimulation. Surprisingly, like alphabeta T cells, suboptimally primed Vgamma2Vdelta2 T cells in vaccinia virus/cidofovir-covaccinated macaques mounted major recall-like expansion after MPV challenge. Finally, Vgamma2Vdelta2 T cells localized in inflamed lung tissues for potential regulation. Our studies provide the first in vivo evidence that viruses, despite their inability to produce exogenous phosphoantigen, can induce expansion, reexpansion, and recall-like expansion of Vgamma2Vdelta2 T cells and stimulate their antimicrobial cytokine response.


Subject(s)
Monkeypox virus/immunology , Mpox (monkeypox)/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Smallpox Vaccine/pharmacology , T-Lymphocyte Subsets/immunology , Animals , Antiviral Agents/pharmacology , Cidofovir , Cytosine/analogs & derivatives , Cytosine/pharmacology , Female , Interferon-gamma/pharmacology , Lung/immunology , Lymphocyte Activation , Macaca fascicularis , Organophosphonates/pharmacology , Smallpox Vaccine/immunology , Vaccinia virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL