Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Nat Immunol ; 16(12): 1228-34, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26523867

ABSTRACT

The molecular mechanisms that link the sympathetic stress response and inflammation remain obscure. Here we found that the transcription factor Nr4a1 regulated the production of norepinephrine (NE) in macrophages and thereby limited experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. Lack of Nr4a1 in myeloid cells led to enhanced NE production, accelerated infiltration of leukocytes into the central nervous system (CNS) and disease exacerbation in vivo. In contrast, myeloid-specific deletion of tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine biosynthesis, protected mice against EAE. Furthermore, we found that Nr4a1 repressed autocrine NE production in macrophages by recruiting the corepressor CoREST to the Th promoter. Our data reveal a new role for macrophages in neuroinflammation and identify Nr4a1 as a key regulator of catecholamine production by macrophages.


Subject(s)
Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Macrophages/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/immunology , Sympathetic Nervous System/immunology , Animals , Cell Line , Cells, Cultured , Central Nervous System/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression/immunology , Humans , Inflammation/genetics , Inflammation/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Norepinephrine/immunology , Norepinephrine/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Sympathetic Nervous System/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/immunology , Tyrosine 3-Monooxygenase/metabolism
2.
J Virol ; 94(3)2020 01 17.
Article in English | MEDLINE | ID: mdl-31723023

ABSTRACT

Following a respiratory virus infection, CXCR3hi CX3CR1lo and CXCR3lo CX3CR1hi CD8 T cells localize to different compartments within the lung and play an important role in host resistance, but mechanisms governing their optimal generation are poorly defined. We serendipitously found that B cell-deficient (µMT-/-) mice were highly resistant to lethal infection with a virulent poxvirus strain and that depletion of CD8 T cells rendered these mice susceptible to infection. B cells were not required for the expansion of virus-specific CD8 T cells, but a greater proportion of activated CD8 T cells acquired an effector-like CXCR3lo CX3CR1hi phenotype in the absence of B cells. After recovery from infection, CD8 T cells in µMT-/- mice contracted normally but failed to survive and seed the memory cell pool in both the lungs and spleen. These findings reveal a previously unappreciated role for B cells in regulating the balance between CD8 T cell-mediated resistance against respiratory viral infection and memory cell development.IMPORTANCE B cells play critical role in host resistance against many respiratory viral infections. However, the role of B cells beyond antibody-producing cells is less well defined. In this study, we made a surprising observation that mice lacking B cells were more resistant to respiratory infection with vaccinia virus than wild-type mice. This enhanced resistance was mediated by CD8 T cells because when we depleted CD8 T cells in B cell-deficient mice, these mice were unable to survive the infection. Interestingly, CD8 T cells in B cell-deficient mice were skewed more toward effector phenotype and less toward memory phenotype, which resulted in severely compromised memory CD8 T cell development. Thus, our study shows a novel role of B cells as regulators of CD8 T cell-mediated host resistance and memory CD8 T cell formation during respiratory viral infection.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Virus Diseases/immunology , Animals , CX3C Chemokine Receptor 1/metabolism , Female , Immunologic Memory , Immunotherapy, Adoptive , Lung , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/metabolism , Spleen , Vaccinia virus/immunology
4.
J Immunol ; 200(8): 2894-2904, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29514949

ABSTRACT

The transition of effector T cells or memory precursors into distinct long-lived memory T cell subsets is not well understood. Although many molecules made by APCs can contribute to clonal expansion and effector cell differentiation, it is not clear if clonal contraction and memory development is passive or active. Using respiratory virus infection, we found that CD8 T cells that cannot express the TNF family molecule lymphotoxin-like, exhibits inducible expression, competes with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes (LIGHT) are unimpaired in their initial response and clonally expand to form effector cell pools. Thereafter, LIGHT-deficient CD8 T cells undergo strikingly enhanced clonal contraction with resultant compromised accumulation of both circulating and tissue-resident memory cells. LIGHT expression at the peak of the effector response regulates the balance of several pro- and antiapoptotic genes, including Akt, and has a preferential impact on the development of the peripheral memory population. These results underscore the importance of LIGHT activity in programming memory CD8 T cell development, and suggest that CD8 effector T cells can dictate their own fate into becoming memory cells by expressing LIGHT.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Respiratory Tract Infections/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Virus Diseases/immunology , Animals , Cell Differentiation/immunology , Female , Mice , Mice, Inbred C57BL , Respiratory Tract Infections/virology
5.
J Virol ; 92(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29875235

ABSTRACT

Respiratory infection with vaccinia virus (VacV) elicits robust CD8+ T cell responses that play an important role in host resistance. In the lung, VacV encounters multiple tissue-resident antigen-presenting cell (APC) populations, but which cell plays a dominant role in priming of virus-specific CD8+ effector T cell responses remains poorly defined. We used Batf3-/- mice to investigate the impact of CD103+ and CD8α+ dendritic cell (DC) deficiency on anti-VacV CD8+ T cell responses. We found that Batf3-/- mice were more susceptible to VacV infection, exhibiting profound weight loss, which correlated with impaired accumulation of gamma interferon (IFN-γ)-producing CD8+ T cells in the lungs. This was largely due to defective priming since early in the response, antigen-specific CD8+ T cells in the draining lymph nodes of Batf3-/- mice expressed significantly reduced levels of Ki67, CD25, and T-bet. These results underscore a specific role for Batf3-dependent DCs in regulating priming and expansion of effector CD8+ T cells necessary for host resistance against acute respiratory VacV infection.IMPORTANCE During respiratory infection with vaccinia virus (VacV), a member of Poxviridae family, CD8+ T cells play important role in resolving the primary infection. Effector CD8+ T cells clear the virus by accumulating in the infected lungs in large numbers and secreting molecules such as IFN-γ that kill virally infected cells. However, precise cell types that regulate the generation of effector CD8+ T cells in the lungs are not well defined. Dendritic cells (DCs) are a heterogeneous population of immune cells that are recognized as key initiators and regulators of T-cell-mediated immunity. In this study, we reveal that a specific subset of DCs that are dependent on the transcription factor Batf3 for their development regulate the magnitude of CD8+ T cell effector responses in the lungs, thereby providing protection during pulmonary VacV infection.


Subject(s)
Basic-Leucine Zipper Transcription Factors/analysis , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/chemistry , Dendritic Cells/immunology , Poxviridae Infections/immunology , Repressor Proteins/analysis , Respiratory Tract Infections/immunology , Vaccinia virus/immunology , Animals , Antigens, CD/analysis , Basic-Leucine Zipper Transcription Factors/deficiency , CD8 Antigens/analysis , Integrin alpha Chains/analysis , Mice , Mice, Knockout , Repressor Proteins/deficiency
6.
J Immunol ; 198(1): 218-228, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27895177

ABSTRACT

Cognate interactions between T follicular helper (Tfh) cells and B cells are essential for promoting protective Ab responses. Whereas costimulatory receptors such as ICOS are accepted as being important for the induction of Tfh cell fate decision, other molecules may play key roles in amplifying or maintaining the Tfh phenotype. In this study, with vaccinia virus infection in mice, we show that OX40 was expressed on Tfh cells that accumulated at the T/B borders in the white pulp of the spleen and that OX40-dependent signals directly shaped the magnitude and quality of the their response to viral Ags. OX40 deficiency in Tfh cells profoundly impaired the acquisition of germinal center (GC) B cell phenotype, plasma cell generation, and virus-specific Ab responses. Most significantly, we found that sustained interactions between OX40 and its ligand, OX40L, beyond the time of initial encounter with dendritic cells were required for the persistence of high numbers of Tfh and GC B cells. Interestingly, OX40 was coexpressed with ICOS on Tfh cells in and around the GC, and ICOS-ICOSL interactions were similarly crucial at late times for maintenance of the Tfh and GC B cells. Thus, OX40 and ICOS act in a cooperative, nonredundant manner to maximize and prolong the Tfh response that is generated after acute virus infection.


Subject(s)
Inducible T-Cell Co-Stimulator Protein/immunology , Lymphocyte Activation/immunology , Receptors, OX40/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/immunology , Disease Models, Animal , Female , Flow Cytometry , Fluorescent Antibody Technique , Germinal Center/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , OX40 Ligand , Poxviridae Infections/immunology , Protein Array Analysis , Receptors, OX40/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Tumor Necrosis Factors/immunology , Tumor Necrosis Factors/metabolism , Vaccinia virus
7.
J Immunol ; 199(8): 2968-2975, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28864473

ABSTRACT

Mucosal immunity to reinfection with a highly virulent virus requires the accumulation and persistence of memory CD8 T cells at the site of primary infection. These cells may derive from memory precursor effector cells (MPECs), which are distinct from short-lived effector cells that provide acute protection but are often destined to die. Using respiratory virus infection, we show that herpes virus entry mediator (HVEM; TNFRSF14), a member of the TNF receptor superfamily, provides key signals for MPEC persistence. HVEM-deficient CD8 T cells expanded normally but were skewed away from MPECs with resultant poor development of circulating and lung-resident memory cells. HVEM was selectively expressed on MPECs whereas MPECs deficient in HVEM failed to survive in adoptive transfer recipients. As a consequence, HVEM-deficient recipients failed to afford protection against respiratory reinfection with influenza virus. HVEM therefore represents a critical signal for MPECs and development of protective mucosal CD8 T cell memory.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Precursor Cells, T-Lymphoid/immunology , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/virology , Cell Self Renewal , Cells, Cultured , Disease Models, Animal , Female , Immunity, Mucosal , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Precursor Cells, T-Lymphoid/virology , Receptors, Tumor Necrosis Factor, Member 14/genetics
8.
Immunol Cell Biol ; 96(4): 370-378, 2018 04.
Article in English | MEDLINE | ID: mdl-29363162

ABSTRACT

Phenotypically diverse memory CD8+ T cells are present in the lungs that either re-circulate or reside within the tissue. Understanding the key cellular interactions that regulate the generation and then persistence of these different subsets is of great interest. Recently, DNGR-1+ dendritic cell (DC) mediated priming was reported to control the generation of lung-resident but not circulating memory cells following respiratory viral infection. Here, we report an important role for Ly6C+ inflammatory monocytes (IMs) in contributing to the persistence of memory CD8+ T cells but not their generation. Effector CD8+ T cells expanded and contracted normally in the absence of IMs, but the memory compartment declined significantly over time. Quite unexpectedly, this defect was confined to tissue resident and circulating CXCR3hi CX3CR1lo memory cells but not CXCR3hi CX3CR1int and CXCR3lo CX3CR1hi subsets. Thus, two developmentally distinct innate cells orchestrate the generation and persistence of memory T cell subsets following a respiratory virus infection. See also: News and Commentary by Lafouresse & Groom.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CX3C Chemokine Receptor 1/metabolism , Immunologic Memory , Inflammation/pathology , Lung/immunology , Monocytes/pathology , Receptors, CXCR3/metabolism , Respiratory Tract Infections/virology , Administration, Intranasal , Animals , Cell Proliferation , Female , HeLa Cells , Humans , Immunity, Innate , Lung/pathology , Lung/virology , Mice, Inbred C57BL , Receptors, CCR2/deficiency , Receptors, CCR2/metabolism , Respiratory Tract Infections/immunology , T-Lymphocyte Subsets/immunology , Vaccinia virus/physiology
9.
J Immunol ; 197(2): 458-69, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27296664

ABSTRACT

Pbx1 controls chromatin accessibility to a large number of genes and is entirely conserved between mice and humans. The Pbx1-d dominant-negative isoform is more frequent in CD4(+) T cells from lupus patients than from healthy controls. Pbx1-d is associated with the production of autoreactive T cells in mice carrying the Sle1a1 lupus-susceptibility locus. Transgenic (Tg) expression of Pbx1-d in CD4(+) T cells reproduced the phenotypes of Sle1a1 mice, with increased inflammatory functions of CD4(+) T cells and impaired Foxp3(+) regulatory T cell (Treg) homeostasis. Pbx1-d-Tg expression also expanded the number of follicular helper T cells (TFHs) in a cell-intrinsic and Ag-specific manner, which was enhanced in recall responses and resulted in Th1-biased Abs. Moreover, Pbx1-d-Tg CD4(+) T cells upregulated the expression of miR-10a, miR-21, and miR-155, which were implicated in Treg and follicular helper T cell homeostasis. Our results suggest that Pbx1-d impacts lupus development by regulating effector T cell differentiation and promoting TFHs at the expense of Tregs. In addition, our results identify Pbx1 as a novel regulator of CD4(+) T cell effector function.


Subject(s)
Cell Differentiation/immunology , DNA-Binding Proteins/immunology , Lupus Erythematosus, Systemic/immunology , Proto-Oncogene Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Flow Cytometry , Homeodomain Proteins/immunology , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Polymerase Chain Reaction , Pre-B-Cell Leukemia Transcription Factor 1 , Transcription Factors/immunology
10.
Immunol Cell Biol ; 95(2): 121-128, 2017 02.
Article in English | MEDLINE | ID: mdl-27725666

ABSTRACT

PTPN22 (protein tyrosine phosphatase non receptor 22) encodes a tyrosine phosphatase that functions as a key regulator of immune homeostasis. In particular, PTPN22 inhibits T-cell receptor signaling and selectively promotes type I interferon responses in myeloid cells. To date, there is little information on the CD8 T-cell-intrinsic role of PTPN22 in response to a viral pathogen. We unexpectedly found that PTPN22-deficient virus-specific CD8 T cells failed to accumulate in wild-type hosts after lymphocytic choriomeningitis virus infection. Lack of PTPN22 expression altered CD8 T-cell activation and antiviral cytokine production, but did not significantly affect the composition of effector and memory cell precursors. Most significantly, in vivo, PTPN22-deficient CD8 T cells showed a profound defect in upregulating STAT-1 after lymphocytic choriomeningitis virus infection and considerably less phosphorylation of STAT-1 in response to IFN-α treatment in vitro compared with their wild-type counterparts. In stark contrast, following transfer into lymphopenic mice, CD8 T-cell expansion and central-like phenotype, was considerably increased in the absence of PTPN22. Collectively, our results suggest that PTPN22 has dual roles in T-cell clonal expansion and effector function; whereas it promotes antigen-driven responses during acute infection by positively regulating interferon signaling in T cells, PTPN22 inhibits homeostatic-driven proliferation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Homeostasis , Lymphocytic choriomeningitis virus/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Animals , Biomarkers/metabolism , Cell Proliferation , Cytokines/metabolism , Interferon Type I/metabolism , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Mice, Inbred C57BL , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 22/deficiency , STAT1 Transcription Factor/metabolism , Signal Transduction , Up-Regulation
11.
J Virol ; 90(1): 129-41, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26468539

ABSTRACT

UNLABELLED: In establishing a respiratory infection, vaccinia virus (VACV) initially replicates in airway epithelial cells before spreading to secondary sites of infection, mainly the draining lymph nodes, spleen, gastrointestinal tract, and reproductive organs. We recently reported that interferon gamma (IFN-γ) produced by CD8 T cells ultimately controls this disseminated infection, but the relative contribution of IFN-γ early in infection is unknown. Investigating the role of innate immune cells, we found that the frequency of natural killer (NK) cells in the lung increased dramatically between days 1 and 4 postinfection with VACV. Lung NK cells displayed an activated cell surface phenotype and were the primary source of IFN-γ prior to the arrival of CD8 T cells. In the presence of an intact CD8 T cell compartment, depletion of NK cells resulted in increased lung viral load at the time of peak disease severity but had no effect on eventual viral clearance, disease symptoms, or survival. In sharp contrast, RAG(-/-) mice devoid of T cells failed to control VACV and succumbed to infection despite a marked increase in NK cells in the lung. Supporting an innate immune role for NK cell-derived IFN-γ, we found that NK cell-depleted or IFN-γ-depleted RAG(-/-) mice displayed increased lung VACV titers and dissemination to ovaries and a significantly shorter mean time to death compared to untreated NK cell-competent RAG(-/-) controls. Together, these findings demonstrate a role for IFN-γ in aspects of both the innate and adaptive immune response to VACV and highlight the importance of NK cells in T cell-independent control of VACV in the respiratory tract. IMPORTANCE: Herein, we provide the first systematic evaluation of natural killer (NK) cell function in the lung after infection with vaccinia virus, a member of the Poxviridae family. The respiratory tract is an important mucosal site for entry of many human pathogens, including poxviruses, but precisely how our immune system defends the lung against these invaders remains unclear. Natural killer cells are a type of cytotoxic lymphocyte and part of our innate immune system. In recent years, NK cells have received increasing levels of attention following the discovery that different tissues contain specific subsets of NK cells with distinctive phenotypes and function. They are abundant in the lung, but their role in defense against respiratory viruses is poorly understood. What this study demonstrates is that NK cells are recruited, activated, and contribute to protection of the lung during a severe respiratory infection with vaccinia virus.


Subject(s)
Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Respiratory Tract Infections/immunology , Vaccinia virus/immunology , Vaccinia/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Lung/immunology , Lung/virology , Lymphocyte Depletion , Mice, Inbred C57BL , Mice, Knockout , Ovary/virology , Survival Analysis , Viral Load
13.
J Immunol ; 192(11): 5415-25, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24748494

ABSTRACT

CD8 T cells are a key component of immunity to many viral infections. They achieve this through using an array of effector mechanisms, but precisely which component/s are required for protection against a respiratory orthopox virus infection remains unclear. Using a model of respiratory vaccinia virus infection in mice, we could specifically determine the relative contribution of perforin, TRAIL, and IFN-γ-mediated pathways in protection against virus induced morbidity and mortality. Unexpectedly, we observed that protection against death was mediated by IFN-γ without any involvement of the perforin or TRAIL-dependent pathways. IFN-γ mRNA and protein levels in the lung peaked between days 3 and 6 postinfection. This enhanced response coincided with the emergence of virus-specific CD8 T cells in the lung and the cessation of weight loss. Transfer experiments indicated that CD8 T cell-autonomous expression of IFN-γ restricts virus-induced lung pathology and dissemination to visceral tissues and is necessary for clearance of virus. Most significantly, we show that CD8 T cell-derived IFN-γ is sufficient to protect mice in the absence of CD4 and B-lymphocytes. Thus, our findings reveal a previously unappreciated mechanism by which effector CD8 T cells afford protection against a highly virulent respiratory orthopox virus infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Lung/immunology , Respiratory Tract Diseases/immunology , Vaccinia virus/immunology , Vaccinia/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Interferon-gamma/genetics , Lung/pathology , Mice , Mice, Knockout , Respiratory Tract Diseases/genetics , Respiratory Tract Diseases/pathology , Vaccinia/genetics , Vaccinia/pathology
14.
Proc Natl Acad Sci U S A ; 110(19): 7808-13, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23603272

ABSTRACT

Host innate immune responses to DNA viruses involve members of the nucleotide-binding domain, leucine-rich repeat and pyrin domain containing protein (NLRP) family, which form "inflammasomes" that activate caspase-1, resulting in proteolytic activation of cytokines interleukin (IL)-1ß and IL-18. We hypothesized that DNA viruses would target inflammasomes to overcome host defense. A Vaccinia virus (VACV) B-cell CLL/lymphoma 2 (Bcl-2) homolog, F1L, was demonstrated to bind and inhibit the NLR family member NLRP1 in vitro. Moreover, infection of macrophages in culture with virus lacking F1L (ΔF1L) caused increased caspase-1 activation and IL-1ß secretion compared with wild-type virus. Virulence of ΔF1L virus was attenuated in vivo, causing altered febrile responses, increased proteolytic processing of caspase-1, and more rapid inflammation in lungs of infected mice without affecting cell death or virus replication. Furthermore, we found that a hexapeptide from F1L is necessary and sufficient for inhibiting the NLRP1 inflammasome in vitro, thus identifying a peptidyl motif required for binding and inhibiting NLRP1. The functional importance of this NLRP1-binding motif was further confirmed by studies of recombinant ΔF1L viruses reconstituted either with the wild-type F1L or a F1L mutant that fails to bind NLRP1. Cellular infection with wild-type F1L reconstituted virus-suppressed IL-1ß production, whereas mutant F1L did not. In contrast, both wild-type and mutant versions of F1L equally suppressed apoptosis. In vivo, the NLR nonbinding F1L mutant virus exhibited an attenuated phenotype similar to ΔF1L virus, thus confirming the importance of F1L interactions with NLRP1 for viral pathogenicity in mice. Altogether, these findings reveal a unique viral mechanism for evading host innate immune responses.


Subject(s)
Gene Expression Regulation, Viral , Immunity, Innate , Inflammasomes/metabolism , Vaccinia virus/metabolism , Viral Proteins/metabolism , Amino Acid Motifs , Animals , Caspases/metabolism , Chlorocebus aethiops , Cytokines/metabolism , HEK293 Cells , HeLa Cells , Humans , Interleukin-1beta/metabolism , Mice , Mice, Inbred BALB C , Mutation , Phenotype , Recombinant Proteins/metabolism , Vero Cells , Virulence
15.
Mamm Genome ; 26(5-6): 264-70, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25930071

ABSTRACT

Studies in mice genetically lacking natural killer T (NKT) cells show that these lymphocytes make important contributions to both innate and adaptive immune responses. However, the usefulness of murine models to study human NKT cells is limited by the many differences between mice and humans, including that their NKT cell frequencies, subsets, and distribution are dissimilar. A more suitable model may be swine that share many metabolic, physiological, and growth characteristics with humans and are also similar for NKT cells. Thus, we analyzed genetically modified pigs made deficient for CD1d that is required for the development of Type I invariant NKT (iNKT) cells that express a semi-invariant T-cell receptor (TCR) and Type II NKT cells that use variable TCRs. Peripheral blood analyzed by flow cytometry and interferon-γ enzyme-linked immuno spot assays demonstrated that CD1d-knockout pigs completely lack iNKT cells, while other leukocyte populations remain intact. CD1d and NKT cells have been shown to be involved in shaping the composition of the commensal microbiota in mice. Therefore, we also compared the fecal microbiota profile between pigs expressing and lacking NKT cells. However, no differences were found between pigs lacking or expressing CD1d. Our results are the first to show that knocking-out CD1d prevents the development of NKT cells in a non-rodent species. CD1d-deficient pigs should offer a useful model to more accurately determine the contribution of NKT cells for human immune responses. They also have potential for understanding how NKT cells impact the health of commercial swine.


Subject(s)
Antigens, CD1d/genetics , Antigens, CD1d/immunology , Natural Killer T-Cells/immunology , Animals , Animals, Genetically Modified , Feces/microbiology , Gene Deletion , Natural Killer T-Cells/metabolism , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Sequence Analysis, RNA , Swine/genetics
16.
J Immunol ; 191(10): 5026-35, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24101548

ABSTRACT

T cell costimulation is a key component of adaptive immunity to viral infection but has also been associated with pathology because of excessive or altered T cell activity. We recently demonstrated that the TNFR family costimulatory molecule OX40 (CD134) is critically required to sustain antiviral T cell and Ab responses that enable control of viral replication in the context of chronic lymphocytic choriomeningitis virus (LCMV) infection. In this study, we investigated whether reinforcing OX40 stimulation through an agonist Ab had the potential to prevent LCMV persistence. We observed that anti-OX40 injection early after LCMV clone 13 infection increased CD8 T cell-mediated immunopathology. More strikingly, OX40 stimulation of virus-specific CD4 T cells promoted expression of the transcriptional repressor Blimp-1 and diverted the majority of cells away from follicular Th cell differentiation. This occurred in both acute and chronic infections, and resulted in dramatic reductions in germinal center and Ab responses to the viral infection. The effect of the OX40 agonist was dependent on IL-2 signaling and the timing of OX40 stimulation. Collectively, our data demonstrate that excessive OX40 signaling can result in deleterious consequences in the setting of LCMV infection.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Receptors, OX40/immunology , Transcription Factors/biosynthesis , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Chlorocebus aethiops , Germinal Center/immunology , Interleukin-2/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , Receptors, OX40/metabolism , Signal Transduction/immunology , Vero Cells , Virus Replication
17.
Immunol Rev ; 244(1): 149-68, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22017437

ABSTRACT

The human respiratory tract is an entry point for over 200 known viruses that collectively contribute to millions of annual deaths worldwide. Consequently, the World Health Organization has designated respiratory viral infections as a priority for vaccine development. Despite enormous advances in understanding the attributes of a protective mucosal antiviral immune response, current vaccines continue to fail in effectively generating long-lived protective CD8(+) T-cell immunity. To date, the majority of licensed human vaccines afford protection against infectious pathogens through the generation of specific immunoglobulin responses. In recent years, the selective manipulation of specific costimulatory pathways, which are critical in regulating T cell-mediated immune responses, has generated increasing interest. Impressive results in animal models have shown that the tumor necrosis factor receptor (TNFR) family member OX40 (CD134) and its binding partner OX40L (CD252) are key costimulatory molecules involved in the generation of protective CD8(+) T-cell responses at mucosal surfaces, such as the lung. In this review, we highlight these new findings with a particular emphasis on their potential as immunological adjuvants to enhance poxvirus-based CD8(+) T-cell vaccines.


Subject(s)
Immunity, Cellular , Immunity, Mucosal , Poxviridae Infections , Poxviridae/immunology , Receptors, OX40/immunology , Respiratory System/immunology , Signal Transduction/immunology , Viral Vaccines , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Communication , Gene Expression/immunology , Humans , Immunization , Immunologic Memory , Mice , Mice, Knockout , OX40 Ligand/genetics , OX40 Ligand/immunology , OX40 Ligand/metabolism , Poxviridae/drug effects , Poxviridae/pathogenicity , Poxviridae Infections/immunology , Poxviridae Infections/prevention & control , Poxviridae Infections/virology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, OX40/genetics , Receptors, OX40/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Respiratory System/cytology , Respiratory System/virology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Viral Vaccines/administration & dosage
18.
PLoS Pathog ; 8(9): e1002913, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22969431

ABSTRACT

During acute viral infections, clearance of the pathogen is followed by the contraction of the anti-viral T cell compartment. In contrast, T cell responses need to be maintained over a longer period of time during chronic viral infections in order to control viral replication and to avoid viral spreading. Much is known about inhibitory signals such as through PD-1 that limit T cell activity during chronic viral infection, but little is known about the stimulatory signals that allow maintenance of anti-viral T cells. Here, we show that the co-stimulatory molecule OX40 (CD134) is critically required in the context of persistent LCMV clone 13 infection. Anti-viral T cells express high levels of OX40 in the presence of their cognate antigen and T cells lacking the OX40 receptor fail to accumulate sufficiently. Moreover, the emergence of T cell dependent germinal center responses and LCMV-specific antibodies are severely impaired. Consequently, OX40-deficient mice fail to control LCMV clone 13 infection over time, highlighting the importance of this signaling pathway during persistent viral infection.


Subject(s)
Adaptive Immunity/genetics , Receptors, OX40/physiology , Virus Diseases/immunology , Virus Diseases/prevention & control , Acute Disease , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chlorocebus aethiops , Germinal Center/metabolism , Germinal Center/physiology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, OX40/genetics , Receptors, OX40/metabolism , Vero Cells , Virus Diseases/genetics , Virus Latency/immunology , Virus Latency/physiology
19.
J Immunol ; 189(5): 2432-40, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22826318

ABSTRACT

The precise immune components required for protection against a respiratory Orthopoxvirus infection, such as human smallpox or monkeypox, remain to be fully identified. In this study, we used the virulent Western Reserve strain of vaccinia virus (VACV-WR) to model a primary respiratory Orthopoxvirus infection. Naive mice infected with VACV-WR mounted an early CD8 T cell response directed against dominant and subdominant VACV-WR Ags, followed by a CD4 T cell and Ig response. In contrast to other VACV-WR infection models that highlight the critical requirement for CD4 T cells and Ig, we found that only mice deficient in CD8 T cells presented with severe cachexia, pulmonary inflammation, viral dissemination, and 100% mortality. Depletion of CD8 T cells at specified times throughout infection highlighted that they perform their critical function between days 4 and 6 postinfection and that their protective requirement is critically dictated by initial viral load and virulence. Finally, the ability of adoptively transferred naive CD8 T cells to protect RAG⁻/⁻ mice against a lethal VACV-WR infection demonstrated that they are both necessary and sufficient in protecting against a primary VACV-WR infection of the respiratory tract.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Vaccinia/immunology , Vaccinia/pathology , Animals , CD8-Positive T-Lymphocytes/virology , Disease Models, Animal , Female , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Recovery of Function/immunology , Respiratory Tract Infections/mortality , Severity of Illness Index , Vaccinia/mortality , Vaccinia virus/immunology
20.
J Immunol ; 186(9): 5294-303, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21441451

ABSTRACT

Follicular Th (T(FH)) cells are specialized in provision of help to B cells that is essential for promoting protective Ab responses. CD28/B7 (B7-1 and B7-2) interactions are required for germinal center (GC) formation, but it is not clear if they simply support activation of naive CD4 T cells during initiation of responses by dendritic cells or if they directly control T(FH) cells and/or directly influence follicular B cell differentiation. Using a model of vaccinia virus infection, we show that B7-2 but not B7-1 deficiency profoundly impaired T(FH) cell development but did not affect CD4 T cell priming and Th1 differentiation. Consistent with this, B7-2 but not B7-1 was required for acquisition of GC B cell phenotype, plasma cell generation, and virus-specific neutralizing Ab responses. Mixed adoptive transfer experiments indicated that bidirectional interactions between CD28 expressed on activated T cells and B7-2 expressed on follicular B cells were essential for maintenance of the T(FH) phenotype and GC B cell development. Our data provide new insight into the source and nature of molecules required for T(FH) cells to direct GC B cell responses.


Subject(s)
B-Lymphocytes/immunology , B7-2 Antigen/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccinia/immunology , Adoptive Transfer , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , B7-2 Antigen/biosynthesis , CD28 Antigens/biosynthesis , CD28 Antigens/immunology , Cell Differentiation/immunology , Cell Separation , Female , Flow Cytometry , Germinal Center/cytology , Germinal Center/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/cytology , Plasma Cells/immunology , Spleen/cytology , Spleen/immunology , Vaccinia virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL