Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Scand J Immunol ; 98(3): e13311, 2023 Sep.
Article En | MEDLINE | ID: mdl-38112131

This is a report from a one-week workshop held in Athens, Greece in July of 2022. The workshop aimed to identify emerging concepts relevant to the fundamentals of immune regulation and areas for future research. Theories of immune regulation emphasize the role of T cell help or co-stimulation (signal 2). The workshop participants considered how new data on the characteristics of agonist antigens, the role of the antigen receptor signals (signal 1) in driving fate decisions, the effect of energetics on immunity and a better understanding of class-control in the immune response, may impact theories of immune regulation. These ideas were discussed in the context of tumour immunology, autoimmunity, pregnancy and transplantation. Here we present the discussions as a narrative of different viewpoints to allow the reader to join the conversation. These discussions highlight the evolving understanding of the nature of specific antigen recognition and how both antigen-specific and non-specific mechanisms impact immune responses.


Antigens , T-Lymphocytes , Humans , Autoimmunity
2.
Cancer Immunol Immunother ; 72(1): 257-264, 2023 Jan.
Article En | MEDLINE | ID: mdl-35753001

Anti-CD19-CAR-T cells are a successful clinical immunotherapy for B cell lymphomas, although some lymphomas can escape attack by downregulating surface CD19 levels. An undesirable consequence of this therapy is that it can also eliminate healthy B cells expressing CD19. Therefore, understanding the dynamics of CD19 expression in B cells under CAR-T cell immunotherapy can help mitigate both escape and adverse outcomes. Previous studies suggested that mechanisms responsible for the loss of CD19 expression in lymphomas usually involves genomic deletion or epigenetic modification which permanently removes CD19 as a therapeutic target in these cells. We examined if healthy B cells can use similar processes to lose CD19 expression and escape CAR-T attack. In the presence of CAR-T cells, untransformed B cells both when cultured in vitro or in vivo in non-tumor bearing animals downregulate expression of CD19. We then used adoptive transfer strategies to remove CD19-low B cells from αCD19-CAR-T pressure in vivo. Intriguingly, these B cells systematically recovered surface expression of CD19 comparable to wild-type levels. These data suggest that unlike many cases of lymphomas, healthy B cells downregulate CD19 in a reversible fashion. Taken together, these data suggest a dynamic regulatory process of CD19 surface expression on healthy B cells that could be exploited to modulate the expression of CD19 on cancer cells to improve immunotherapy or minimize the depletion of endogenous B cell compartment during treatment.


Antigens, CD19 , Receptors, Chimeric Antigen , Animals , Antigens, CD19/metabolism , Down-Regulation , Immunotherapy , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes , Humans , Mice
3.
Sci Rep ; 12(1): 19487, 2022 11 14.
Article En | MEDLINE | ID: mdl-36376544

Peripheral T cells express a diverse repertoire of antigen-specific receptors, which together protect against the full range of pathogens. In this context, the total repertoire of memory T cells which are maintained by trophic signals, long after pathogen clearance, is critical. Since these trophic factors include cytokines and self-peptide-MHC, both of which are available from endogenous antigen-presenting cells (APC), we hypothesized that enhancing APC numbers in vivo can be a viable strategy to amplify the population of memory T cells. We evaluated this by acutely treating intact mice with FMS-like tyrosine kinase 3 ligand (Flt3l), which promotes expansion of APCs. Here we report that this treatment allowed for, an expansion of effector-memory CD4+ and CD8+ T cells as well as an increase in their expression of KLRG1 and CD25. In the lymph nodes and spleen, the expansion was limited to a specific CD8 (CD44-low but CD62L-) subset. Functionally, this subset is distinct from naïve T cells and could produce significant amounts of effector cytokines upon restimulation. Taken together, these data suggest that the administration of Flt3L can impact both APC turnover as well as a corresponding flux of specific subsets of CD8+ T cells in an intact peripheral immune compartment.


CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Mice , Animals , Spleen/metabolism , Lymphocyte Count , Cytokines/metabolism , Mice, Inbred C57BL , Immunologic Memory
4.
Eur J Immunol ; 52(9): 1431-1440, 2022 09.
Article En | MEDLINE | ID: mdl-35816444

It is commonly believed that IL-12 produced by DCs in response to pathogens is the first signal that stimulates the production of IFN-γ by NK cells. However, IL-12 production by DCs in response to bacterial LPS depends on either engagement of CD40 by CD40L on activated T cells or IFN-γ from NK cells. This suggests that during the primary immune response, NK cells produce IFN-γ before IL-12 production by DCs. Here, using single-cell measurements, cell sorting and mouse lines deficient in IL-12, IL-23, type I IFN receptor and the IL-18 receptor, we show that a subset of BM-derived DCs characterized by low expression of MHC class II (MHCIIlow ) stimulates IFN-γ production by NK cells. The expression of Toll-like Receptor (TLR) 4 on DCs but not NK cells was required for such NK-derived IFN-γ. In addition, soluble factor(s) produced by LPS-activated MHCIIlow DCs were sufficient to induce IFN-γ production by NK cells independent of IL-12, IL-23, and IL-18. This response was enhanced in the presence of a low dose of IL-2. These results delineate a previously unknown pathway of DC-mediated IFN-γ production by NK cells, which is independent of commonly known cytokines.


Interleukin-12 , Interleukin-18 , Animals , Cells, Cultured , Dendritic Cells , Interferon-gamma/metabolism , Interleukin-12/metabolism , Interleukin-18/metabolism , Interleukin-23/metabolism , Lipopolysaccharides/pharmacology , Mice
5.
J Burn Care Res ; 43(4): 792-801, 2022 07 01.
Article En | MEDLINE | ID: mdl-34739051

The World Health Organization estimates ~180,000 deaths occur annually from burn-related injuries. Many victims who survive the initial burn trauma succumb to bacterial infections that lead to sepsis during treatment. Although advancements in burn care continue to improve in high-income countries due to their burn centers and advanced research, low and middle-income countries continue to see high frequencies of burn injuries and burn-related deaths due to secondary infections. Bacterial-derived sepsis is the most life-threatening danger for people that survive burn injuries. Here we provide evidence for the first time that a subeschar seroma forms postburn even in the absence of infection in mice. The seroma fills with a volume estimated at 500 µL of fluid, 25% of the blood supply, free of red blood cells. The seroma fluid supports robust Pseudomonas aeruginosa (PA) growth and contains inflammatory cytokines and chemokines, which recruit immature neutrophils and monocytes to the seroma in the absence of endothelial breakdown. These immune cells fail to contain PA expansion and dissemination. This recruitment of monocytes and immature neutrophils may result in sequestering these critical immune cells away from other tissues during a pivotal time during bacterial dissemination, promoting PA-mediated sepsis.


Burns , Pseudomonas Infections , Sepsis , Soft Tissue Injuries , Animals , Disease Models, Animal , Humans , Mice , Pseudomonas aeruginosa , Sepsis/microbiology , Seroma
6.
Cell Rep ; 37(2): 109816, 2021 10 12.
Article En | MEDLINE | ID: mdl-34644571

Cytokines are typically single gene products, except for the heterodimeric interleukin (IL)-12 family. The two subunits (IL-12p40 and IL-12p35) of the prototype IL-12 are known to be simultaneously co-expressed in activated myeloid cells, which secrete the fully active heterodimer to promote interferon (IFN)γ production in innate and adaptive cells. We find that chimeric mice containing mixtures of cells that can only express either IL-12p40 or IL-12p35, but not both together, generate functional IL-12. This alternate two-cell pathway requires IL-12p40 from hematopoietic cells to extracellularly associate with IL-12p35 from radiation-resistant cells. The two-cell mechanism is sufficient to propel local T cell differentiation in sites distal to the initial infection and helps control systemic dissemination of a pathogen, although not parasite burden, at the site of infection. Broadly, this suggests that early secretion of IL-12p40 monomers by sentinel cells at the infection site may help prepare distal host tissues for potential pathogen arrival.


Dendritic Cells/metabolism , Interleukin-12 Subunit p35/metabolism , Interleukin-12 Subunit p40/metabolism , Leishmania major/pathogenicity , Leishmaniasis, Cutaneous/metabolism , Stromal Cells/metabolism , T-Lymphocytes/metabolism , Animals , Cell Communication , Dendritic Cells/immunology , Dendritic Cells/parasitology , Disease Models, Animal , Female , Host-Parasite Interactions , Interferon-gamma/metabolism , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p40/genetics , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Multimerization , Signal Transduction , Stromal Cells/immunology , Stromal Cells/parasitology , T-Lymphocytes/immunology , T-Lymphocytes/parasitology
7.
Viral Immunol ; 34(6): 365-375, 2021.
Article En | MEDLINE | ID: mdl-33600238

Despite advances in slowing the progression of acquired immunodeficiency syndrome (AIDS), there is no viable cure for human immunodeficiency virus (HIV). The challenge toward a cure is mainly the formation and maintenance of a latent reservoir of cells that harbor the virus in both replication-competent and replication-defective states. This small niche of quiescent cells has been identified to reside primarily in quiescent and memory CD4+ T cells, but parameters that could reliably distinguish an infected T cell from an uninfected one, if any, are not clear. In addition, the migratory properties and specific anatomical reservoirs of latent T cells are difficult to measure at a high resolution in humans. A functional cure of HIV would require targeting this population using innovative new clinical strategies. One constraint toward the empirical development of such approaches is the absence of a native small animal model for AIDS. Since HIV does not efficiently infect murine cells, probing molecular-genetic questions involving latently infected T cells homing to deep tissue sites, interacting with stroma and persisting through different treatment regimens, is challenging. The goal of this article is to discuss how examining the dynamics of T cells in mouse models can provide a framework for effectively studying these questions, even without infecting mice with HIV. The inflammatory and cytokine milieu found in early human HIV infections are being increasingly understood as a result of clinical measurements. Mouse studies that recreate this milieu can potentially be used to subsequently map the fate of T cells activated in this context as well as their migratory routes. In essence, such a framework could allow complementary studies in mice to enhance our understanding of aspects of the biology of HIV latency. This can be the basis of a modular approach to small animal HIV modeling, amenable to preclinical curative strategy development.


HIV Infections , HIV-1 , Animals , CD4-Positive T-Lymphocytes , HIV Infections/drug therapy , Mice , Virus Latency , Virus Replication
8.
Proc Natl Acad Sci U S A ; 117(25): 14342-14353, 2020 06 23.
Article En | MEDLINE | ID: mdl-32513716

Immature T cells undergo a process of positive selection in the thymus when their new T cell receptor (TCR) engages and signals in response to self-peptides. As the T cell matures, a slew of negative regulatory molecules, including the inhibitory surface glycoprotein CD5, are up-regulated in proportion to the strength of the self-peptide signal. Together these regulators dampen TCR-proximal signaling and help avoid any subsequent peripheral activation of T cells by self-peptides. Paradoxically, antigen-specific T cells initially expressing more CD5 (CD5hi) have been found to better persist as effector/memory cells after a peripheral challenge. The molecular mechanisms underlying such a duality in CD5 function is not clear. We found that CD5 alters the basal activity of the NF-κB signaling in resting peripheral T cells. When CD5 was conditionally ablated, T cells were unable to maintain higher expression of the cytoplasmic NF-κB inhibitor IκBα. Consistent with this, resting CD5hi T cells expressed more of the NF-κB p65 protein than CD5lo cells, without significant increases in transcript levels, in the absence of TCR signals. This posttranslationally stabilized cellular NF-κB depot potentially confers a survival advantage to CD5hi T cells over CD5lo ones. Taken together, these data suggest a two-step model whereby the strength of self-peptide-induced TCR signal lead to the up-regulation of CD5, which subsequently maintains a proportional reserve of NF-κB in peripheral T cells poised for responding to agonistic antigen-driven T cell activation.


CD5 Antigens/metabolism , Gene Expression Regulation, Developmental/immunology , NF-KappaB Inhibitor alpha/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antigen Presentation/immunology , CD5 Antigens/genetics , Cell Line, Tumor , Cell Separation , Cell Survival/immunology , Female , Flow Cytometry , Lipopolysaccharides/immunology , Lymphocyte Activation , Mice , Mice, Knockout , Models, Animal , Primary Cell Culture , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Thymus Gland/cytology , Thymus Gland/growth & development , Thymus Gland/immunology , Transcription Factor RelA/metabolism , Up-Regulation
9.
Mol Immunol ; 123: 64-73, 2020 07.
Article En | MEDLINE | ID: mdl-32422416

T cells can help confer protective immunity by eliminating infections and tumors or drive immunopathology by damaging host cells. Both outcomes require a series of steps from the activation of naïve T cells to their clonal expansion, differentiation and migration to tissue sites. In addition to specific recognition of the antigen via the T cell receptor (TCR), multiple accessory signals from costimulatory molecules, cytokines and metabolites also influence each step along the progression of the T cell response. Current efforts to modify effector T cell function in many clinical contexts focus on the latter - which encompass antigen-independent and broad, contextual regulators. Not surprisingly, such approaches are often accompanied by adverse events, as they also affect T cells not relevant to the specific treatment. In contrast, fine tuning T cell responses by precisely targeting antigen-specific TCR signals has the potential to radically alter therapeutic strategies in a focused manner. Development of such approaches, however, requires a better understanding of functioning of the TCR and the biochemical signaling network coupled to it. In this article, we review some of the recent advances which highlight important roles of TCR signals throughout the activation and differentiation of T cells during an immune response. We discuss how, an appreciation of specific signaling modalities and variant ligands that influence the function of the TCR has the potential to influence design principles for the next generation of pharmacologic and cellular therapies, especially in the context of tumor immunotherapies involving adoptive cell transfers.


Adoptive Transfer/methods , Immunity, Cellular/physiology , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigen-Presenting Cells/immunology , Cell Differentiation/immunology , Humans , Immunotherapy/methods , Lymphocyte Activation , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Signal Transduction/immunology
10.
Sci Rep ; 10(1): 8218, 2020 05 19.
Article En | MEDLINE | ID: mdl-32427927

Natural killer T (NKT) cells rapidly respond to antigenic stimulation with cytokine production and direct cytotoxicity. These innate-like characteristics arise from their differentiation into mature effector cells during thymic development. A subset of mature NKT cells remain thymic resident, but their activation and function remain poorly understood. We examined the roles of CD28 and CTLA-4 in driving the activation of thymic resident NKT cells. In contrast to studies with peripheral NKT cells, the proliferation of thymic NKT cells was significantly impaired when CD28 engagement was blocked, but unaffected by CTLA-4 activation or blockade. Within NKT subsets, however, stage 3 NKT cells, marked by higher NK1.1 expression, were significantly more sensitive to the loss of CD28 signals compared to NK1.1- stage 2 NKT cells. In good agreement, CD28 blockade suppressed NKT cell cytokine secretion, lowering the ratio of IFN-γ:IL-4 production by NK1.1+ NKT cells. Intriguingly, the activation-dependent upregulation of the master transcription factor PLZF did not require CD28-costimulation in either of the thymic NKT subsets, underlining a dichotomy between requirements for early activation vs subsequent proliferation and effector function by these cells. Collectively, our studies demonstrate the ability of CD28 co-stimulation to fine tune subset-specific responses by thymic resident NKT cells and contextually shape the milieu in this primary lymphoid organ.


CD28 Antigens/immunology , Natural Killer T-Cells/immunology , Thymus Gland/immunology , Animals , Cell Proliferation , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets
11.
Trends Immunol ; 41(6): 466-480, 2020 06.
Article En | MEDLINE | ID: mdl-32414695

Latent HIV-1 persists indefinitely during antiretroviral therapy (ART) as an integrated silent genome in long-lived memory CD4+ T cells. In untreated infections, immune activation increases the turnover of intrinsically long-lived provirus-containing CD4+ T cells. Those are 'washed out' as a result of their activation, which when coupled to viral protein expression can facilitate local inflammation and recruitment of uninfected cells to activation sites, causing latently infected cells to compete for survival. De novo infection can counter this washout. During ART, inflammation and CD4+ T cell activation wane, resulting in reduced cell turnover and a persistent reservoir. We propose accelerating reservoir washout during ART by triggering sequential waves of polyclonal CD4+ T cell activation while simultaneously enhancing virus protein expression. Reservoir reduction as an adjunct to other therapies might achieve lifelong viral control.


HIV Infections , HIV-1 , CD4-Positive T-Lymphocytes/immunology , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/drug effects , HIV-1/immunology , Humans , Lymphocyte Activation , Virus Latency/drug effects , Virus Latency/immunology
12.
Brain Behav Immun ; 80: 275-285, 2019 08.
Article En | MEDLINE | ID: mdl-30953766

Neurotransmitters are known to modulate the course of an immune response by targeting cells in both the innate and adaptive immune systems. Increasing evidence suggests that T cells, by expressing specific neurotransmitter receptors (NR) are directly regulated by them, leading to altered activation and skewed differentiation of the adaptive immune response. Given that gene expression in T cells changes in lineage- and activation-dependent fashion, it is expected that sensitivity to neurotransmitters may also vary along these lines. Here we generate an important resource for further analysis of this tier of immunoregulation, by identifying the distinct profile of NR transcripts that are expressed by peripheral T cells in mice, at different states of activation and differentiation. We find that only about 15% of the total annotated NR genes are transcribed in these T cells and most of them do not change in different subsets of T cells (CD8, CD4 - Naïve vs Memory vs Treg), or even when T cells migrate to different tissues. We suggest that the T cell-expressed NRs, found across all these subsets identifies a core, constitutive NR signature for the T cell lineage. In contrast, a very limited number (<2) of NRs were observed to mark each of the post-activation T cell states, suggesting that very specific neurotransmitter signals are available to modulate T cell responses in vivo in these subsets.


Lymphocyte Activation , Receptors, Neurotransmitter/metabolism , T-Lymphocytes/metabolism , Animals , Female , Male , Mice, Inbred C57BL , Receptors, Neurotransmitter/genetics , T-Lymphocyte Subsets/metabolism
13.
Immunol Lett ; 192: 61-71, 2017 12.
Article En | MEDLINE | ID: mdl-29111199

Peripheral T cell tolerance is challenging to induce in partially lymphopenic hosts and this is relevant for clinical situations involving transplant tolerance. While the shortage of regulatory cells is thought to be one reason for this, T cell-intrinsic tolerance processes such as anergy are also poorly triggered in such hosts. In order to understand the latter, we used a T cell deficient mouse model system where adoptively transferred autoreactive T cells are significantly tolerized in a cell intrinsic fashion, without differentiation to regulatory T cells. Intriguingly these T cells often retain sufficient effector functions to trigger autoimmune pathology. Here we find that the high population density of the autoreactive T cells that accumulated in such a host limits the progression of the cell-intrinsic tolerance process in T cells. Accordingly, reducing the cell density during a second transfer allowed T cells to further tune down their responsiveness to antigenic stimulation. The retuning of T cells was reflected by a loss of the T cell's abilities to proliferate, produces cytokines or help B cells. We further suggest, based on altering the levels of chronic antigen using miniosmotic pumps, that the effects of cell-density on T cell re-tuning may reflect the effective changes in the antigen dose perceived by individual T cells. This could proportionally elicit more negative feedback downstream of the TCR. Consistent with this, the retuned T cells showed signaling defects both proximal and distal to the TCR. Therefore, similar to the immunogenic activation of T cells, cell-intrinsic T cell tolerance may also involve a quantitative and progressive process of tuning down its antigen-responsiveness. The progress of such tuning seems to be stabilized at multiple intermediate stages by factors such as cell density, rather than just absolute antigen levels.


CD4-Positive T-Lymphocytes/immunology , Lymphopenia/immunology , Peripheral Tolerance , Adoptive Transfer , Animals , Autoantigens/immunology , Autoimmunity , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , DNA-Binding Proteins/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
PLoS One ; 12(8): e0182164, 2017.
Article En | MEDLINE | ID: mdl-28771533

The oral cavity is home to unique resident microbial communities whose interactions with host immunity are less frequently studied than those of the intestinal microbiome. We examined the stimulatory capacity and the interactions of two oral bacteria, Porphyromonas gingivalis (P. gingivalis) and Fusobacterium nucleatum (F. nucleatum), on Dendritic Cell (DC) activation, comparing them to the effects of the well-studied intestinal microbe Escherichia coli (E. coli). Unlike F. nucleatum and E. coli, P. gingivalis failed to activate DCs, and in fact silenced DC responses induced by F. nucleatum or E. coli. We identified a variant strain of P. gingivalis (W50) that lacked this immunomodulatory activity. Using biochemical approaches and whole genome sequencing to compare the two substrains, we found a point mutation in the hagA gene. This protein is though to be involved in the alteration of the PorSS/gingipain pathway, which regulates protein secretion into the extracellular environment. A proteomic comparison of the secreted products of the two substrains revealed enzymatic differences corresponding to this phenotype. We found that P. gingivalis secretes gingipain(s) that inactivate several key proinflammatory mediators made by DCs and/or T cells, but spare Interleukin-1 (IL-1) and GM-CSF, which can cause capillary leaks that serve as a source of the heme that P. gingivalis requires for its survival, and GM-CSF, which can cause epithelial-cell growth. Taken together, our results suggest that P. gingivalis has evolved potent mechanisms to modulate its virulence factors and dampen the innate immune response by selectively inactivating most proinflammatory cytokines.


Bacterial Proteins/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate , Porphyromonas gingivalis/immunology , Animals , Antibiosis , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cytokines/analysis , Cytokines/metabolism , DNA, Bacterial/chemistry , DNA, Bacterial/isolation & purification , DNA, Bacterial/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Escherichia coli/genetics , Female , Fusobacterium/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Inflammation Mediators/metabolism , Interleukin-1/analysis , Interleukin-1/metabolism , Lectins/chemistry , Lectins/genetics , Lectins/metabolism , Male , Mice , Mice, Transgenic , Porphyromonas gingivalis/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/microbiology
15.
Pathog Dis ; 74(7)2016 10.
Article En | MEDLINE | ID: mdl-27620200

The adaptive immune system is expected to protect the host from infectious agents and malignancies, while avoiding robust activation against self-peptides. However, T cells are notoriously inept at protection whenever the pathogen or tumor is persistent in the body for longer periods of time. While this has been thought of as an adaptation to limit the immunopathology from continued effector T-cell responses, it is also likely an extension of the T cell's intrinsic mechanisms which evolved to tolerate self-peptides. Here we deliberate on how the need to tolerate self-peptides might stem from a paradoxical requirement-the utility of such molecules in maintaining a diverse repertoire of pathogen-specific memory T cells in the body. Understanding the mechanisms underlying this intriguing nexus, therefore, has the potential to reveal therapeutic strategies not only for improving immune responses to chronic infections and tumors but also the long-term efficacy of vaccines aimed at cellular immune responses.


Autoimmunity , Immunity, Cellular , Immunologic Memory , T-Lymphocyte Subsets/immunology , Animals , Antigens/immunology , Cell Differentiation/immunology , Cytokines/metabolism , Humans , Infections/immunology , Infections/metabolism , Infections/microbiology , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
16.
Cytokine ; 76(1): 53-7, 2015 Nov.
Article En | MEDLINE | ID: mdl-26242928

How dendritic cells (DCs) gather information from the local milieu at a site of infection or injury and communicate this to influence adaptive immunity is not well understood. We and others have reported that soon after microbial encounter, DCs secrete the p40 subunit of IL-12, by itself, in a monomeric form. Based on recent data that this p40 monomer subsequently associates with p35 released from other cells to generate functional IL-12, we proposed that p40 can function as a DC-derived probe which samples the composition of the local milieu by looking for other binding partners. In this opinion, we discuss how such a sampling function might generate an elaborate combinatorial "code" of heterodimeric cytokines, capable of conveying location-specific information to cells downstream of DC activation, including NK and T cells.


Interleukin-12 Subunit p40 , Protein Multimerization , Animals , Cytokines/chemistry , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Humans , Interleukin-12/immunology , Interleukin-12 Subunit p40/chemistry , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Lymphocyte Activation , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
J Immunol ; 192(12): 6028-36, 2014 Jun 15.
Article En | MEDLINE | ID: mdl-24821971

IL-12p40 partners with the p35 and p19 polypeptides to generate the heterodimeric cytokines IL-12 and IL-23, respectively. These cytokines play critical and distinct roles in host defense. The assembly of these heterodimers is thought to take place within the cell, resulting in the secretion of fully functional cytokines. Although the p40 subunit alone can also be rapidly secreted in response to inflammatory signals, its biological significance remains unclear. In this article, we show that the secreted p40 monomer can generate de novo IL-12-like activities by combining extracellularly with p35 released from other cells. Surprisingly, an unbiased proteomic analysis reveals multiple such extracellular binding partners for p40 in the serum of mice after an endotoxin challenge. We biochemically validate the binding of one of these novel partners, the CD5 Ag-like glycoprotein, to the p40 monomer. Nevertheless, the assembled p40-CD5L heterodimer does not recapitulate the biological activity of IL-12. These findings underscore the plasticity of secreted free p40 monomer, suggesting that p40 functions as an adaptor that is able to generate multiple de novo composites in combination with other locally available polypeptide partners after secretion.


Apoptosis Regulatory Proteins/immunology , Dimerization , Interleukin-12/immunology , Receptors, Immunologic/immunology , Animals , Apoptosis Regulatory Proteins/genetics , CD5 Antigens/genetics , CD5 Antigens/immunology , Interleukin-12/genetics , Mice , Mice, Knockout , Proteomics , Receptors, Immunologic/genetics , Receptors, Scavenger
18.
Immunity ; 37(4): 735-46, 2012 Oct 19.
Article En | MEDLINE | ID: mdl-23021952

After an immune response, the expanded population of antigen-specific CD4(+) T cells contract to steady state levels. We have found that the contraction is neither cell-autonomous nor mediated by competition for generic trophic factors, but regulated by relatively rare subsets of neighboring CD4(+) T cells not necessarily of a conventional regulatory T cell lineage. These regulators, referred to as deletors, specifically limit the frequency of particular antigen-specific T cells even though they are not reactive to the same agonist as their targets. Instead, an isolated deletor could outcompete the target for recognition of a shared, nonstimulatory endogenous peptide-MHC ligand. This mechanism was sufficient to prevent even agonist-driven autoimmune disease in a lymphopenic environment. Such a targeted regulation of homeostasis within narrow colonies of T cells with related TCR specificities for subthreshold ligands might help to prevent the loss of unrelated TCRs during multiple responses, preserving the valuable diversity of the repertoire.


Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , T-Cell Antigen Receptor Specificity , Animals , Autoimmunity , Cell Survival , Cells, Cultured , Ligands , Lymphopenia/immunology , Mice , Receptors, Antigen, T-Cell/immunology
19.
Eur J Immunol ; 42(11): 2875-80, 2012 Nov.
Article En | MEDLINE | ID: mdl-22806568

The behavior of self-reactive T cells in the peripheral immune system has often been studied by following the fate of adoptively transferred antigen-specific T cells in antigen expressing mice. In most cases, after a period of expansion, such cells undergo a slow clonal deletion, accompanied by the onset of anergy and/or suppression in the remaining cells. Here, we demonstrate that at initial frequencies approaching those found in normal repertoires, it is possible to completely avoid deletion and still maintain peripheral tolerance. At starting numbers of <1000 T cells, stimulation by chronic self-antigens resulted in a period of robust clonal expansion, followed by a steady plateau phase extending beyond 4 months. Despite their stable persistence, the self-reactive T cells did not convert to a Foxp3⁺ fate. However, they displayed a considerable block in their ability to make IL-2, consistent with the onset of anergy - in a precursor frequency or deletion independent fashion.


Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Clonal Anergy/immunology , Adoptive Transfer , Animals , Flow Cytometry , Immune Tolerance/immunology , Interleukin-2/immunology , Male , Mice , Mice, Knockout , Mice, Transgenic , Specific Pathogen-Free Organisms
20.
Transplantation ; 94(3): 234-40, 2012 Aug 15.
Article En | MEDLINE | ID: mdl-22797707

BACKGROUND: The hyperacute rejection mediated by preexisting antibodies is a major impediment to the success of transplants across allogeneic and xenogeneic barriers. We report a new mouse model that allows us to not only monitor the sensitization of B cells mediating the hyperacute response but also validate therapeutic strategies for tolerizing them. MODEL: The new model system uses 5C.C7,RAG2 T-cell receptor transgenic T cells and B10.S(9R),CD3[Latin Small Letter Open E] hosts for adoptive transfer experiments. RESULTS AND CONCLUSIONS: In the allogeneic hosts, transgenic T cells expanded briefly before being chronically deleted. Once the deletion was initiated, a second graft of donor cells was used to assess a hyperacute response. The rapid rejection of the second cohort correlated with the appearance of donor-specific antibodies in the serum. Interestingly, chronically stimulated T cells were relatively resistant to hyperacute rejection, suggesting an explanation for the slower rejection kinetics of the first cohort even as the second cohort of identical donor cells was being hyperacutely rejected. Finally, we could tolerize the potential for a hyperacute response, by pretreating recipients with a single infusion of naive donor B cells before the first T-cell transfer. This treatment not only abrogated the development of a hyperacute response but also allowed the primary graft to survive in vivo for extended periods.


Disease Models, Animal , Graft Rejection/immunology , Graft vs Host Reaction , Alleles , Animals , B-Lymphocytes/cytology , Cell Separation , Cohort Studies , Crosses, Genetic , Flow Cytometry , Graft Survival , Mice , Mice, Knockout , Mice, Transgenic , T-Lymphocytes/cytology
...