Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 20(2): 195-205, 2019 02.
Article in English | MEDLINE | ID: mdl-30643267

ABSTRACT

The developmental programs that generate a broad repertoire of regulatory T cells (Treg cells) able to respond to both self antigens and non-self antigens remain unclear. Here we found that mature Treg cells were generated through two distinct developmental programs involving CD25+ Treg cell progenitors (CD25+ TregP cells) and Foxp3lo Treg cell progenitors (Foxp3lo TregP cells). CD25+ TregP cells showed higher rates of apoptosis and interacted with thymic self antigens with higher affinity than did Foxp3lo TregP cells, and had a T cell antigen receptor repertoire and transcriptome distinct from that of Foxp3lo TregP cells. The development of both CD25+ TregP cells and Foxp3lo TregP cells was controlled by distinct signaling pathways and enhancers. Transcriptomics and histocytometric data suggested that CD25+ TregP cells and Foxp3lo TregP cells arose by coopting negative-selection programs and positive-selection programs, respectively. Treg cells derived from CD25+ TregP cells, but not those derived from Foxp3lo TregP cells, prevented experimental autoimmune encephalitis. Our findings indicate that Treg cells arise through two distinct developmental programs that are both required for a comprehensive Treg cell repertoire capable of establishing immunotolerance.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphoid Progenitor Cells/physiology , T-Lymphocytes, Regulatory/physiology , Thymus Gland/growth & development , Animals , Autoantigens/immunology , Colitis/immunology , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , Humans , Immune Tolerance/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphoid Progenitor Cells/transplantation , Mice , Mice, Transgenic , Mycobacterium tuberculosis/immunology , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Signal Transduction , Specific Pathogen-Free Organisms , Thymus Gland/cytology , Thymus Gland/immunology
2.
Immunity ; 48(4): 745-759.e6, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29669252

ABSTRACT

It is unclear how quiescence is enforced in naive T cells, but activation by foreign antigens and self-antigens is allowed, despite the presence of inhibitory signals. We showed that active transforming growth factor ß (TGF-ß) signaling was present in naive T cells, and T cell receptor (TCR) engagement reduced TGF-ß signaling during T cell activation by downregulating TGF-ß type 1 receptor (TßRI) through activation of caspase recruitment domain-containing protein 11 (CARD11) and nuclear factor κB (NF-κB). TGF-ß prevented TCR-mediated TßRI downregulation, but this was abrogated by interleukin-6 (IL-6). Mitigation of TCR-mediated TßRI downregulation through overexpression of TßRI in naive and activated T cells rendered T cells less responsive and suppressed autoimmunity. Naive T cells in autoimmune patients exhibited reduced TßRI expression and increased TCR-driven proliferation compared to healthy subjects. Thus, TCR-mediated regulation of TßRI-TGF-ß signaling acts as a crucial criterion to determine T cell quiescence and activation.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , CD4-Positive T-Lymphocytes/immunology , Guanylate Cyclase/metabolism , Lymphocyte Activation/immunology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptors, Antigen, T-Cell/immunology , Transforming Growth Factor beta1/metabolism , Animals , Autoimmunity/immunology , CARD Signaling Adaptor Proteins/genetics , Cell Line , Cell Proliferation , Colitis/immunology , Colitis/pathology , Disease Models, Animal , Down-Regulation/immunology , Guanylate Cyclase/genetics , HEK293 Cells , Humans , Interleukin-6/immunology , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Receptor, Transforming Growth Factor-beta Type I/biosynthesis , Signal Transduction/immunology , Transforming Growth Factor beta1/biosynthesis
3.
Proc Natl Acad Sci U S A ; 119(40): e2205062119, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36161903

ABSTRACT

Limiting CD4+ T cell responses is important to prevent solid organ transplant rejection. In a mouse model of costimulation blockade-dependent cardiac allograft tolerance, we previously reported that alloreactive CD4+ conventional T cells (Tconvs) develop dysfunction, losing proliferative capacity. In parallel, induction of transplantation tolerance is dependent on the presence of regulatory T cells (Tregs). Whether susceptibility of CD4+ Tconvs to Treg suppression is modulated during tolerance induction is unknown. We found that alloreactive Tconvs from transplant tolerant mice had augmented sensitivity to Treg suppression when compared with memory T cells from rejector mice and expressed a transcriptional profile distinct from these memory T cells, including down-regulated expression of the transcription factor Special AT-rich sequence-binding protein 1 (Satb1). Mechanistically, Satb1 deficiency in CD4+ T cells limited their expression of CD25 and IL-2, and addition of Tregs, which express higher levels of CD25 than Satb1-deficient Tconvs and successfully competed for IL-2, resulted in greater suppression of Satb1-deficient than wild-type Tconvs in vitro. In vivo, Satb1-deficient Tconvs were more susceptible to Treg suppression, resulting in significantly prolonged skin allograft survival. Overall, our study reveals that transplantation tolerance is associated with Tconvs' susceptibility to Treg suppression, via modulated expression of Tconv-intrinsic Satb1. Targeting Satb1 in the context of Treg-sparing immunosuppressive therapies might be exploited to improve transplant outcomes.


Subject(s)
Graft Survival , Matrix Attachment Region Binding Proteins , T-Lymphocytes, Regulatory , Transcription Factors , Transplantation Tolerance , Animals , Graft Survival/genetics , Graft Survival/immunology , Immunologic Memory/genetics , Interleukin-2/metabolism , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation Tolerance/genetics , Transplantation Tolerance/immunology
4.
Am J Transplant ; 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38519004

ABSTRACT

This study aimed to dissect the relationship between specific gut commensal bacterial subgroups, their functional metabolic pathways, and their impact on skin allograft outcome and alloimmunity. We previously showed that oral broad-spectrum antibiotic (Abx) pretreatment in mice delayed skin, heart, and lung allograft rejection and dampened alloimmune responses. Here, rationally designed Abx combinations targeting major bacterial groups were used to elucidate their individual contribution to modulating alloimmune responses. Abx cocktails targeting intestinal gram-negative, gram-positive, or anaerobic/gram-positive bacteria by oral gavage, all delayed skin allograft rejection, and reduced alloreactive T cell priming to different extents. Notably, the most pronounced extension of skin allograft survival and attenuation of alloimmunity were achieved when all gut bacterial groups were simultaneously targeted. These results suggest a model in which the strength of the alloimmune response is additively tuned up by gut microbial diversity. Shotgun metagenomic sequencing enabled strain-level resolution and identified a shared commensal, Parabacteroides distasonis, as the most enriched following all Abx treatments. Oral administration of P.distasonis to mice harboring a diverse microbiota significantly prolonged skin allograft survival, identifying a probiotic with therapeutic benefit in transplantation.

5.
Am J Transplant ; 24(6): 933-943, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38228228

ABSTRACT

Following solid organ transplantation, small precursor populations of polyclonal CD8+ T cells specific for any graft-expressed antigen preferentially expand their high-affinity clones. This phenomenon, termed "avidity maturation," results in a larger population of CD8+ T cells with increased sensitivity to alloantigen, posing a greater risk for graft rejection. Using a mouse model of minor-mismatched skin transplantation, coupled with the tracking of 2 skin graft-reactive CD8+ T cell receptor-transgenic tracer populations with high and low affinity for the same peptide-major histocompatibility complex, we explored the conventional paradigm that CD8+ T cell avidity maturation occurs through T cell receptor affinity-based competition for cognate antigen. Our data revealed "interclonal CD8-CD8 help," whereby lower/intermediate affinity clones help drive the preferential expansion of their higher affinity counterparts in an interleukin-2/CD25-dependent manner. Consequently, the CD8-helped high-affinity clones exhibit greater expansion and develop augmented effector functions in the presence of their low-affinity counterparts, correlating with more severe graft damage. Finally, interclonal CD8-CD8 help was suppressed by costimulation blockade treatment. Thus, high-affinity CD8+ T cells can leverage help from low-affinity CD8+ T cells of identical specificity to promote graft rejection. Suppressing provision of interclonal CD8-CD8 help may be important to improve transplant outcomes.


Subject(s)
CD8-Positive T-Lymphocytes , Graft Rejection , Mice, Inbred C57BL , Skin Transplantation , Animals , CD8-Positive T-Lymphocytes/immunology , Mice , Graft Rejection/immunology , Isoantigens/immunology , Mice, Transgenic , Mice, Inbred BALB C , Graft Survival/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics
6.
Am J Transplant ; 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38219866

ABSTRACT

Mouse models have been instrumental in understanding mechanisms of transplant rejection and tolerance, but cross-study reproducibility and translation of experimental findings into effective clinical therapies are issues of concern. The Mouse Models in Transplantation symposium gathered scientists and physician-scientists involved in basic and clinical research in transplantation to discuss the strengths and limitations of mouse transplant models and strategies to enhance their utility. Participants recognized that increased procedure standardization, including the use of prespecified, defined endpoints, and statistical power analyses, would benefit the field. They also discussed the generation of new models that incorporate environmental and genetic variables affecting clinical outcomes as potentially important. If implemented, these strategies are expected to improve the reproducibility of mouse studies and increase their translation to clinical trials and, ideally, new Food and Drug Administration-approved drugs.

7.
Radiographics ; 44(3): e230031, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38329903

ABSTRACT

Infective endocarditis (IE) is a complex multisystemic disease resulting from infection of the endocardium, the prosthetic valves, or an implantable cardiac electronic device. The clinical presentation of patients with IE varies, ranging from acute and rapidly progressive symptoms to a more chronic disease onset. Because of its severe morbidity and mortality rates, it is necessary for radiologists to maintain a high degree of suspicion in evaluation of patients for IE. Modified Duke criteria are used to classify cases as "definite IE," "possible IE," or "rejected IE." However, these criteria are limited in characterizing definite IE in clinical practice. The use of advanced imaging techniques such as cardiac CT and nuclear imaging has increased the accuracy of these criteria and has allowed possible IE to be reclassified as definite IE in up to 90% of cases. Cardiac CT may be the best choice when there is high clinical suspicion for IE that has not been confirmed with other imaging techniques, in cases of IE and perivalvular involvement, and for preoperative treatment planning or excluding concomitant coronary artery disease. Nuclear imaging may have a complementary role in prosthetic IE. The main imaging findings in IE are classified according to the site of involvement as valvular (eg, abnormal growths [ie, "vegetations"], leaflet perforations, or pseudoaneurysms), perivalvular (eg, pseudoaneurysms, abscesses, fistulas, or prosthetic dehiscence), or extracardiac embolic phenomena. The differential diagnosis of IE includes evaluation for thrombus, pannus, nonbacterial thrombotic endocarditis, Lambl excrescences, papillary fibroelastoma, and caseous necrosis of the mitral valve. The location of the lesion relative to the surface of the valve, the presence of a stalk, and calcification or enhancement at contrast-enhanced imaging may offer useful clues for their differentiation. ©RSNA, 2024 Test Your Knowledge questions for this article are available in the supplemental material.


Subject(s)
Aneurysm, False , Endocarditis, Bacterial , Endocarditis , Humans , Endocarditis, Bacterial/diagnosis , Endocarditis, Bacterial/microbiology , Endocarditis, Bacterial/pathology , Endocarditis/diagnostic imaging , Tomography, X-Ray Computed/methods , Multimodal Imaging
8.
Am J Transplant ; 23(10): 1526-1535, 2023 10.
Article in English | MEDLINE | ID: mdl-37356668

ABSTRACT

Obesity is associated with dysbiosis and a state of chronic inflammation that contributes to the pathogenesis of metabolic diseases, including diabetes. We have previously shown that obese mice develop glucose intolerance, increased alloreactivity, and accelerated transplant rejection. In the present study, we investigated the influence of the microbiota on diet-induced obesity (DIO)-associated transplant rejection and hyperglycemia. Antibiotic treatment prolonged graft survival and reduced fasting glycemia in high-fat diet (HFD)-fed specific-pathogen-free (SPF) mice, supporting a role for the microbiota in promoting accelerated graft rejection and hyperglycemia induced by DIO. Further supporting a microbiota-dependent effect, fecal microbiota transfer from DIO SPF mice into germ-free mice also accelerated graft rejection when compared with lean mice-fecal microbiota transfer. Notably, HFD could be also detrimental to the graft independently from microbiota, obesity, and hyperglycemia. Thus, whereas HFD-associated hyperglycemia was exclusively microbiota-dependent, HFD affected transplant outcomes via both microbiota-dependent and -independent mechanisms. Importantly, hyperglycemia in DIO SPF mice could be reduced by the addition of the gut commensal Alistipes onderdonkii, which alleviated both HFD-induced inflammation and glucose intolerance. Thus, microbial dysbiosis can be manipulated via antibiotics or select probiotics to counter some of the pathogenic effects of obesity in transplantation.


Subject(s)
Gastrointestinal Microbiome , Glucose Intolerance , Hyperglycemia , Animals , Mice , Graft Rejection/etiology , Glucose Intolerance/complications , Dysbiosis/etiology , Obesity/etiology , Diet, High-Fat/adverse effects , Hyperglycemia/complications , Inflammation/etiology , Mice, Inbred C57BL
9.
Am J Transplant ; 23(2): 272-277, 2023 02.
Article in English | MEDLINE | ID: mdl-36804134

ABSTRACT

Intestinal commensals can exert immunomodulatory effects on the host, with beneficial or detrimental consequences depending on underlying diseases. We have previously correlated longer survival of minor mismatched skin grafts in mice with the presence of an intestinal commensal bacterium, Alistipes onderdonkii. In this study, we investigated its sufficiency and mechanism of action. Oral administration of A onderdonkii strain DSM19147 but not DSM108265 was sufficient to prolong minor mismatched skin graft survival through inhibition of tumor necrosis factor production. Through metabolomic and metagenomic comparisons between DSM19147 and DSM108265, we identified candidate gene products associated with the anti-inflammatory effect of DSM19147. A onderdonkii DSM19147 can lower inflammation both at a steady state and after transplantation and may serve as an anti-inflammatory probiotic beneficial for transplant recipients.


Subject(s)
Bacteroidetes , Graft Survival , Probiotics , Skin Transplantation , Animals , Mice , Administration, Oral , Allografts , Graft Rejection/drug therapy , Graft Rejection/etiology , Graft Rejection/prevention & control , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous , Probiotics/administration & dosage
10.
Immunity ; 41(5): 830-42, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25517615

ABSTRACT

Spontaneous T cell responses against tumors occur frequently and have prognostic value in patients. The mechanism of innate immune sensing of immunogenic tumors leading to adaptive T cell responses remains undefined, although type I interferons (IFNs) are implicated in this process. We found that spontaneous CD8(+) T cell priming against tumors was defective in mice lacking stimulator of interferon genes complex (STING), but not other innate signaling pathways, suggesting involvement of a cytosolic DNA sensing pathway. In vitro, IFN-? production and dendritic cell activation were triggered by tumor-cell-derived DNA, via cyclic-GMP-AMP synthase (cGAS), STING, and interferon regulatory factor 3 (IRF3). In the tumor microenvironment in vivo, tumor cell DNA was detected within host antigen-presenting cells, which correlated with STING pathway activation and IFN-? production. Our results demonstrate that a major mechanism for innate immune sensing of cancer occurs via the host STING pathway, with major implications for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA/immunology , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Membrane Proteins/immunology , Adaptive Immunity , Adaptor Proteins, Signal Transducing/genetics , Adoptive Transfer , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Cell Line, Tumor , Cell Proliferation , Dendritic Cells/immunology , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon-beta/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Nucleotidyltransferases , Receptors, Antigen, T-Cell/immunology , Receptors, Purinergic P2X7/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics , Tumor Microenvironment/immunology
11.
Am J Transplant ; 22(10): 2348-2359, 2022 10.
Article in English | MEDLINE | ID: mdl-35633180

ABSTRACT

Oral antigen exposure is a powerful, non-invasive route to induce immune tolerance to dietary antigens, and has been modestly successful at prolonging graft survival in rodent models of transplantation. To harness the mechanisms of oral tolerance for promoting long-term graft acceptance, we developed a mouse model where the antigen ovalbumin (OVA) was introduced orally prior to transplantation with skin grafts expressing OVA. Oral OVA treatment pre-transplantation promoted permanent graft acceptance and linked tolerance to skin grafts expressing OVA fused to the additional antigen 2W. Tolerance was donor-specific, as secondary donor-matched, but not third-party allografts were spontaneously accepted. Oral OVA treatment promoted an anergic phenotype in OVA-reactive CD4+ and CD8+ conventional T cells (Tconvs) and expanded OVA-reactive Tregs pre-transplantation. However, skin graft acceptance following oral OVA resisted partial depletion of Tregs and blockade of PD-L1. Mechanistically, we revealed a role for the proximal gut draining lymph nodes (gdLNs) in mediating this effect, as an intestinal infection that drains to the proximal gdLNs prevented tolerance induction. Our study extends previous work applying oral antigen exposure to transplantation and serves as proof of concept that the systemic immune mechanisms supporting oral tolerance are sufficient to promote long-term graft acceptance.


Subject(s)
Isoantigens , Skin Transplantation , Animals , Antigens , B7-H1 Antigen , Graft Survival , Immune Tolerance , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin , Transplantation Tolerance
12.
Am J Transplant ; 22(2): 402-413, 2022 02.
Article in English | MEDLINE | ID: mdl-34551205

ABSTRACT

Obesity initiates a chronic inflammatory network linked to perioperative complications and increased acute rejection rates in organ transplantation. Bariatric surgery is the most effective treatment of obesity recommended for morbidly obese transplant recipients. Here, we delineated the effects of obesity and bariatric surgery on alloimmunity and transplant outcomes in diet-induced obese (DIO) mice. Allograft survival was significantly shorter in DIO-mice. When performing sleeve gastrectomies (SGx) prior to transplantation, we found attenuated T cell-derived alloimmune responses resulting in prolonged allograft survival. Administering taurodeoxycholic acid (TDCA) and valine, metabolites depleted in DIO-mice and restored through SGx, prolonged graft survival in DIO-mice comparable with SGx an dampened Th1 and Th17 alloimmune responses while Treg frequencies and CD4+ T cell-derived IL-10 production were augmented. Moreover, in recipient animals treated with TDCA/valine, levels of donor-specific antibodies had been reduced. Mechanistically, TDCA/valine restrained inflammatory M1-macrophage polarization through TGR5 that compromised cAMP signaling and inhibited macrophage-derived T cell activation. Consistently, administering a TGR5 agonist to DIO-mice prolonged allograft survival. Overall, we provide novel insights into obesity-induced inflammation and its impact on alloimmunity. Furthermore, we introduce TDCA/valine as a noninvasive alternative treatment for obese transplant patients.


Subject(s)
Heart Transplantation , Obesity, Morbid , Allografts , Animals , Graft Rejection/etiology , Graft Survival , Heart Transplantation/adverse effects , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Taurodeoxycholic Acid , Valine
13.
Blood ; 135(8): 568-581, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31880771

ABSTRACT

Gastrointestinal (GI) tract involvement is the major cause of morbidity and mortality in acute graft-versus-host disease (GVHD), and pathological damage is largely attributable to inflammatory cytokine production. Recently, granulocyte-macrophage colony stimulating factor (GM-CSF) has been identified as a cytokine that mediates inflammation in the GI tract, but the transcriptional program that governs GM-CSF production and the mechanism by which GM-CSF links adaptive to innate immunity within this tissue site have not been defined. In the current study, we identified Bhlhe40 as a key transcriptional regulator that governs GM-CSF production by CD4+ T cells and mediates pathological damage in the GI tract during GVHD. In addition, we observed that GM-CSF was not regulated by either interleukin 6 (IL-6) or IL-23, which are both potent inducers of GVHD-induced colonic pathology, indicating that GM-CSF constitutes a nonredundant inflammatory pathway in the GI tract. Mechanistically, GM-CSF had no adverse effect on regulatory T-cell reconstitution, but linked adaptive to innate immunity by enhancing the activation of donor-derived dendritic cells in the colon and subsequent accumulation of these cells in the mLNs. In addition, GM-CSF promoted indirect alloantigen presentation, resulting in the accumulation of donor-derived T cells with a proinflammatory cytokine phenotype in the colon. Thus, Bhlhe40+ GM-CSF+ CD4+ T cells constitute a colitogenic T-cell population that promotes indirect alloantigen presentation and pathological damage within the GI tract, positioning GM-CSF as a key regulator of GVHD in the colon and a potential therapeutic target for amelioration of this disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/pathology , Graft vs Host Disease/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Homeodomain Proteins/immunology , Isoantigens/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Colon/immunology , Colon/pathology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Graft vs Host Disease/immunology , Mice, Inbred C57BL
14.
Proc Natl Acad Sci U S A ; 116(47): 23682-23690, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31685610

ABSTRACT

Following antigen stimulation, naïve T cells differentiate into memory cells that mediate antigen clearance more efficiently upon repeat encounter. Donor-specific tolerance can be achieved in a subset of transplant recipients, but some of these grafts are rejected after years of stability, often following infections. Whether T cell memory can develop from a tolerant state and whether these formerly tolerant patients develop antidonor memory is not known. Using a mouse model of cardiac transplantation in which donor-specific tolerance is induced with costimulation blockade (CoB) plus donor-specific transfusion (DST), we have previously shown that systemic infection with Listeria monocytogenes (Lm) months after transplantation can erode or transiently abrogate established tolerance. In this study, we tracked donor-reactive T cells to investigate whether memory can be induced when alloreactive T cells are activated in the setting of tolerance. We show alloreactive T cells persist after induction of cardiac transplantation tolerance, but fail to acquire a memory phenotype despite becoming antigen experienced. Instead, donor-reactive T cells develop T cell-intrinsic dysfunction evidenced when removed from the tolerant environment. Notably, Lm infection after tolerance did not rescue alloreactive T cell memory differentiation or functionality. CoB and antigen persistence were sufficient together but not separately to achieve alloreactive T cell dysfunction, and conventional immunosuppression could substitute for CoB. Antigen persistence was required, as early but not late surgical allograft removal precluded the acquisition of T cell dysfunction. Our results demonstrate transplant tolerance-associated T cell-intrinsic dysfunction that is resistant to memory development even after Lm-mediated disruption of tolerance.


Subject(s)
Graft Survival/immunology , Immune Tolerance/immunology , T-Lymphocyte Subsets/immunology , Transplantation Immunology , Allografts , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Forkhead Transcription Factors/analysis , Genes, Reporter , Graft Rejection/immunology , H-2 Antigens/immunology , Heart Transplantation , Histocompatibility Antigens Class II/immunology , Immunologic Memory , Isoantigens/immunology , Listeria monocytogenes , Listeriosis/immunology , Lymphocyte Transfusion , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Postoperative Complications/immunology , T-Lymphocytes, Regulatory/immunology , Tissue Donors
15.
Cell Immunol ; 351: 104068, 2020 05.
Article in English | MEDLINE | ID: mdl-32139072

ABSTRACT

Cellular metabolism is central to T cell function and proliferation, with most of the research to date focusing on cancer and autoimmunity. Cellular metabolism is associated with a host of physiological phenomena, from epigenetic changes, to cellular function and fate. For the purpose of this review, we will discuss the metabolism of T cells relating to their differentiation and function. We will cover a variety of metabolic processes, ranging from glycolysis to amino acid metabolism. Understanding how T cell metabolism informs T cell function may be useful to understand alloimmune responses and design novel therapies to improve graft outcome.


Subject(s)
Organ Transplantation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation Tolerance/immunology , Animals , Cell Differentiation/immunology , Graft Rejection/immunology , Graft Survival/immunology , Humans
16.
Clin Transplant ; 34(11): e14059, 2020 11.
Article in English | MEDLINE | ID: mdl-32762055

ABSTRACT

An unprecedented global pandemic caused by a novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has quickly overwhelmed the health care systems worldwide. While there is an absence of consensus among the community in how to manage solid organ transplant recipients and donors, a platform provided by the American Society of Transplantation online community "Outstanding Questions in Transplantation," hosted a collaborative multicenter, multinational discussions to share knowledge in a rapidly evolving global situation. Here, we present a summary of the discussion in addition to the latest published literature.


Subject(s)
COVID-19 , Organ Transplantation , Pandemics , Postoperative Complications , SARS-CoV-2 , COVID-19/epidemiology , COVID-19/immunology , COVID-19/therapy , Global Health , Graft Rejection/prevention & control , Humans , Immunocompromised Host , Immunosuppressive Agents/therapeutic use , International Cooperation , Postoperative Complications/epidemiology , Postoperative Complications/immunology , Postoperative Complications/therapy , Societies, Medical
18.
Am J Transplant ; 19(5): 1560-1567, 2019 05.
Article in English | MEDLINE | ID: mdl-30659772

ABSTRACT

Regular exercise reduces risk of various chronic diseases and can prevent the development and recurrence of cancer, making it a promising nonpharmacological modulator of disease. Yet the effect of regular exercise on solid organ transplant outcome remains uncertain. Using a model of voluntary wheel-running exercise and skin transplantation in mice, we hypothesized that exercise strengthens the alloimmune response, leading to an increased rate of rejection. Instead, we found that regular exercise in mice resulted in prolonged graft survival, with mean allograft survival time increasing by almost 50%. We observed this graft survival extension in exercised mice despite evidence of a slightly enhanced alloimmune response, comprised of increased proliferation of alloreactive CD4+ T cells, as well as increased interferon-γ production by these cells. Exercise was not associated with significant changes in numbers of conventional CD4+ or CD8+ T cells, NK cells, or Foxp3+ regulatory T cells. In conclusion, our study suggests that exercise increases skin graft resistance to a similar or slightly higher level of alloimmunity and supports regular exercise as an important beneficial pursuit for transplant recipients.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/prevention & control , Graft Survival/immunology , Physical Conditioning, Animal/methods , Skin Transplantation/adverse effects , T-Lymphocytes, Regulatory/immunology , Animals , Female , Graft Rejection/etiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous
19.
Trends Immunol ; 37(12): 831-843, 2016 12.
Article in English | MEDLINE | ID: mdl-27743777

ABSTRACT

Transplantation of solid organs between genetically distinct individuals leads, in the absence of immunosuppression, to T cell-dependent transplant rejection. Activation of graft-reactive T cells relies on the presentation of transplant-derived antigens (intact donor MHC molecules or processed peptides on host MHC molecules) by mature dendritic cells (DCs). This review focuses on novel insights regarding the steps for maturation and differentiation of DCs that are necessary for productive presentation of transplant antigens to host T cells. These steps include the licensing of DCs by the microbiota, their activation and maturation following recognition of allogeneic non-self, and their capture of donor cell exosomes to amplify the presentation of transplant antigens.


Subject(s)
Antigen Presentation , Dendritic Cells/immunology , Graft Rejection/immunology , Organ Transplantation , T-Lymphocytes/immunology , Transplantation Immunology , Transplantation Tolerance/immunology , Animals , Cell Differentiation , Exosomes/immunology , Humans , Isoantigens/immunology , Lymphocyte Activation , Microbiota/immunology
20.
Proc Natl Acad Sci U S A ; 113(27): 7551-6, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27317748

ABSTRACT

Fibroblasts are common cell types in cancer stroma and lay down collagen required for survival and growth of cancer cells. Although some cancer therapy strategies target tumor fibroblasts, their origin remains controversial. Multiple publications suggest circulating mesenchymal precursors as a source of tumor-associated fibroblasts. However, we show by three independent approaches that tumor fibroblasts derive primarily from local, sessile precursors. First, transplantable tumors developing in a mouse expressing green fluorescent reporter protein (EGFP) under control of the type I collagen (Col-I) promoter (COL-EGFP) had green stroma, whereas we could not find COL-EGFP(+) cells in tumors developing in the parabiotic partner lacking the fluorescent reporter. Lack of incorporation of COL-EGFP(+) cells from the circulation into tumors was confirmed in parabiotic pairs of COL-EGFP mice and transgenic mice developing autochthonous intestinal adenomas. Second, transplantable tumors developing in chimeric mice reconstituted with bone marrow cells from COL-EGFP mice very rarely showed stromal fibroblasts expressing EGFP. Finally, cancer cells injected under full-thickness COL-EGFP skin grafts transplanted in nonreporter mice developed into tumors containing green stromal cells. Using multicolor in vivo confocal microscopy, we found that Col-I-expressing fibroblasts constituted approximately one-third of the stromal mass and formed a continuous sheet wrapping the tumor vessels. In summary, tumors form their fibroblastic stroma predominantly from precursors present in the local tumor microenvironment, whereas the contribution of bone marrow-derived circulating precursors is rare.


Subject(s)
Cancer-Associated Fibroblasts/physiology , Neoplasms, Experimental/pathology , Actins/metabolism , Animals , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Collagen Type I/metabolism , Green Fluorescent Proteins , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL