Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Ultrasound Med Biol ; 49(9): 2152-2159, 2023 09.
Article in English | MEDLINE | ID: mdl-37394375

ABSTRACT

OBJECTIVE: Biofilm formation in medical catheters is a major source of hospital-acquired infections which can produce increased morbidity and mortality for patients. Histotripsy is a non-invasive, non-thermal focused ultrasound therapy and recently has been found to be effective at removal of biofilm from medical catheters. Previously established histotripsy methods for biofilm removal, however, would require several hours of use to effectively treat a full-length medical catheter. Here, we investigate the potential to increase the speed and efficiency with which biofilms can be ablated from catheters using histotripsy. METHODS: Pseudomonas aeruginosa (PA14) biofilms were cultured in in vitro Tygon catheter mimics and treated with histotripsy using a 1 MHz histotripsy transducer and a variety of histotripsy pulsing rates and scanning methods. The improved parameters identified in these studies were then used to explore the bactericidal effect of histotripsy on planktonic PA14 suspended in a catheter mimic. RESULTS: Histotripsy can be used to remove biofilm and kill bacteria at substantially increased speeds compared with previously established methods. Near-complete biofilm removal was achieved at treatment speeds up to 1 cm/s, while a 4.241 log reduction in planktonic bacteria was achieved with 2.4 cm/min treatment. CONCLUSION: These results represent a 500-fold increase in biofilm removal speeds and a 6.2-fold increase in bacterial killing speeds compared with previously published methods. These findings indicate that histotripsy shows promise for the treatment of catheter-associated biofilms and planktonic bacteria in a clinically relevant time frame.


Subject(s)
Anti-Bacterial Agents , Cross Infection , Humans , Anti-Bacterial Agents/therapeutic use , Catheters , Biofilms
2.
Am J Cardiol ; 191: 43-50, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36640599

ABSTRACT

Bleeding events result in morbidity and mortality in patients who underwent percutaneous coronary intervention (PCI). There are limited data on the predicting bleeding complications in patients who underwent stent implantation and subsequent dual antiplatelet therapy (PRECISE-DAPT) and Academic Research Consortium for High Bleeding Risk (ARC-HBR) scores' ability to predict in-hospital outcomes in patients who underwent PCI. Consecutive patients who underwent PCI at tertiary centers from January 2016 to March 2018 were identified and the bleeding risk scores were calculated. The primary end point was the National Cardiovascular Data Registry-defined in-hospital bleeding stratified by low versus high predicted bleeding risk. The major and net adverse cardiovascular events were also examined. The discriminatory ability of the risk models was determined using receiver operating characteristic curves. Among 3,659 patients studied, the in-hospital major bleeding was 3.3% (n = 121). The patients characterized as high bleeding risk by either criterion had significantly higher bleeding rates than those meeting the low-risk criteria (ARC-HBR 5.4% vs 3.3%, p <0.001; PRECISE-DAPT 5.8% vs 2.4%, p <0.001), and higher major adverse cardiovascular events and net adverse clinical events. These risk estimates showed moderate and similar predictive ability (ARC-HBR high-risk area under the receiver operating characteristic curve [AUC] 0.62, PRECISE-DAPT ≥25 AUC 0.61, p = 0.49), with no incremental benefit to adding the estimates (AUC 0.60). The subgroup analysis revealed that women had higher bleeding rates than men (5.53% vs 2.39%, p <0.001); however, the predictive ability of the criteria were similar in women and men. The patients identified as having a high bleeding risk by the PRECISE-DAPT and the ARC-HBR criteria before PCI are at high risk for in-hospital bleeding and adverse outcomes independent of gender. The 2 scores have moderate predictive ability for bleeds. Further study is needed to determine strategies to reduce risk in this population.


Subject(s)
Cardiovascular Diseases , Percutaneous Coronary Intervention , Male , Humans , Female , Platelet Aggregation Inhibitors/therapeutic use , Percutaneous Coronary Intervention/adverse effects , Hemorrhage/etiology , Risk Factors , Cardiovascular Diseases/etiology , Treatment Outcome , Risk Assessment
3.
Cancer Immunol Res ; 11(1): 38-55, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36260656

ABSTRACT

V domain immunoglobulin suppressor of T-cell activation (VISTA) is a premier target for cancer treatment due to its broad expression in many cancer types and enhanced expression upon development of adaptive immune checkpoint resistance. In the CT26 colorectal cancer model, monotherapy of small tumors with anti-VISTA resulted in slowed tumor growth. In a combination therapy setting, large CT26 tumors showed complete adaptive resistance to anti-PD-1/CTLA-4, but inclusion of anti-VISTA led to rejection of half the tumors. Mechanisms of enhanced antitumor immunity were investigated using single-cell RNA sequencing (scRNA-seq), multiplex image analysis, and flow cytometry of the tumor immune infiltrate. In both treatment models, anti-VISTA upregulated stimulated antigen presentation pathways and reduced myeloid-mediated suppression. Imaging revealed an anti-VISTA stimulated increase in contacts between T cells and myeloid cells, further supporting the notion of increased antigen presentation. scRNA-seq of tumor-specific CD8+ T cells revealed that anti-VISTA therapy induced T-cell pathways highly distinct from and complementary to those induced by anti-PD-1 therapy. Whereas anti-CTLA-4/PD-1 expanded progenitor exhausted CD8+ T-cell subsets, anti-VISTA promoted costimulatory genes and reduced regulators of T-cell quiescence. Notably, this is the first report of a checkpoint regulator impacting CD8+ T-cell quiescence, and the first indication that quiescence may be a target in the context of T-cell exhaustion and in cancer. This study builds a foundation for all future studies on the role of anti-VISTA in the development of antitumor immunity and provides important mechanistic insights that strongly support use of anti-VISTA to overcome the adaptive resistance seen in contemporary treatments involving PD-1 and/or CTLA-4. See related Spotlight by Wei, p. 3.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , B7 Antigens/immunology , Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Immunoglobulins
4.
IDCases ; 26: e01273, 2021.
Article in English | MEDLINE | ID: mdl-34584844

ABSTRACT

Myocarditis occurs with a variety of infectious agents including viruses, bacteria, protozoa and parasites. We present a rare case of myocarditis secondary to Toxoplasma gondii in a 23-year-old immunocompetent male presenting with acute chest pain. Workup revealed evidence of biventricular myocarditis on cardiac magnetic resonance imaging, elevated Toxoplasma serologies with rising titers over time. The patient was treated with sulfadiazine and pyrimethamine for eighteen days with resolution of symptoms. This case highlights alternative diagnostic and treatment modalities for Toxoplasma myocarditis in immunocompetent hosts.

5.
J Am Heart Assoc ; 10(17): e020890, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34431361

ABSTRACT

Background Black men and women are at higher risk for, and suffer greater morbidity and mortality from, atherosclerotic cardiovascular disease (ASCVD) compared with adults of European Ancestry (EA). Black patients with familial hypercholesterolemia are at particularly high risk for ASCVD complications because of lifelong exposure to elevated levels of low-density-lipoprotein cholesterol. Methods and Results This retrospective study analyzed ASCVD prevalence and risk factors in 808 adults with heterozygous familial hypercholesterolemia from 5 US-based lipid clinics, and compared findings in Black versus EA patients. Multivariate logistic regression models were used to determine the strongest predictors of ASCVD as a function of race. No significant difference was noted in the prevalence of ASCVD in Black versus EA patients with familial hypercholesterolemia (39% versus 32%, respectively; P=0.15). However, Black versus EA patients had significantly greater prevalence of modifiable risk factors, including body mass index (mean, 32±7 kg/m2 versus 29±6 kg/m2; P<0.001), hypertension (82% versus 50%; P<0.001), diabetes (39% versus 15%; P<0.001), and current smoking (16% versus 8%; P=0.006). Black versus EA patients also had significantly lower usage of statins (61% versus 73%; P=0.004) and other lipid-lowering agents. In a fully adjusted multivariate model, race was not independently associated with ASCVD (odds ratio, 0.92; 95% CI, 0.60-1.49; P=0.72). Conclusions The strongest predictors of ASCVD in Black patients with familial hypercholesterolemia were hypertension and cigarette smoking. These data support wider usage of statins and other lipid-lowering therapies and greater attention to modifiable risk, specifically blood pressure management and smoking cessation.


Subject(s)
Atherosclerosis , Black People , Cardiovascular Diseases , Health Status Disparities , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hyperlipoproteinemia Type II , Adult , Atherosclerosis/ethnology , Cardiovascular Diseases/ethnology , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/ethnology , Hypertension/ethnology , Male , Racial Groups , Retrospective Studies , Risk Factors , United States
6.
Acta Neuropathol Commun ; 9(1): 91, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34006329

ABSTRACT

V-type immunoglobulin domain-containing suppressor of T-cell activation (VISTA) is a negative checkpoint regulator (NCR) that is involved in T-cell quiescence, inhibition of T-cell activation, and in myeloid cells regulates cytokine production, chemotaxis, phagocytosis, and tolerance induction. In the central nervous system (CNS), VISTA is expressed by microglia, the resident macrophage of the parenchyma, and expression is decreased during neuroinflammation; however, the function of VISTA in microglia is unknown. Here, we extensively analyzed VISTA expression in different MS lesion stages and characterized the function of VISTA in the CNS by deleting VISTA in microglia. VISTA is differentially expressed in distinct MS lesion stages. In mice, VISTA deletion in Cx3Cr1-expressing cells induced a more amoeboid microglia morphology, indicating an immune-activated phenotype. Expression of genes associated with cell cycle and immune-activation was increased in VISTA KO microglia. In response to LPS and during experimental autoimmune encephalomyelitis (EAE), VISTA KO and WT microglia shared similar transcriptional profiles and VISTA deletion did not affect EAE disease progression or microglia responses. VISTA KO in microglia in vitro decreased the uptake of myelin. This study demonstrates that VISTA is involved in microglia function, which likely affects healthy CNS homeostasis and neuroinflammation.


Subject(s)
Homeostasis/physiology , Membrane Proteins/deficiency , Microglia/metabolism , Multiple Sclerosis/metabolism , Myelin Sheath/metabolism , Phagocytosis/physiology , Animals , Animals, Newborn , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Jurkat Cells , Male , Membrane Proteins/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Myelin Sheath/genetics , Myelin Sheath/pathology , Transcription, Genetic/physiology
7.
Science ; 367(6475)2020 01 17.
Article in English | MEDLINE | ID: mdl-31949051

ABSTRACT

Negative checkpoint regulators (NCRs) temper the T cell immune response to self-antigens and limit the development of autoimmunity. Unlike all other NCRs that are expressed on activated T lymphocytes, V-type immunoglobulin domain-containing suppressor of T cell activation (VISTA) is expressed on naïve T cells. We report an unexpected heterogeneity within the naïve T cell compartment in mice, where loss of VISTA disrupted the major quiescent naïve T cell subset and enhanced self-reactivity. Agonistic VISTA engagement increased T cell tolerance by promoting antigen-induced peripheral T cell deletion. Although a critical player in naïve T cell homeostasis, the ability of VISTA to restrain naïve T cell responses was lost under inflammatory conditions. VISTA is therefore a distinctive NCR of naïve T cells that is critical for steady-state maintenance of quiescence and peripheral tolerance.


Subject(s)
B7 Antigens/physiology , Membrane Proteins/physiology , Peripheral Tolerance/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , B7 Antigens/genetics , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Tolerance/genetics , Receptors, Antigen, T-Cell/physiology
8.
Front Immunol ; 10: 2641, 2019.
Article in English | MEDLINE | ID: mdl-31803182

ABSTRACT

The role of negative checkpoint regulators (NCRs) in human health and disease cannot be overstated. V-domain Ig-containing Suppressor of T-cell Activation (VISTA) is an Ig superfamily protein predominantly expressed within the hematopoietic compartment and has been studied for its role in the negative regulation of T cell responses. The findings presented in this study show that, unlike all other NCRs, VISTA deficiency dramatically impacts on macrophage cytokine and chemokine production, as well as the chemotactic response of VISTA-deficient macrophages. A select group of inflammatory chemokines, including CCL2, CCL3, CCL4, and CCL5, was strikingly elevated in culture supernatants from VISTA KO macrophages. VISTA deficiency also altered chemokine receptor recycling and profoundly disrupted myeloid chemotaxis. The impact of VISTA deficiency on chemotaxis in vivo was apparent with the reduced ability of both KO macrophages and MDSCs to migrate to the tumor microenvironment. This is the first demonstration of an NCR impacting on myeloid mediator production and chemotaxis, and will guide the use of anti-VISTA therapeutics to manipulate the chemotaxis of inflammatory macrophages or immunosuppressive MDSCs in inflammatory diseases and cancer.


Subject(s)
Chemokines/physiology , Chemotaxis/physiology , Macrophages/physiology , Membrane Proteins/physiology , Myeloid-Derived Suppressor Cells/physiology , Animals , Cell Line, Tumor , Female , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Tumor Microenvironment
9.
Transfusion ; 59(10): 3077-3083, 2019 10.
Article in English | MEDLINE | ID: mdl-31490558

ABSTRACT

BACKGROUND: Astronauts on exploration missions may be at risk for traumatic injury and medical conditions that lead to life threatening hemorrhage. Resuscitation protocols are limited by the austere conditions of spaceflight. Solutions may be found in low-resource terrestrial settings. The existing literature on alternative blood product administration and walking blood banks was evaluated for applicability to spaceflight. STUDY DESIGN AND METHODS: A literature review was done using PubMed and Google Scholar. References were crosschecked for additional publications not identified using the initial search terms. Twenty-seven articles were identified, including three controlled trials, six retrospective cohort analyses, 15 reviews, one case report, and two experimental studies. RESULTS: Solutions to blood transfusion in austere settings include lyophilized blood products, hemoglobin-based oxygen carriers (HBOCs), and fresh whole blood. Many of these products are investigational. Protocols for walking blood banks include methods for screening and activating donors, transfusion, and monitoring for adverse reactions. Microgravity and mission limitations create additional challenges for transfusion, including baseline physiologic changes, difficulty reconstituting lyophilized products, risk of air emboli during transfusion, equipment constraints, and limited evacuation and surgical options. CONCLUSION: Medical planning for space exploration should consider the possibility of acute blood loss. A model for "floating" blood banks based on terrestrial walking blood bank protocols from austere environments is presented, with suggestions for future development. Constraints on volume, mass, storage, and crew, present challenges to blood transfusion in space and must be weighed against the benefits of expanding medical capabilities.


Subject(s)
Blood Banks , Blood Transfusion , Hemorrhage/therapy , Resuscitation , Space Flight , Animals , Humans
10.
J Immunol ; 202(9): 2795-2805, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30885956

ABSTRACT

Recent studies have underscored the critical role of retinoic acid (RA) in the development of lineage-committed CD4 and CD8 T cells in vivo. We have shown that under acute graft-versus-host disease (GVHD) inflammatory conditions, RA is upregulated in the intestine and is proinflammatory, as GVHD lethality was attenuated when donor allogeneic T cells selectively expressed a dominant negative RA receptor α that blunted RA signaling. RA can function in an autocrine and paracrine fashion, and as such, the host cell lineage responsible for the production of RA metabolism and the specific RA-metabolizing enzymes that potentiate GVHD severity are unknown. In this study, we demonstrate that enhancing RA degradation in the host and to a lesser extent donor hematopoietic cells by overexpressing the RA-catabolizing enzyme CYP26A1 reduced GVHD. RA production is facilitated by retinaldehyde isoform-2 (RALDH2) preferentially expressed in dendritic cells (DCs). Conditionally deleted RA-synthesizing enzyme RALDH2 in host or to a lesser extent donor DCs reduced GVHD lethality. Improved survival in recipients with RALDH2-deleted DCs was associated with increased T cell death, impaired T effector function, increased regulatory T cell frequency, and augmented coinhibitory molecule expression on donor CD4+ T cells. In contrast, retinaldehydrogenase isoform-1 (RALDH1) is dominantly expressed in intestinal epithelial cells. Unexpectedly, conditional host intestinal epithelial cells RALDH1 deletion failed to reduce GVHD. These data demonstrate the critical role of both donor and especially host RALDH2+ DCs in driving murine GVHD and suggest RALDH2 inhibition or CYP26A1 induction as novel therapeutic strategies to prevent GVHD.


Subject(s)
Aldehyde Oxidoreductases/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Gene Expression Regulation, Enzymologic/immunology , Graft vs Host Disease/immunology , Aldehyde Oxidoreductases/genetics , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/immunology , Tretinoin/immunology
11.
Immunol Rev ; 276(1): 66-79, 2017 03.
Article in English | MEDLINE | ID: mdl-28258694

ABSTRACT

Utilization of negative checkpoint regulators (NCRs) for cancer immunotherapy has garnered significant interest with the completion of clinical trials demonstrating efficacy. While the results of monotherapy treatments are compelling, there is increasing emphasis on combination treatments in an effort to increase response rates to treatment. One of the most recently discovered NCRs is VISTA (V-domain Ig-containing Suppressor of T cell Activation). In this review, we describe the functions of this molecule in the context of cancer immunotherapy. We also discuss factors that may influence the use of anti-VISTA antibody in combination therapy and how genomic analysis may assist in providing indications for treatment.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B7 Antigens/metabolism , Immunotherapy/methods , Neoplasms/therapy , Animals , B7 Antigens/genetics , B7 Antigens/immunology , Combined Modality Therapy , Genome , Humans , Lymphocyte Activation , Neoplasms/immunology
12.
Am J Transl Res ; 8(2): 1016-26, 2016.
Article in English | MEDLINE | ID: mdl-27158387

ABSTRACT

Retinoid x receptors (RXRs) are master regulators that control cell growth, differentiation, and survival and form heterodimers with many other family members. Here we show that treatment with the RXR agonist IRX4204 enhances the differentiation of CD4(+) T cells into inducible regulatory T cells (iTreg) and suppresses the development of T helper (Th) 17 cells in vitro. Furthermore in a murine model of multiple sclerosis (experimental autoimmune encephalomyelitis (EAE)), treatment with IRX4204 profoundly attenuates both active and Th17-mediated passive disease. In the periphery, treatment with IRX4204 is associated with decreased numbers of CD4(+) T cells that produce pro-inflammatory cytokines. In addition, CD4(+) T cells express decreased levels of Ki-67 and increased expression of CTLA-4. Our findings demonstrate IRX4204 treatment during EAE results in immune modulation and profound attenuation of disease severity.

13.
Methods Mol Biol ; 1220: 461-86, 2015.
Article in English | MEDLINE | ID: mdl-25388268

ABSTRACT

Mast cells (MCs) play an important role in both inflammatory and immunosuppressive responses [1]. The importance of MCs in maintaining peripheral tolerance was discovered in a FoxP3(+) regulatory T-cell (Treg)-mediated skin transplant model [2]. MCs can directly mediate tolerance by releasing anti-inflammatory mediators (reviewed in ref. 3) or by interacting with other immune cells in the graft. Here we will present protocols used to study the role of MCs in peripheral tolerance with the emphasis on how MCs can regulate T-cell functionality. First we will introduce the skin transplant model followed by reconstitution of mast cell-deficient mice (B6.Cg-Kit (W-sh) ). This includes the preparation of MCs from the bone marrow. Finally the methods used to study the influence of MCs on T-cell responses and Treg functionality will be presented by modulating the balance between tolerance and inflammation.


Subject(s)
Mast Cells/immunology , Peripheral Tolerance , Skin Transplantation , Adoptive Transfer , Animals , Antibodies/immunology , Bone Marrow Cells/cytology , Cell Degranulation , Cytokines/metabolism , Graft Rejection , Mast Cells/cytology , Mice , Microscopy, Fluorescence , Models, Animal , Skin Transplantation/adverse effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Tissue and Organ Harvesting
14.
J Immunol ; 194(2): 505-13, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25505290

ABSTRACT

Chemokine-dependent localization of specific B cell subsets within the immune microarchitecture is essential to ensure successful cognate interactions. Although cognate interactions between T cells and memory B cells (B(mem)) are essential for the secondary humoral immune responses, the chemokine response patterns of B(mem) cells are largely unknown. In contrast to naive B cells, this study shows that Ag-specific B(mem) cells have heightened expression of CCR6 and a selective chemotactic response to the CCR6 ligand, CCL20. Although CCR6 appears be nonessential for the initial clonal expansion and maintenance of B(mem), CCR6 is essential for the ability of B(mem) to respond to a recall response to their cognate Ag. This dependency was deemed intrinsic by studies in CCR6-deficient mice and in bone marrow chimeric mice where CCR6 deficiency was limited to the B cell lineage. Finally, the mis-positioning of CCR6-deficient B(mem) was revealed by immunohistological analysis with an altered distribution of CCR6-deficient B(mem) from the marginal and perifollicular to the follicular/germinal center area.


Subject(s)
Antigens/immunology , B-Lymphocytes/immunology , Chemotaxis/immunology , Immunologic Memory/physiology , Receptors, CCR6/immunology , Allografts , Animals , B-Lymphocytes/cytology , Bone Marrow Transplantation , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Chemotaxis/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Germinal Center/cytology , Germinal Center/immunology , Mice , Mice, Knockout , Receptors, CCR6/genetics , Transplantation Chimera/immunology
15.
Proc Natl Acad Sci U S A ; 111(41): 14846-51, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25267631

ABSTRACT

V domain-containing Ig suppressor of T-cell activation (VISTA) is a negative checkpoint regulator that suppresses T cell-mediated immune responses. Previous studies using a VISTA-neutralizing monoclonal antibody show that VISTA blockade enhances T-cell activation. The current study describes a comprehensive characterization of mice in which the gene for VISTA has been deleted. Despite the apparent normal hematopoietic development in young mice, VISTA genetic deficiency leads to a gradual accumulation of spontaneously activated T cells, accompanied by the production of a spectrum of inflammatory cytokines and chemokines. Enhanced T-cell responsiveness was also observed upon immunization with neoantigen. Despite the presence of multiorgan chronic inflammation, aged VISTA-deficient mice did not develop systemic or organ-specific autoimmune disease. Interbreeding of the VISTA-deficient mice with 2D2 T-cell receptor transgenic mice, which are predisposed to the development of experimental autoimmune encephalomyelitis, drastically enhanced disease incidence and intensity. Disease development is correlated with the increase in the activation of encephalitogenic T cells in the periphery and enhanced infiltration into the CNS. Taken together, our data suggest that VISTA is a negative checkpoint regulator whose loss of function lowers the threshold for T-cell activation, allowing for an enhanced proinflammatory phenotype and an increase in the frequency and intensity of autoimmunity under susceptible conditions.


Subject(s)
Autoimmunity/genetics , Autoimmunity/immunology , B7 Antigens/genetics , Genetic Predisposition to Disease , Inflammation/pathology , Aging/pathology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , B7 Antigens/deficiency , B7 Antigens/metabolism , Chemokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Hematopoiesis , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Th1 Cells/immunology , Th17 Cells/immunology
16.
Arthritis Res Ther ; 16(5): 469, 2014.
Article in English | MEDLINE | ID: mdl-25606596

ABSTRACT

Immune checkpoint regulators are critical modulators of the immune system, allowing the initiation of a productive immune response and preventing the onset of autoimmunity. Co-inhibitory and co-stimulatory immune checkpoint receptors are required for full T-cell activation and effector functions such as the production of cytokines. In autoimmune rheumatic diseases, impaired tolerance leads to the development of diseases such as rheumatoid arthritis, systemic lupus erythematosus, and Sjogren's syndrome. Targeting the pathways of the inhibitory immune checkpoint molecules CD152 (cytotoxic T lymphocyte antigen-4) and CD279 (programmed death-1) in cancer shows robust anti-tumor responses and tumor regression. This observation suggests that, in autoimmune diseases, the converse strategy of engaging these molecules may alleviate inflammation owing to the success of abatacept (CD152-Ig) in rheumatoid arthritis patients. We review the preclinical and clinical developments in targeting immune checkpoint regulators in rheumatic disease.


Subject(s)
Costimulatory and Inhibitory T-Cell Receptors/immunology , Inflammation/immunology , Rheumatic Diseases/immunology , Signal Transduction/immunology , Antirheumatic Agents/immunology , Antirheumatic Agents/therapeutic use , Humans , Inflammation/drug therapy , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Models, Immunological , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Rheumatic Diseases/drug therapy , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
Trends Immunol ; 34(11): 556-63, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23954143

ABSTRACT

Fine-tuning the immune response and maintaining tolerance to self-antigens involves a complex network of co-stimulatory and co-inhibitory molecules. The recent FDA approval of ipilimumab, a monoclonal antibody blocking cytotoxic T lymphocyte antigen (CTLA)-4, demonstrates the impact of checkpoint regulators in disease. This is reinforced by ongoing clinical trials targeting not only CTLA-4, but also the programmed death (PD)-1 and B7-H4 pathways in various disease states. Recently, two new B7 family inhibitory ligands, V-domain Ig suppressor of T cell activation (VISTA) and B7-H6 were identified. Here, we review recent understanding of B7 family members and their concerted regulation of the immune response to either self or foreign pathogens. We also discuss clinical developments in targeting these pathways in different disease settings, and introduce VISTA as a putative therapeutic target.


Subject(s)
B7 Antigens/immunology , CTLA-4 Antigen/immunology , Immune Tolerance/immunology , Immunity/immunology , Programmed Cell Death 1 Receptor/immunology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/immunology , Animals , Humans
18.
J Exp Med ; 209(11): 2127-35, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23008335

ABSTRACT

Nutrient deprivation based on the loss of essential amino acids by catabolic enzymes in the microenvironment is a critical means to control inflammatory responses and immune tolerance. Here we report the novel finding that Tph-1 (tryptophan hydroxylase-1), a synthase which catalyses the conversion of tryptophan to serotonin and exhausts tryptophan, is a potent regulator of immunity. In models of skin allograft tolerance, tumor growth, and experimental autoimmune encephalomyelitis, Tph-1 deficiency breaks allograft tolerance, induces tumor remission, and intensifies neuroinflammation, respectively. All of these effects of Tph-1 deficiency are independent of its downstream product serotonin. Because mast cells (MCs) appear to be the major source of Tph-1 and restoration of Tph-1 in the MC compartment in vivo compensates for the defect, these experiments introduce a fundamentally new mechanism of MC-mediated immune suppression that broadly impacts multiple arms of immunity.


Subject(s)
Immune Tolerance/immunology , Inflammation/immunology , Mast Cells/immunology , Tryptophan Hydroxylase/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Flow Cytometry , Gene Expression , Immune Tolerance/genetics , Inflammation/genetics , Male , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Reverse Transcriptase Polymerase Chain Reaction , Skin Transplantation/immunology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism , Transplantation, Homologous , Tryptophan/blood , Tryptophan/metabolism , Tryptophan Hydroxylase/deficiency , Tryptophan Hydroxylase/genetics
19.
Immunity ; 35(4): 550-61, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22035846

ABSTRACT

Peripheral tolerance orchestrated by regulatory T cells, dendritic cells (DCs), and mast cells (MCs) has been studied in several models including skin allograft tolerance. We now define a role for MCs in controlling DC behavior ("conditioning") to facilitate tolerance. Under tolerant conditions, we show that MCs mediated a marked increase in tumor necrosis factor (TNFα)-dependent accumulation of graft-derived DCs in the dLN compared to nontolerant conditions. This increase of DCs in the dLN is due to the local production of granulocyte macrophage colony-stimulating factor (GM-CSF) by MCs that induces a survival advantage of graft-derived DCs. DCs that migrated to the dLN from the tolerant allograft were tolerogenic; i.e., they dominantly suppress T cell responses and control regional immunity. This study underscores the importance of MCs in conditioning DCs to mediate peripheral tolerance and shows a functional impact of peripherally produced TNFα and GM-CSF on the migration and function of tolerogenic DCs.


Subject(s)
Dendritic Cells/immunology , Mast Cells/immunology , Skin/immunology , Transplantation Tolerance , Animals , Cell Movement , Cells, Cultured , Dendritic Cells/cytology , Humans , Mice , Mice, Inbred C57BL , Phenotype , Skin Transplantation , Tumor Necrosis Factor-alpha/immunology
20.
J Exp Med ; 208(9): 1767-75, 2011 Aug 29.
Article in English | MEDLINE | ID: mdl-21859847

ABSTRACT

It is known that vitamin A and its metabolite, retinoic acid (RA), are essential for host defense. However, the mechanisms for how RA controls inflammation are incompletely understood. The findings presented in this study show that RA signaling occurs concurrent with the development of inflammation. In models of vaccination and allogeneic graft rejection, whole body imaging reveals that RA signaling is temporally and spatially restricted to the site of inflammation. Conditional ablation of RA signaling in T cells significantly interferes with CD4(+) T cell effector function, migration, and polarity. These findings provide a new perspective of the role of RA as a mediator directly controlling CD4(+) T cell differentiation and immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/physiology , Cell Movement/physiology , Immunity, Cellular/physiology , Models, Immunological , Signal Transduction/physiology , Tretinoin/immunology , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/metabolism , Immunity, Cellular/drug effects , Immunization , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Knockout , Signal Transduction/drug effects , Skin Transplantation/immunology , Transplantation, Homologous , Tretinoin/metabolism , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...