scholarly journals Arginase I Release from Activated Neutrophils Induces Peripheral Immunosuppression in a Murine Model of Stroke

2015 ◽  
Vol 35 (10) ◽  
pp. 1657-1663 ◽  
Author(s):  
Trisha R Sippel ◽  
Takeru Shimizu ◽  
Frank Strnad ◽  
Richard J Traystman ◽  
Paco S Herson ◽  
...  

Transient suppression of peripheral immunity is a major source of complication for patients suffering from ischemic stroke. The release of Arginase I (ArgI) from activated neutrophils has recently been associated with T-cell dysfunction in a number of pathologies. However, this pathway has not been previously explored in ischemic stroke. Using the murine model of transient middle cerebral artery occlusion, we explored effects of stroke on peripheral T-cell function and evaluated the role of neutrophils and ArgI. Stimulation of splenic T cells from post-stroke animals with anti-CD3/CD28 resulted in decreased proliferation and interferon-γ production when compared with sham-surgery controls. Flow cytometric analysis of intrasplenic leukocytes exposed the presence of a transient population of activated neutrophils that correlated quantitatively with elevated ArgI levels in culture media. In vitro activation of purified resting neutrophils from unmanipulated controls confirmed the capacity for murine neutrophils to release ArgI from preformed granules. We observed decreased expression of the L-arg-sensitive CD3ζ on T cells, consistent with decreased functional activity. Critically, L-arg supplementation restored the functional response of post-stroke T cells to mitogenic stimulation. Together, these data outline a novel mechanism of reversible, neutrophil-mediated peripheral immunosuppression related to ArgI release following ischemic stroke.

Hypertension ◽  
2016 ◽  
Vol 68 (suppl_1) ◽  
Author(s):  
Isha S Dhande ◽  
Mykola Mamenko ◽  
Yaming Zhu ◽  
Oleh Pochynyuk ◽  
Scott Wenderfer ◽  
...  

The genetic mechanism of end organ injury in hypertension may involve gene variation in genes participating in inflammation and its regulation. We identified a novel truncating mutation in the hypertensive end organ injury-prone spontaneously hypertensive rat (SHR-A3/SHRSP) line affecting the C-terminus of STIM1, a protein involved in the store-operated Ca 2+ entry (SOCE) pathway. The genomes of injury-resistant SHR lines, including SHR-B2 used here, encode the ‘wild-type’ STIM1. SOCE is required by T cells to activate the transcription factor NFAT and regulate T cell proliferation and cytokine production. T cell receptor (TCR) stimulation depletes intracellular Ca 2+ stores, activates the ER Ca 2+ -sensor STIM1 and results in SOCE. We tested the effect of STIM1 mutation on lymphocyte SOCE and found it was dramatically reduced in SHR-A3, but not in SHR-B2 (maximal [Ca 2+ ] i : 150.5±34.9 vs 521.5±42.6 nM, p<0.0001). Flow cytometric analysis of circulating T cell subsets revealed comparable levels of CD4 + and CD8 + T cells in both lines, however circulating CD4 + CD25 + FoxP3 + T regs were reduced in SHR-A3 compared to SHR-B2 (4.34±0.59 vs 7.12±0.33%, p=0.01). TCR-induced lymphocyte proliferation was similar in both SHR-A3 and SHR-B2. T cell cytokine production in response to TCR stimulation was markedly impaired CD4 + T cells from SHR-A3 compared with SHR-B2 (IL-2: 168 ±83.4 vs 1385±377.0 pg/mL, p=0.01; IFNγ: 235±69.5 vs 2119±434.7 pg/mL, p=0.002). IL-2 and IFNγ production was completely inhibited by Pyr6, an inhibitor of Stim1-dependent SOCE. However, circulating levels of IL-2 and IFNγ were not different between the two lines. Based on our findings, we conclude that TCR-mediated effector signaling is impaired due to defective SOCE in SHR-A3 rats. Defects in SOCE in SHR-A3 attributable to STIM1 mutation alter T cell function, reduce T reg numbers and may disturb regulatory interactions between T cells and other immune cells involved in end organ injury.


Blood ◽  
2006 ◽  
Vol 109 (4) ◽  
pp. 1568-1573 ◽  
Author(s):  
Paulo C. Rodriguez ◽  
David G. Quiceno ◽  
Augusto C. Ochoa

Abstract l-arginine (l-Arg) plays a central role in several biologic systems including the regulation of T-cell function. l-Arg depletion by myeloid-derived suppressor cells producing arginase I is seen in patients with cancer inducing T-cell anergy. We studied how l-Arg starvation could regulate T-cell–cycle progression. Stimulated T cells cultured in the absence of l-Arg are arrested in the G0-G1phase of the cell cycle. This was associated with an inability of T cells to up-regulate cyclin D3 and cyclin-dependent kinase 4 (cdk4), but not cdk6, resulting in an impaired downstream signaling with a decreased phosphorylation of Rb protein and a low expression and binding of E2F1. Silencing of cyclin D3 reproduced the cell cycle arrest caused by l-Arg starvation. The regulation of cyclin D3 and cdk4 by l-Arg starvation occurs at transcriptional and posttranscriptional levels. Signaling through GCN2 kinase is triggered during amino acid starvation. Experiments demonstrated that T cells from GCN2 knock-out mice did not show a decreased proliferation and were able to up-regulate cyclin D3 when cultured in the absence of l-Arg. These results contribute to the understanding of a central mechanism by which cancer and other diseases characterized by high arginase I production may cause T-cell dysfunction.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3294-3294
Author(s):  
Anja Troeger ◽  
Jenna Wood ◽  
David A. Williams

Abstract Rho GTPases are well known regulators of actin dynamics, gene transcription, kinase pathways and cell cycle progression. RhoH, a hematopoietic specific, GTPase-deficient Rho GTPase was first defined as a hypermutable gene in diffuse large B cell lymphoma (Pasqualucci et al., 2001), although its role in the pathobiology of B cell malignancies still remains unclear. Subsequently, RhoH has been implicated in TCR signaling and T cell development in humans and mice. As RhoH is required for activation and localization of ZAP70 and LCK to the immune synapse, knockout of RhoH results in T cell deficiency (Gu et al., 2006). However, despite a profound T cell defect in Rhoh-/- mice, we previously noted an apparent paradoxical delay in disease onset in the Em-TCL1Tg murine model of B cell chronic lymphocytic leukemia (CLL) after deletion of RhoH. We previously demonstrated that this is partly due to Rhoh-/- CLL cell-intrinsic changes resulting in impaired access to supportive niches and defective microenvironmental interactions (Troeger et al., 2012). However, there is accumulating evidence that progressive immune dysregulation also plays an important role in CLL progression. Specifically, an increase in circulating follicular helper T cells (Tfh), inverted CD4:CD8 ratios, a predominance of a memory T cell phenotype, defective T cell motility and an impaired immunological synapse formation have been reported in CLL patients and Em-TCL1Tg mice (Hofbauer et al., 2011). Interestingly, the immunomodulatory drug lenalidomide has proven effective in modulating CLL-associated changes in T cell function and altered Rho GTPase activity (Ramsay et al., 2013) and we have previously demonstrated that lenalidomide treatment of CLL cells in vivo and in vitro resulted in decreased RhoH expression, suggesting that Rho GTPases are involved in T cell- B cell/CLL interactions. Moreover, lenalidomide treatment has been shown to restore immune synapse formation and T cell function in CLL patients indicating that RhoH may similarly modulate T cell- B cell crosstalk including modified LFA1 signaling. Here we aimed to assess the impact of RhoH on the germinal center reaction and changes in T cell populations over time in knock-out mice and a murine model of CLL. We demonstrate that RhoH is required for normal germinal center (GC) formation and induction of T cell dependent B cell responses in vivo. Thus, while IgM levels were only mildly reduced in Rhoh-/- mice, these animals exhibited significantly reduced IgG1 serum levels 21 days after TNP-KLH treatment (WT vs. Rhoh-/-mice: IgG1 3706951ng/ml +/-871537 vs 122176ng/ml +/-14006; mean+/- SEM; p=0.01), indicating a defect in immunoglobulin class switching. In keeping with these observations, we detected a severe defect in CXCR5+ Tfh cells in the spleens (WT vs Rhoh-/- mice: 10.09%+/-1.23 vs. 1.71%+/-0.45; mean+/- SEM; p=0.01) and peripheral blood (WT vs Rhoh-/- mice: 4.69%+/-0.51 vs 0.36% +/-0.13) of young Rhoh-/- mice, and a profound defect in both naïve CD4+ and CD8+ T cells. However, while over time in mice with CLL disease CD8+ activated effector memory T cells expanded in Em-TCL1Tg;Rhoh-/- to levels comparable to WT mice, deficiency in CD4+ Th cells persisted in Rhoh-/- animals. As it has been shown that CLL cell proliferation depends on Th cell support (Plander et al., 2009), we next aimed to assess the impact of the T cell phenotype on disease progression by performing adoptive transfer experiments. After 6 weeks, recipients of WT CLL cells demonstrated a significantly accelerated disease progression compared to those injected with Rhoh-/- CLL cells (WT vs. Rhoh-/- recipients:238.1+/-65.8 K/µl vs 195.2+/-54.8 K/µl WT CLL cells; and 2.3+/-1.4 K/µl vs 5.9+/-2.5 K/µl Rhoh-/- CLL cells) and accordingly also demonstrated improved survival (survival probability: WT CLL: 0.4+/-0.15 vs. 0.3+/-0.14 and Rhoh-/- CLL: 0.75+/-0.13 vs. 0.82+/-0.12; p<0.05). These data clarify that the lack of CD4+ Th cell support and impaired GC reaction due to RhoH-deficiency have little impact on disease progression in this model of CLL. The findings further confirm that the slower disease progression and improved survival observed in the murine model of Rhoh-/- CLL is mainly mediated by cell autonomous characteristics of the CLL cells and a sufficiently sustained immune surveillance by CD8+ T cells. Thus, inhibition of RhoH may represent an attractive tool for future targeted therapies in CLL. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A559-A559
Author(s):  
Yu Si ◽  
Kirsten Bruderek ◽  
Simon Merz ◽  
Philip Jansen ◽  
Stephan Lang ◽  
...  

BackgroundA high neutrophil-to-lymphocyte ratio in the circulation and high frequencies of tumor-associated neutrophils (TAN) in malignant tissue are associated with poor outcome and tumor progression in patients with cancer. It is hypothesized that immunosuppressive neutrophil activity (aka PMN-MDSC activity) contributes to this effect. In addition, this MDSC activity represents a major resistance mechanism in different types of immunotherapy. The exact cellular identity of human PMN-MDSC is still under debate. Improved immunomonitoring and functional characterization of MDSC is needed in order to exploit these cells as novel biomarkers and targets for combination immunotherapy.MethodsIn this study, we sought to identify the neutrophil subset that contained the highest T cell suppressive activity. To this end, we purified different subsets of circulating neutrophils by FACS and performed multi-parameter immunofluorescence together with digital pathology on 2-D and 3-D tumor tissue samples.ResultsWe found that a population of circulating, mature, arginase I+ neutrophils that co-purified with mononuclear cells in density gradients, most potently suppressed T cell function in multiple in vitro assays. These PMN-MDSC were also superior to monocytic MDSC in T cell suppression. Using a novel technology of tissue whole mount labelling, clearing and imaging we derived 3-D spatial maps of neutrophil – T cell interaction in human tumors. We found that T cells, which were conjugated to arginase I+, myeloperoxidase+ TAN, had significantly reduced expression of proliferation and cytotoxicity markers. In patients, frequent conjugation of T cells to those PMN-MDSC was associated with poor prognosis. In contrast to circulating PMN-MDSC, tissue PMN-MDSC expressed high amounts of LOX-1 (oxidized low density lipoprotein receptor 1) and a high intratumoral frequency of LOX-1+ PMN-MDSC was associated with poor survival.ConclusionsWe identified and characterized PMN-MDSC activity in human cancer patients. Our findings will facilitate and improve MDSC immunomonitoring and MDSC targeting in combination therapies.Ethics ApprovalUse of patient material was approved by the Ethics committee of the Medical Faculty of the University of Duisburg-Essen


2009 ◽  
Vol 296 (3) ◽  
pp. H689-H697 ◽  
Author(s):  
Karen Y. Stokes ◽  
LeShanna Calahan ◽  
Candiss M. Hamric ◽  
Janice M. Russell ◽  
D. Neil Granger

Hypercholesterolemia is associated with phenotypic changes in endothelial cell function that lead to a proinflammatory and prothrombogenic state in different segments of the microvasculature. CD40 ligand (CD40L) and its receptor CD40 are ubiquitously expressed and mediate inflammatory responses and platelet activation. The objective of this study was to determine whether CD40/CD40L, in particular T-cell CD40L, contributes to microvascular dysfunction induced by hypercholesterolemia. Intravital microscopy was used to quantify blood cell adhesion in cremasteric postcapillary venules, endothelium-dependent vasodilation responses in arterioles, and microvascular oxidative stress in wild-type (WT) C57BL/6, CD40-deficient (−/−), CD40L−/−, or severe combined immune deficient (SCID) mice placed on a normal (ND) or high-cholesterol (HC) diet for 2 wk. WT-HC mice exhibited an exaggerated leukocyte and platelet recruitment in venules and impaired vasodilation responses in arterioles compared with ND counterparts. A deficiency of CD40, CD40L, or lymphocytes attenuated these responses to HC. The HC phenotype was rescued in CD40L−/− and SCID mice by a transfer of WT T cells. Bone marrow chimeras revealed roles for both vascular- and blood cell-derived CD40 and CD40L in the HC-induced vascular responses. Hypercholesterolemia induced an oxidative stress in both arterioles and venules of WT mice, which was abrogated by either CD40 or CD40L deficiency. The transfer of WT T cells into CD40L−/− mice restored the oxidative stress. These results implicate CD40/CD40L interactions between circulating cells and the vascular wall in both the arteriolar and venular dysfunction elicited by hypercholesterolemia and identify T-cell-associated CD40L as a key mediator of these responses.


Genes ◽  
2021 ◽  
Vol 12 (6) ◽  
pp. 813
Author(s):  
Norwin Kubick ◽  
Pavel Klimovich ◽  
Patrick Henckell Flournoy ◽  
Irmina Bieńkowska ◽  
Marzena Łazarczyk ◽  
...  

Understanding the evolution of interleukins and interleukin receptors is essential to control the function of CD4+ T cells in various pathologies. Numerous aspects of CD4+ T cells’ presence are controlled by interleukins including differentiation, proliferation, and plasticity. CD4+ T cells have emerged during the divergence of jawed vertebrates. However, little is known about the evolution of interleukins and their origin. We traced the evolution of interleukins and their receptors from Placozoa to primates. We performed phylogenetic analysis, ancestral reconstruction, HH search, and positive selection analysis. Our results indicated that various interleukins' emergence predated CD4+ T cells divergence. IL14 was the most ancient interleukin with homologs in fungi. Invertebrates also expressed various interleukins such as IL41 and IL16. Several interleukin receptors also appeared before CD4+ T cells divergence. Interestingly IL17RA and IL17RD, which are known to play a fundamental role in Th17 CD4+ T cells first appeared in mollusks. Furthermore, our investigations showed that there is not any single gene family that could be the parent group of interleukins. We postulate that several groups have diverged from older existing cytokines such as IL4 from TGFβ, IL10 from IFN, and IL28 from BCAM. Interleukin receptors were less divergent than interleukins. We found that IL1R, IL7R might have diverged from a common invertebrate protein that contained TIR domains, conversely, IL2R, IL4R and IL6R might have emerged from a common invertebrate ancestor that possessed a fibronectin domain. IL8R seems to be a GPCR that belongs to the rhodopsin-like family and it has diverged from the Somatostatin group. Interestingly, several interleukins that are known to perform a critical function for CD4+ T cells such as IL6, IL17, and IL1B have gained new functions and evolved under positive selection. Overall evolution of interleukin receptors was not under significant positive selection. Interestingly, eight interleukin families appeared in lampreys, however, only two of them (IL17B, IL17E) evolved under positive selection. This observation indicates that although lampreys have a unique adaptive immune system that lacks CD4+ T cells, they could be utilizing interleukins in homologous mode to that of the vertebrates' immune system. Overall our study highlights the evolutionary heterogeneity within the interleukins and their receptor superfamilies and thus does not support the theory that interleukins evolved solely in jawed vertebrates to support T cell function. Conversely, some of the members are likely to play conserved functions in the innate immune system.


2021 ◽  
Vol 9 (8) ◽  
pp. e002628
Author(s):  
Jitao Guo ◽  
Andrew Kent ◽  
Eduardo Davila

Adoptively transferred T cell-based cancer therapies have shown incredible promise in treatment of various cancers. So far therapeutic strategies using T cells have focused on manipulation of the antigen-recognition machinery itself, such as through selective expression of tumor-antigen specific T cell receptors or engineered antigen-recognition chimeric antigen receptors (CARs). While several CARs have been approved for treatment of hematopoietic malignancies, this kind of therapy has been less successful in the treatment of solid tumors, in part due to lack of suitable tumor-specific targets, the immunosuppressive tumor microenvironment, and the inability of adoptively transferred cells to maintain their therapeutic potentials. It is critical for therapeutic T cells to overcome immunosuppressive environmental triggers, mediating balanced antitumor immunity without causing unwanted inflammation or autoimmunity. To address these hurdles, chimeric receptors with distinct signaling properties are being engineered to function as allies of tumor antigen-specific receptors, modulating unique aspects of T cell function without directly binding to antigen themselves. In this review, we focus on the design and function of these chimeric non-antigen receptors, which fall into three broad categories: ‘inhibitory-to-stimulatory’ switch receptors that bind natural ligands, enhanced stimulatory receptors that interact with natural ligands, and synthetic receptor-ligand pairs. Our intent is to offer detailed descriptions that will help readers to understand the structure and function of these receptors, as well as inspire development of additional novel synthetic receptors to improve T cell-based cancer therapy.


2000 ◽  
Vol 355 (1400) ◽  
pp. 1093-1101 ◽  
Author(s):  
P. C. Doherty ◽  
J. M. Riberdy ◽  
G. T. Belz

The recent development of techniques for the direct staining of peptide–specific CD8 + T cells has revolutionized the analysis of cell–mediated immunity (CMI) in virus infections. This approach has been used to quantify the acute and long–term consequences of infecting laboratory mice with the readily eliminated influenza A viruses (fluA) and a persistent γherpesvirus (γHV). It is now, for the first time, possible to work with real numbers in the analysis of CD8 + T CMI, and to define various characteristics of the responding lymphocytes both by direct flow cytometric analysis and by sorting for further in vitro manipulation. Relatively little has yet been done from the latter aspect, though we are rapidly accumulating a mass of numerical data. The acute, antigen–driven phases of the fluA and γHV–specific response look rather similar, but CD8 + T–cell numbers are maintained in the long term at a higher ‘set point’ in the persistent infection. Similarly, these ‘memory’ T cells continue to divide at a much greater rate in the γHV–infected mice. New insights have also been generated on the nature of the recall response following secondary challenge in both experimental systems, and the extent of protection conferred by large numbers of virus–specific CD8 + T cells has been determined. However, there are still many parameters that have received little attention, partly because they are difficult to measure. These include the rate of antigen–specific CD8 + T–cell loss, the extent of the lymphocyte ‘diaspora’ to other tissues, and the diversity of functional characteristics, turnover rates, clonal life spans and recirculation profiles. The basic question for immunologists remains how we reconcile the extraordinary plasticity of the immune system with the mechanisms that maintain a stable milieu interieur. This new capacity to quantify CD8 + T–cell responses in readily manipulated mouse models has obvious potential for illuminating homeostatic control, particularly if the experimental approaches to the problem are designed in the context of appropriate predictive models.


Molecules ◽  
2015 ◽  
Vol 20 (10) ◽  
pp. 19014-19026 ◽  
Author(s):  
Masao Goto ◽  
Manabu Wakagi ◽  
Toshihiko Shoji ◽  
Yuko Takano-Ishikawa

2009 ◽  
Vol 206 (2) ◽  
pp. 421-434 ◽  
Author(s):  
Randall H. Friedline ◽  
David S. Brown ◽  
Hai Nguyen ◽  
Hardy Kornfeld ◽  
JinHee Lee ◽  
...  

Cytotoxic T lymphocyte antigen-4 (CTLA-4) plays a critical role in negatively regulating T cell responses and has also been implicated in the development and function of natural FOXP3+ regulatory T cells. CTLA-4–deficient mice develop fatal, early onset lymphoproliferative disease. However, chimeric mice containing both CTLA-4–deficient and –sufficient bone marrow (BM)–derived cells do not develop disease, indicating that CTLA-4 can act in trans to maintain T cell self-tolerance. Using genetically mixed blastocyst and BM chimaeras as well as in vivo T cell transfer systems, we demonstrate that in vivo regulation of Ctla4−/− T cells in trans by CTLA-4–sufficient T cells is a reversible process that requires the persistent presence of FOXP3+ regulatory T cells with a diverse TCR repertoire. Based on gene expression studies, the regulatory T cells do not appear to act directly on T cells, suggesting they may instead modulate the stimulatory activities of antigen-presenting cells. These results demonstrate that CTLA-4 is absolutely required for FOXP3+ regulatory T cell function in vivo.


Sign in / Sign up

Export Citation Format

Share Document