Laminin-8 (alpha4beta1gamma1) is synthesized by lymphoid cells, promotes lymphocyte migration and costimulates T cell proliferation

2001 ◽  
Vol 114 (2) ◽  
pp. 423-433 ◽  
Author(s):  
T. Geberhiwot ◽  
D. Assefa ◽  
J. Kortesmaa ◽  
S. Ingerpuu ◽  
C. Pedraza ◽  
...  

Laminins are a growing family of large heterotrimeric proteins with cell adhesive and signalling functions. They are major components of basement membranes and are found in many organs, including the vasculature and other compartments of bone marrow, thymus, lymph nodes and spleen. However, expression, recognition and use of laminin isoforms by lymphoid cells are poorly understood. In the present study, lymphoid T cells (Jurkat) were found to synthesize laminin alpha4, beta1 and gamma1 mRNAs and polypeptides and to assemble the chains into laminin-8. Lymphoblastoid B (NAD-20) cells, lymphoid NK (NKL) cells and blood lymphocytes also contained laminin-8 and, after cell permeabilization, practically all blood lymphocytes reacted with mAbs to laminin beta1 and gamma1 chains. Following stimulation, blood lymphocytes secreted laminin-8, and this laminin isoform, but not laminin-10/11(alpha5beta1gamma1/alpha5beta2gamma1), promoted chemokine-induced migration of the cells. In an activation-dependent manner, purified blood CD4 T cells adhered to immobilized laminin-8 and laminin-10/11 by using alpha6beta1 integrin, but minimally to laminin-1 (alpha1beta1gamma1). Accordingly, laminin-8 and laminin-10/11, but not laminin-1, strongly costimulated proliferation of the T cells via the same integrin. Thus, lymphoid cells are able to synthesize and secrete complete laminin molecules. In addition, synthesis of laminin-8 and recognition of laminin-8 and -10/11 by lymphocytes indicate relevance of these laminin isoforms in lymphocyte physiology.

2021 ◽  
Vol 11 ◽  
Author(s):  
Linda Voss ◽  
Karina Guttek ◽  
Annika Reddig ◽  
Annegret Reinhold ◽  
Martin Voss ◽  
...  

Repositioning of approved drugs for identifying new therapeutic purposes is an alternative, time and cost saving strategy to classical drug development. Here, we screened a library of 786 FDA-approved drugs to find compounds, which can potentially be repurposed for treatment of T cell-mediated autoimmune diseases. Investigating the effect of these diverse substances on mitogen-stimulated proliferation of both, freshly stimulated and pre-activated (48 h) peripheral blood mononuclear cells (PBMCs), we discovered Adefovir Dipivoxil (ADV) as very potent compound, which inhibits T cell proliferation in a nanomolar range. We further analyzed the influence of ADV on proliferation, activation, cytokine production, viability and apoptosis of freshly stimulated as well as pre-activated human T cells stimulated with anti-CD3/CD28 antibodies. We observed that ADV was capable of suppressing the proliferation in both T cell stimulation systems in a dose-dependent manner (50% inhibition [IC50]: 63.12 and 364.8 nM for freshly stimulated T cells and pre-activated T cells, respectively). Moreover, the drug impaired T cell activation and inhibited Th1 (IFN-γ), Th2 (IL-5), and Th17 (IL-17) cytokine production dose-dependently. Furthermore, ADV treatment induced DNA double-strand breaks (γH2AX foci expression), which led to an increase of p53-phospho-Ser15 expression. In response to DNA damage p21 and PUMA are transactivated by p53. Subsequently, this caused cell cycle arrest at G0/G1 phase and activation of the intrinsic apoptosis pathway. Our results indicate that ADV could be a new potential candidate for treatment of T cell-mediated autoimmune diseases. Prospective studies should be performed to verify this possible therapeutic application of ADV for such disorders.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1804-1804
Author(s):  
David J. Chung ◽  
Marco Rossi ◽  
Jennifer Pressley ◽  
David H. Munn ◽  
James W. Young

Abstract Effective immunotherapy must overcome tolerance toward tumor antigens and avoid subsequent inhibition of stimulated antitumor immunity. The specific contribution of immune regulatory mechanisms intrinsic to dendritic cells (DCs), especially with regard to regulatory T cells (T regs), is of emerging importance. We have found that all conventional, immunogenic human DCs express the immunomodulatory enzyme indoleamine 2,3-dioxygenase (IDO) and that IDO protein expression and activity are markedly increased in mature compared with immature DCs. Priming of resting T cells with mature IDO+ DCs in an autologous mixed leukocyte reaction (MLR) increases the proportion of CD4+CD25+ T cells capable of suppressing allogeneic T cell proliferation in secondary MLRs as much as 10-fold above baseline (Figure 1A). Conversely, 1-methyl-tryptophan, a competitive inhibitor of IDO, dampens the inhibitory activity (Figure 1A). Further characterization of the suppressor T cells was performed after cytofluorographic sorting into CD4+CD25hi, CD4+CD25int, and CD4+CD25low/− subpopulations. Post-sort analysis revealed that the majority (>60%) of the CD4+CD25hi cells coexpressed Foxp3, which was absent in the CD4+CD25low/− cells. Separate studies showed that these Foxp3+ cells express little or no CD127 (IL-7R-alpha). Candidate CD4+CD25hi T regs inhibited DC-stimulated allogeneic T cell proliferation in a dose dependent manner, with >90% inhibition at a suppressor to responder T cell ratio of 1:1 and ∼50% inhibition at a ratio as low as 1:25 (Figure 1B). CD4+CD25low/− cells were not inhibitory, and CD4+CD25int cells exerted intermediate suppression depending on dose (Figure 1B). CD4+CD25hi T regs exert similar inhibition of autologous T cell responses to stimulation de novo by DCs. Both the priming and effector phases of T reg suppression were contact dependent. Moreover, depletion of the trace population of CD4+CD25hi T cells at the outset of autologous priming largely abolished the relative expansion of this population. These results clearly demonstrate that mature conventional human DCs support relative but significant expansion of autologous, constitutive CD4+CD25hi T cells, which coexpress Foxp3, express little or no CD127, and exert significant suppression of both allogeneic and autologous T cells stimulated de novo by DCs. Although contrary to the anticipated enrichment of IDO in immature DCs because of their expected tolerogenicity, these findings underscore the importance of regulatory mechanisms exerted by immunogenic cells like mature conventional DCs. While this may provide a physiologic means of turning off an otherwise unchecked immune response, this IDO-mediated pathway in DCs provides a rational target for optimizing host immune responses against tumor antigens. This should result in more sustained benefit from active immunotherapy with DC-based vaccines. FIGURE 1: A) T cell mediated suppression of secondary allogeneic MLR. Autologous T eels primed in the presence of the IDO inhibitor. 1-metnyl-D-tryptophan (1-MT), do not develop as much suppressor activity as T cells primed in the absence of 1-MT, resulting in loss inhibition of the secondary MLR Data presented are representative of 6 experiments. B) CD4−CD25***** T cells art potent inhibitors of T cell proliferation. FACS-sorted CD4−CD25******, CD4−CD25*****, and CD4−CD25***** T cells (‘suppressor T cells’) were added to allogeneic MLRs composed of autologous DCs + allogeneic T cells. DC to T cell ratio was 1:30. Suppressor T cell to responder T cell ratios were 1:25, 1:5, and 1:1. After 4–5 days in culture, responder T cell proliferation was assessed by measuring 3HTdR incorporation and comparing with controls containing no suppressor cells (black bar). CD4−CD25***** cells inhibited T cell proliferation in a dose-dependent manner. CD4−CD25***** cells inhibited proliferation to a lesser degree, and CD4-CD25***** cells showed no inhibition. While the CD4−CD25***** cells and to a lesser extent the CD4−CD25***** cells suppressed the proliferative response of alloreactive T cells in the MLRs, they were themselves anergic to the allogeneic DCs in proportion to CD25 and FOXP3 expression (data not shown). Values are pooled from 3 independent experiments (N=4 for each condition), and error bars indicate standard deviation of the mean. FIGURE 1:. A) T cell mediated suppression of secondary allogeneic MLR. Autologous T eels primed in the presence of the IDO inhibitor. 1-metnyl-D-tryptophan (1-MT), do not develop as much suppressor activity as T cells primed in the absence of 1-MT, resulting in loss inhibition of the secondary MLR Data presented are representative of 6 experiments. B) CD4−CD25***** T cells art potent inhibitors of T cell proliferation. FACS-sorted CD4−CD25******, CD4−CD25*****, and CD4−CD25***** T cells (‘suppressor T cells’) were added to allogeneic MLRs composed of autologous DCs + allogeneic T cells. DC to T cell ratio was 1:30. Suppressor T cell to responder T cell ratios were 1:25, 1:5, and 1:1. After 4–5 days in culture, responder T cell proliferation was assessed by measuring 3HTdR incorporation and comparing with controls containing no suppressor cells (black bar). CD4−CD25***** cells inhibited T cell proliferation in a dose-dependent manner. CD4−CD25***** cells inhibited proliferation to a lesser degree, and CD4-CD25***** cells showed no inhibition. While the CD4−CD25***** cells and to a lesser extent the CD4−CD25***** cells suppressed the proliferative response of alloreactive T cells in the MLRs, they were themselves anergic to the allogeneic DCs in proportion to CD25 and FOXP3 expression (data not shown). Values are pooled from 3 independent experiments (N=4 for each condition), and error bars indicate standard deviation of the mean.


1985 ◽  
Vol 161 (4) ◽  
pp. 641-656 ◽  
Author(s):  
T Hara ◽  
S M Fu

Three monoclonal antibodies (mAb), of IgG1, IgG2a, and IgM isotypes, raised against the T3 complex, were used to probe the activation of human T cells. The IgM antibody 235 was not mitogenic for peripheral blood mononuclear cells (PMC). It efficiently blocked the proliferation of PMC induced by T cell mitogens, alloantigens, and soluble antigens. The other two antibodies were mitogenic, and behaved similarly to Leu 4 and OKT3, respectively. In T cell preparations with less than 0.1% monocytes (as assayed by nonspecific esterase staining), all three mAb were not mitogenic. They failed to induce either interleukin 2 (IL-2) receptor expression or IL-2 secretion. Addition of IL-1 failed to collaborate with anti-T3 mAb to induce these T cells to proliferate, but IL-2 enhanced T cell proliferation slightly. Monocyte-depleted T cells, however, proliferated in response to all three anti-T3 mAb, when TPA was added, in a dose-dependent manner. TPA induced a low level of IL-2 receptor expression in monocyte-depleted T cells, without inducing IL-2 secretion. Anti-T3 plus TPA induced a marked enhancement in both quantity and intensity of IL-2 receptor expression. IL-2 secretion was also detected. These results indicate that anti-T3 IgM can deliver an inductive signal despite its blockage of T cell proliferation, and that two signals are necessary and perhaps sufficient to induce human T cell activation and proliferation.


2018 ◽  
Vol 115 (7) ◽  
pp. 1588-1592 ◽  
Author(s):  
Abdulrahman M. Saadalla ◽  
Abu Osman ◽  
Michael F. Gurish ◽  
Kristen L. Dennis ◽  
Nichole R. Blatner ◽  
...  

Mast cells (MCs) are tissue resident sentinels that mature and orchestrate inflammation in response to infection and allergy. While they are also frequently observed in tumors, the contribution of MCs to carcinogenesis remains unclear. Here, we show that sequential oncogenic events in gut epithelia expand different types of MCs in a temporal-, spatial-, and cytokine-dependent manner. The first wave of MCs expands focally in benign adenomatous polyps, which have elevated levels of IL-10, IL-13, and IL-33, and are rich in type-2 innate lymphoid cells (ILC2s). These vanguard MCs adhere to the transformed epithelial cells and express murine mast cell protease 2 (mMCP2; a typical mucosal MC protease) and, to a lesser extent, the connective tissue mast cell (CTMC) protease mMCP6. Persistence of MCs is strictly dependent on T cell-derived IL-10, and their loss in the absence of IL-10–expressing T cells markedly delays small bowel (SB) polyposis. MCs expand profusely in polyposis-prone mice when T cells overexpress IL-10. The frequency of polyp-associated MCs is unaltered in response to broad-spectrum antibiotics, arguing against a microbial component driving their recruitment. Intriguingly, when polyps become invasive, a second wave of mMCP5+/mMCP6+ CTMCs expands in the tumor stroma and at invasive tumor borders. Ablation of mMCP6 expression attenuates polyposis, but invasive properties of the remaining lesions remain intact. Our findings argue for a multistep process in SB carcinogenesis in which distinct MC subsets, and their elaborated proteases, guide disease progression.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3557-3557
Author(s):  
Yukio Kondo ◽  
Luis Espinoza ◽  
Takamasa Katagiri ◽  
Zhirong Qi ◽  
Shinji Nakao

Abstract Abstract 3557 Poster Board III-494 Immunization of allogeneic stem cell transplant (SCT) recipients with leukemia-associated antigens (LAAs) is an attractive approach to the augmentation of graft-versus-leukemia (GVL) effect. However, the induction of CTLs specific to LAAs is hampered by various inhibitory molecules expressed on leukemic cells that restrain the T cell function in connection with their receptors on T cells. Even if the cellular immunity is rebuilt after SCT by T cells of the donor origin, overcoming such an escape mechanism is required to effectively induce the CTLs specific to TAAs by vaccination after allogeneic SCT. Glucocorticoid-induced TNFR-related protein (GITR) belongs to the TNF receptor superfamily and is expressed on NK cells, CD25+ regulatory T cells and activated T cells. The binding of GITR ligand (GITRL) on leukemic cells to GITR on NK cells restrains NK cell activity but the influence on T cells of the GITR/GITRL binding has not been clarified. Myeloid dendritic cells derived from myeloid leukemic cells express GITRL which inhibits induction of LAA-specific CTLs (Blood 2008; 112:817a). The mechanisms of the negative effect on the induction of LAA-specific T cells through the GITR/GITRL interaction was investigated to improve the efficiency of the CTL induction. The expression of GITRL was observed on leukemic cells from 9 of 16 patients with myeloid leukemia and a monocytic leukemia cell line THP-1, and soluble GITRL (sGITRL) was detectable in the serum from 3 of 5 patients as well as in the culture supernatant of THP-1 cells. CFSE-labeled pan T cell, CD4+ T cell and CD8+ T cell proliferation in response to microbeads coated with anti-CD3 and anti-CD28 monoclonal antibodies (CD3/CD28 microbeads) was suppressed to 55.0%, 63.6%, 65.8% of the controls in a culture supernatant of THP-1 cells, and was restored to 86.9%, 65.1% and 76.8% respectively by addition of sGITR to block the binding of sGITRL in the supernatant and GITR on T cells. Flow cytometry detected GITRL in exosomes, which express HLA class II, purified from the culture supernatant of THP-1 with anti-HLA class II antibody-coated microbeads, and CFSE-labeled pan T cell, CD4+ T cell and CD8+ T cell proliferation was restrained as well by the addition of GITRL+ exosomes in a dose dependent manner (27.6%, 54.1%, 27.9% reduction of proliferation with 10 μl exosome, respectively). Indoleamine 2, 3-dioxygenase (IDO) activity in plasmacytoid DC (pDC) is negatively correlated with the activity of CD4+ T cells induced by their interaction with the pDC through the GITR/GITRL interaction in a mouse model. Kynurenine (Kyn), a metabolite of tryptophan in leukemic cells that is broken down by IDO, suppressed CFSE-labeled pan T cell, CD4+ T cell and CD8+ T cell proliferation in response to CD3/CD28 microbeads in a dose dependent manner (24.5%, 12.3%, 18.3% reduction in the proliferation at 100 μM, respectively). Significantly higher concentrations of Kyn were detected in the supernatant of THP-1 cells after incubation in the presence of sGITR than a control, and the production of Kyn was suppressed by the addition of an IDO inhibitor, 1-Methyl Tryptophan (1MT) (Fig). Moreover, the addition of sGITR to leukemic cells from five patients with AML induced Kyn (Fig). These findings indicate that GITRL on leukemic cells and sGITRL secreted by leukemic cells as an exosome protein suppress the induction of LAA-specific CTLs by directly binding GITR on LAA-specific CTLs, increasing the IDO activity in leukemic cells and inducing Kyn secretion from leukemic cells. The administration of anti-IDO agents or anti-GITRL blocking Abs combined with LAA vaccination may therefore effectively induce LAA-specific T cells in SCT recipients. Fig GITR/GITRL binding induces kyn secretion from THP-1 cell and primary AML cells. Fig. GITR/GITRL binding induces kyn secretion from THP-1 cell and primary AML cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3905-3905
Author(s):  
Bernd Jahrsdoerfer ◽  
Karen Dahlke ◽  
Magdalena Hagn ◽  
Kai Sontheimer ◽  
Thamara Beyer ◽  
...  

Abstract Abstract 3905 Immune regulation is central for the development of an efficient cellular immune response. Both Treg cells and plasmacytoid DC can suppress T cell proliferation in a granzyme B (GzmB)-dependent and perforin-independent manner. In the present study we found that, depending on stimulation with interleukin (IL-) 21, B cells (BC) can also express GzmB and effectively suppress T cell proliferation. GzmB expression in BC is enhanced by BC receptor engagement, and is suppressed by CD40 ligation. Since CD4+ T cells are a main source of IL-21, we tested whether they can induce GzmB in BC. We found that incompletely activated CD4+ T cells, but not fully activated T cells induce GzmB in co-cultured BC. Using confocal microscopy, we showed that BC-derived GzmB is enzymatically active and that GzmB+ BC transfer GzmB to CD4+ T cells. Furthermore, GzmB+ BC decreased CD4+ T cell expression of the TCR-zeta chain, a GzmB target, which is required for T cell proliferation. Our results suggest BC may regulate cellular adaptive immune responses by Treg cell-like mechanisms. Inhibition of BC-derived GzmB may represent a novel strategy to induce more effective and comprehensive cellular immune responses. Disclosures: No relevant conflicts of interest to declare.


1991 ◽  
Vol 173 (3) ◽  
pp. 721-730 ◽  
Author(s):  
P S Linsley ◽  
W Brady ◽  
L Grosmaire ◽  
A Aruffo ◽  
N K Damle ◽  
...  

A successful immune response requires intercellular contact between T and B lymphocytes. We recently showed that CD28, a T cell surface protein that regulates an activation pathway, could mediate intercellular adhesion with activated B cells by interaction with the B7 antigen. Here we show that CD28 is the primary receptor for B7 on activated peripheral blood T cells, that CD28 binds to B7 in the absence of other accessory molecules, and that interaction between CD28 and B7 is costimulatory for T cell activation. To characterize the binding of CD28 to B7, we have produced genetic fusions of the extracellular portions of B7 and CD28, and immunoglobulin (Ig) C gamma 1 chains. 125I-labeled B7 Ig bound to CD28-transfected Chinese hamster ovary (CHO) cells, and to immobilized CD28 Ig with a Kd approximately 200 nM. B7 Ig also inhibited CD28-mediated cellular adhesion. The function of CD28-B7 interactions during T cell activation was investigated with soluble fusion proteins and with B7-transfected CHO cells. Immobilized B7 Ig and B7+ CHO cells costimulated T cell proliferation. Stimulation of T cells with B7+ CHO cells also specifically increased levels of interleukin 2 transcripts. These results demonstrate that the CD28 signaling pathway could be activated by B7, resulting in increased T cell cytokine production and T cell proliferation. Cellular interactions mediated by B7 and CD28 may represent an important component of the functional interactions between T and B lymphoid cells.


F1000Research ◽  
2018 ◽  
Vol 7 ◽  
pp. 316 ◽  
Author(s):  
Hridesh Banerjee ◽  
Lawrence P. Kane

T-cell immunoglobulin and mucin domain 3 (Tim-3) is a transmembrane protein that in both mice and humans has been shown to possess various functions in a context-dependent manner. Thus, Tim-3 has been associated with both inhibitory and co-stimulatory function, depending in part on the specific cell type and immune response course. Though originally described on T cells, Tim-3 is now known to be expressed by both lymphoid and non-lymphoid cells within the immune system and even by non-immune cells. In addition, though widely thought of as a negative regulator of immunity, Tim-3 has been shown in more recent studies to have a positive function on both myeloid and lymphoid cells, including T cells. Tim-3 is often expressed at a high level on exhausted T cells in tumors and chronic infection and may engage in crosstalk with other so-called “checkpoint” molecules such as PD-1. Thus, Tim-3 has emerged as a possible therapeutic target, which is being actively explored both pre-clinically and clinically. However, recent research suggests a more complex in vivo role for this protein, compared with other targets in this area.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2423-2423
Author(s):  
Ruishu Deng ◽  
Kaniel Cassady ◽  
Xiaofan Li ◽  
Sheng Yao ◽  
Mingfeng Zhang ◽  
...  

Abstract Interactions of B7H1 (PD-L1) on antigen presenting cells with its two ligands, PD-1 and B7.1, on T cells play a pivotal role in controlling T cell activation, proliferation, anergy, and apoptosis. However, the interactions between the two pathways remain unknown. Using a graft-versus-host disease (GVHD) model of C57BL/6 donor to MHC-mismatched BALB/c recipient, we report here that:1) Wild-type (WT) conventional alloreactive CD4+ T (Tcon) cells induces more severe acute GVHD in B7H1 deficient recipients due to lack of both B7H1/PD-1 and B7H1/B7.1 interaction, resulting in reduced Tcon proliferation and apoptosis and therefore increased expansion of Tcon cells. In contrast, PD-1 deficient Tcon cells induced less severe GVHD in B7H1 deficient recipients compared to WT recipients due to lack of B7H1/B7.1 interaction and reduced Tcon cell proliferation. 2) Specific blockade of the B7H1/B7.1 axis by use of an anti-B7H1 mAb exacerbate acute GVHD. This is because anti-B7H1 mAb reduces donor Tcon cell proliferation, IL-2 production, expression of PD-1, and activation-induced apoptosis, resulting in expansion of alloreactive Tcon cells. In contrast, in the absence of PD-1 on donor T cells, specific blockade of B7H1/B7.1 interaction ameliorates acute GVHD due to reduction of donor Tcon proliferation without impact on Tcon cell apoptosis. 3) B7H1 fused to an immunoglobulin Fc domain (B7H1-Ig), when produced in vivo by hydrodynamic injection of B7H1 plasmid, ameliorates GVHD by augmenting both proliferation and apoptosis of WT- alloreactive Tcon cells. Conversely, in the absence of PD-1 on donor T cells, B7H1-Ig treatment has no impact on apoptosis but augments donor T cell proliferation and worsens GVHD. These results indicate that interaction of B7.1 with B7H1 augments Tcon cell proliferation, IL-2 production, and expression of PD-1. Higher expression of PD-1 leads to increased donor Tcon cell apoptosis mediated by the B7H1/PD1 pathway therefore ameliorates GVHD. But in the absence of PD-1, B7H1/B7.1 interaction only augments T cell proliferation and causes more severe GVHD. Additionally, by engaging both PD-1 and B7.1, B7H1-Ig can be a powerful therapeutic reagent for down-regulating the T cell immune response (This work was supported by NIH R01 AI066008). Disclosures No relevant conflicts of interest to declare.


2015 ◽  
Vol 35 (suppl_1) ◽  
Author(s):  
Iuliia Peshkova ◽  
Petr Makhov ◽  
Vivianly Hou ◽  
Ekaterina Koltsova

Abdominal aortic aneurysm (AAA) is a cardiovascular disease, which is characterized by aortic wall dilation with subsequent rupture and internal bleeding. Inflammatory reactions in the vessel wall are likely essential for AAA pathogenesis, just like they are important for the development of another vascular pathology- atherosclerosis, which can predispose to AAA formation in a context-dependent manner. While inflammatory cytokines were shown to promote atherosclerosis and AAA, little is known about contribution of anti-inflammatory cytokines with regard to their ability to control vascular inflammation. Interleukin (IL)-27 signaling is required to suppress atherosclerosis development but its function in AAA remains unknown. We utilize Angiotensin II (AngII) model to evaluate the role of IL-27R signaling in pathogenesis of AAA. AngII containing pumps were surgically implanted into Il27ra-/- x Ldlr-/-; Il27ra -/- x Apoe-/- and Il27ra+/- littermate controls and AAA progression was analyzed 4 weeks later. Surprisingly, we found attenuated AAA progression in Il27ra-/- mice compared to Il27ra+/- and wt counterparts. The latter developed large AAA with visual hemorrhage into the artery wall, while Il27ra-/- mice developed small AAA with fewer myeloid cells and T cells. Moreover, opposite to aortic arches, T cells in abdominal aortas of Il27ra-/- mice produced less inflammatory IL-17A, while IFNγ production was unchanged. Interestingly, we found enhanced production of “Th2-like” cytokines IL-4 and IL-13, by NK cells and Type 2 Innate lymphoid cells (ILC2) in Il27ra deficient mice, which correlated with the protection from AAA. Overall, we conclude that immunoregulatory cytokine IL-27 can differentially control atherosclerosis and AAA development by regulating innate and adaptive immune cell recruitment and cytokine production in the aortic wall.


Sign in / Sign up

Export Citation Format

Share Document