Ex Vivo Generation Of CD4+ T Cells To Prevent and Treat Infection From Antibiotic-Resistant Klebsiella Pneumoniae In Immunocompromised Patients

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2022-2022 ◽  
Author(s):  
Manuel Franco-Colon ◽  
Kong Chen ◽  
Dhanalakshmi Chinnasamy ◽  
Marianna Sabatino ◽  
David Stroncek ◽  
...  

Abstract Antibiotic resistance is becoming an increasingly significant challenge facing the healthcare system. Klebsiella (K.)pneumoniae is an important cause of Gram-negative nosocomial infections. Recently strains of K. pneumoniae producing carbapenemase (KPC) have emerged worldwide. KPC-Klebsiella infection is a significant problem for stem cell transplantation (SCT) recipients and patients whose immunity is impaired by leukemia, aplastic anemia, cancer or genetic abnormalities of host defense. Immunocompromised patients infected with KPC-Klebsiella (frequently pan-resistant to antibiotics) have few treatment options and face mortality rates over 50%. Novel strategies to improve the odds of survival for these patients are needed. Emerging evidence suggests that, in addition to neutrophil bacterial defense, Th17 cells and IL-17 augment immunity against many bacteria including K. pneumonia. Th17 cells bridge innate and adaptive responses preventing bacterial translocation by maintaining tight intestinal mucosal junctions. IL-17 promotes hematopoietic stem cell function, myelopoesis, and recruitment of myeloid cells. In an animal model, antigen-specific MHC class II-restricted Th17 cells recognizing K. pneumoniae outer membrane protein (OMP) have been described. These T cells provide serotype-independent protective mucosal immunity against K. pneumoniae, including the multi-drug resistant strains. Here we studied the endogenous reactivity of T cells derived peripheral blood of normal healthy donors against Klebsiella antigens. For antigenic stimulation PBL were exposed to recombinant K. pneumoniae outer membrane protein X (OmpX) and lysate of K. pneumoniae serotype 2 (KP). Flow cytometry revealed intracellular production of IL-17A by CD4+ T cells selectively in 4 out of 5 donors upon stimulation with KP lysate and in 3 out of 5 donors upon stimulation with OmpX protein. The observed Th17 reactivity was confined only to the effector memory compartment (TEM), suggesting an antigen-specific mechanism, but not to unrelated peptide libraries (cancer testis antigen SSX2 and CMV pp65) indicating the existence of pre-established antigen-specific immunity against K. pneumoniae in normal healthy donors. Next we tested the feasibility of expanding the Klebsiella-specific CD4+ T cells in vitro under Th17-polarizing conditions. We used irradiated autologous PBMCs as antigen presenting cells. Significant enrichment of OmpX- and KP-specific cells was achieved in 2 out of 4 tested donors following one week antigenic stimulation. Resulting Th effector cells retained a clinically-desirable feature of polyfunctionality in terms of ability to specifically produce not only IL-17A, but also IFN-γ and IL-2. In summary, for the first time we demonstrate that human Th17 cells derived from normal healthy donors can specifically recognize outer membrane proteins derived from K. pneumoniae. Our observation is analogous to findings from an animal model of Th17-mediated serotype-independed immunity against K. pneumoniae. Importantly, we also show that it is feasible to expand the OmpX-reactive CD4+ T cells in vitro. These results raise the possibility of testing the role of adoptively-transferred antibacterial Th17 cells as a novel strategy of augmenting the host defenses of vulnerable patients colonized with antibiotic-resistant bacteria who are at risk for Gram-negative sepsis. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2445-2445
Author(s):  
Pawel Muranski ◽  
Manuel Franco-Colon ◽  
Dhanalakshmi Chinnasamy ◽  
Kong Chen ◽  
Scott Stegemann ◽  
...  

Abstract Klebsiella (K.)pneumoniae is an important cause of Gram-negative nosocomial infections. Recent worldwide emergence of K. pneumoniae producing carbapenemase (KPC) poses a significant problem for stem cell transplantation (SCT) recipients and patients whose immunity is impaired. Such patients have few treatment options and face mortality rates over 50%. Novel strategies are needed. Emerging evidence suggests that, in addition to granulocytes, Th17 cells and IL-17 augment immunity against many bacteria including K. pneumonia. Th17 cells bridge innate and adaptive responses preventing bacterial translocation by maintaining tight intestinal mucosal junctions, promote hematopoietic stem cell function, myelopoiesis, and recruitment of myeloid cells. In an animal model, antigen-specific MHC class II-restricted Th17 cells recognizing K. pneumoniae outer membrane protein (Omp). These T cells provide serotype-independent protective mucosal immunity against K. pneumoniae, including the multi-drug resistant strains. Here we studied the endogenous reactivity of T cells of normal healthy donors against Klebsiella antigens. PBL were exposedtorecombinant K. pneumoniae outer membrane protein X (OmpX) and lysate of K. pneumoniae serotype 2 (KP). Flow cytometry revealed intracellular production of IL-17A by CD4+ T cells selectively in 4 out of 5 donors upon stimulation with KP lysate and in 3 out of 5 donors upon stimulation with OmpX protein. The observed Th17 reactivity was confined only to the effector memory compartment (TEM), suggesting an antigen-specific mechanism, but not to unrelated virus and tumor antigen control peptide libraries indicating pre-established antigen-specific immunity against K. pneumoniae in normal healthy donors. Next the feasibility of expanding the Klebsiella-specific CD4+ T cells in vitro under Th17-polarizing conditions was tested. Magnetic bead purified CD4+ memory T cells from normal donors were co-cultured for 10-14 days with autologous monocytes or irradiated PBMCs pulsed with KP lysate, recombinant OmpX or control peptide libraries derived from BK virus. Cultures were maintained in media containing Th17 polarizing cytokines (TGF-1, IL-1β, IL-6, and IL-23) and supplemented with IL-7, IL-15, and IL-2. Two rounds of stimulation were performed and resulting T cells were tested by FACS for their ability to recognize target antigens. Both KP-lysate and OmpX-stimulated T cells from majority of the donors demonstrated robust antigen-specific production of IL-17A, TNF-α, and IFN-γ upon exposure to respective cognate antigens with some cross-reactivity between them. Control autologous BK-virus specific T cells showed minimal reactivity against KP lysate or OmpX, underscoring antigen-specific nature of observed antibacterial responses. Comparable induction of anti-KP responses was achieved using CD4+ T cells from pre-transplant patients with leukemia, but there was marked impairment when T cells isolated from recipients of allogeneic SCT were used. Importantly, T cells generated using KP serotype 2 lysate demonstrated robust recognition of KP lysate from KP396 (serotype 1), while significantly lower cross-reactivity was observed against lysate from E. coli and no responses were seen upon stimulation with antigens from Gram-positive bacteria (S. aureus and S. pneumoniae), In summary, for the first time we demonstrate that human Th17 cells from healthy donors specifically recognize K. pneumoniae OMP, analogous to the animal model of Th17-mediated serotype-independent immunity against K. pneumoniae. Notably, we efficiently expanded the KP-reactive CD4+ Th17 cells in vitro and these cells recognized KP antigens independently of serotype, indicating the feasibility of targeting simultaneously many strains of KP including the multi-drug-resistant isolates. These results raise the possibility of testing adoptively-transferred antibacterial Th17 cells as a novel strategy to augment host defenses of patients colonized with antibiotic-resistant bacteria at risk for Gram-negative sepsis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2698-2698
Author(s):  
Elena E. Solomou ◽  
A. Tsanaktsi ◽  
V. Fertakis ◽  
K. Dallas ◽  
S. Karambina ◽  
...  

Abstract IL17-producing T cells have been recently described as a distinct T cell helper population (Th17 cells) characterized by expression of membrane CD4 and IL23R and intracellular expression of the orphan nuclear receptor RORgt. In Th17 cells the transcription factor RORgt induces the transcription of IL17 gene, whereas in Th1 cells the transcription factor Tbet is responsible for the transcription of IFNg gene. Th1 along with Th17 cells are thought to contribute to the pathogenesis of autoimmune diseases. In murine models Th17 cells are fully polarized. In humans a proportion of Th17 cells are also positive for interferon gamma (IFN-g); they are named Th1/17 cells and their function is yet unclear. In patients with colitis and seronegative arthritis Th17 cells are increased. The induction of Th17 and Th1/17 in patients with MDS has not been previously evaluated. To examine the expression of Th17 and Th1/17 cells in this disease, peripheral blood mononuclear cells (PBMC) from patients with MDS were cultured in vitro for 6 days in RPMI-1640, 15% FBS supplemented with PHA (0.1 μg/mL) and IL-2 (10 ng/mL). Percentages of CD4+IL23R+IL-17+ T cells (Th17) and CD4+IL23R+IL17+IFN-g+ T cells (Th1/17) in patients with MDS were determined by flow cytometry: Th17 cells were markedly increased in patients (n=30) compared to healthy controls (n=15), (17.5% ± 3.4 vs 2.5% ± 0.4, p=0.008). Th1/Th17 cells were also significantly increased in MDS patients compared to controls (15.17% ± 2.80 vs 2.56% ± 0.80, p=0.008). None of the patients had been on immunosuppressive treatment or transfused before sampling. In multi-transfused patients with no underlying hematologic disease examined (n=3) the Th17 and Th1/17 populations were comparable to those of healthy donors. In patients with MDS the majority of the Th17 cells expressed also IFNg (90.07% ± 2.87) whereas in healthy controls only 59.7% ± 5.5 of the Th17 cells were also positive for IFNg (p<0.0001). There were no differences between different subtypes of MDS (RA, RARS, and RAEB). Using confocal microscopy, purified CD4+ T cells from PBMC cultures from patients (n=5) showed increased Tbet and RORgt expression at the single-cell level compared to controls (n=3),(T-bet: 22.03 ± 1.20 vs 11.60 ± 0.35 arbitrary units respectively, p<0.0001 and RORãt: 28.90 ± 0.35 vs 21.03 ± 1.20 arbitrary units, p=0.0008. For each sample 100 cells were analyzed). We next asked whether kinases involved in the induction of Tbet are also involved in the induction of RORgt. We analyzed the effects of rottlerin, a PKC-theta inhibitor, SB203580, a p38 MAPK pathway inhibitor, and PD98059, an ERK pathway inhibitor, on Th17 and Th1/17 cell induction in patients (n=7) and controls (n=4). Rottlerin decreased the Th17 content in patients and controls by 45.0%, and the Th1/17 content by 64.8%. SB203580 showed a 17% and 18% decrease on Th17 and on Th1/17 content, respectively, in patients and controls. PD98059 showed no effect on Th17 and Th1/17 populations in patients and controls. By immunoblots, in normal CD4+T cells rottlerin decreased both T-bet and RORgt protein levels by 50% and 20%, respectively. SB203580, decreased RORgt levels by 25%, and PD98059 did not obviously decrease Tbet but decreased RORgt levels by 20%. CD4+IL23R+IL-17+ T cells and CD4+IL23R+IL17+IFN-g+ T cells are increased in most patients with MDS. T cells have recently been implicated in MDS pathogenesis. Although more studies are needed in order to define the role of Th17 and Th1/17 cells in the pathogenesis of MDS, our in vitro data with the kinase inhibitors may suggest a probable therapeutic target for patients with MDS.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 468-468
Author(s):  
Pawel Muranski ◽  
Sid P Kerkar ◽  
Zachary A Borman ◽  
Robert Reger ◽  
Luis Sanchez-Perez ◽  
...  

Abstract Abstract 468 We have recently demonstrated that Th17-polarized TCR transgenic CD4+ T cells specific for TRP-1 melanoma antigen are superior to Th1-polarized cells in mediating effective anti-tumor responses against advanced disease after adoptive transfer. The therapeutic activity of Th17-skewed cells is critically dependent on their ability to secrete IFN-γ, suggesting that the Th17 subset might evolve in vivo. However, the developmental program of Th17-polarized cells in vivo remains substantially un- elucidated. We developed a novel TCR-transduction technique that enabled us to rapidly confer specificity for a cognate antigen upon any population of T cells, regardless of its genetic background, its previous polarization history or its state of differentiation. Using adoptive transfers into tumor-bearing hosts, we were able to study the functionality of these genetically-engineered T cells in vivo. In vitro, CD4+ T cells cultured in type 17 conditions acquired end-effector phenotype (CD62Llow, CD45RBlow), but proliferated slower than cells grown in type 1 condition. Thus, we hypothesized that Th17-polarized cells might represent a less mature, more central-memory like subset. This notion was supported by their ability to secrete high quantities of IL-2 and higher expression of IL-7 receptor. In contrast, Th1-polarized cells upon in vitro re-stimulation upregulated PRDM1 that encodes BLIMP1, a molecule associated with the end-effector senescent phenotype. Moreover, Th1-skewed cells overexpressed caspase 3 and were prone to activation-induced cell death as measured by annexin V assay, while type 17 cells were resistant to apoptosis, and robustly expanded in secondary cultures. Using the TCR gene transfer technique we tested the treatment outcomes when Th17-polarized cells deficient for IL-17A were used. In contrast to wild-type (WT)-derived Th17 cells that effectively eradicated established tumors, we observed significant impairment of treatment with IL-17A-deficent cells. Similarly, we observed reduction in treatment efficacy when CCR6-deficient Th17 cells were transferred. CCR6 is a receptor for CCL20, a chemokine highly induced Th17 cells and thought to contribute to the trafficking of those cells to the site of inflammation. In both cases however, the addition of exogenous vaccination and IL-2 significantly improved treatment efficacy. Thus, we concluded that Th17-associated factors play the role in the anti-cancer activity of type 17 cells. To address the question whether plasticity of Th17-skewed effectors is important for their function upon ACT, we treated animals with TCR-transduced Th17-skewed cells derived from IFN-γ-deficient CD4+ cells as well as from t-bet-deficient mice, which are not able to develop type 1 responses. In contrast to WT-derived Th17 effectors, IFN-γ-deficient cells did not show any anti-tumor activity, while t-bet-deficient Th17 cells were able to mediate only minimal delay in tumor growth, suggesting that indeed the capacity to acquire Th1-like properties is essential for the anti-tumor function of Th17-skewed lymphocytes. Overall, here we demonstrate that TCR gene engineered Th17-polarized cells can efficiently treat advanced tumor. The high activity of in vitro-generated anti-tumor Th17 cells relies on the contribution of type 17-associated characteristics, including both the secretion of inflammatory factors IL-17A and CCL20, as well as the superior capacity to survive and expand upon the secondary stimulation. Importantly however, type 1-defining t-bet-mediated plasticity in the lineage commitment is required for the full therapeutic effect, underscoring the dualistic nature of Th17-skewed cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 711-711
Author(s):  
Srimoyee Ghosh ◽  
Sergei B Koralov ◽  
Irena Stevanovic ◽  
Mark S Sundrud ◽  
Yoshiteru Sasaki ◽  
...  

Abstract Abstract 711 Naïve CD4 T cells differentiate into diverse effector and regulatory subsets to coordinate the adaptive immune response. TH1 and TH2 effector subsets produce IFN-γ and IL-4, respectively, whereas proinflammatory TH17 cells are key regulators of autoimmune inflammation, characteristically produce IL-17 and IL-22 and differentiate in the presence of inflammatory cytokines like IL-6 and IL-21 together with TGF-β. Naive T cells can also differentiate into tissue-protective induced T regulatory (iTreg) cells. NFAT proteins are highly phosphorylated and reside in the cytoplasm of resting cells. Upon dephosphorylation by the Ca2+/calmodulin-dependent serine phosphatase calcineurin, NFAT proteins translocate to the nucleus, where they orchestrate developmental and activation programs in diverse cell types. In this study, we investigated the role of the Ca/NFAT signaling pathway in regulating T cell differentiation and the development of autoimmune diseases. We generated transgenic mice conditionally expressing a hyperactivable version of NFAT1 (AV-NFAT1) from the ROSA26 locus. To restrict AV-NFAT1 expression to the T cell compartment, ROSA26-AV-NFAT1 transgenic mice were bred to CD4-Cre transgenic mice. Naïve CD4 T cells freshly isolated from AV mice produced significantly less IL-2 but increased amounts of the inhibitory cytokine IL-10. To investigate the role of NFAT1 in the generation of TH1, TH2, Tregand TH17 cells, the respective cell types were generated from CD4 T cells of AV mice by in vitro differentiation. T cells from AV-NFAT1 mice exhibited a dysregulation of cytokine expression, producing more IFN-γ and less IL-4. While the numbers of CD4+CD25+ “natural” Treg cells in peripheral lymphoid organs and their in vitro suppressive functions were slightly decreased in AV mice, iTreg generation from CD4+CD25- T cells of AV mice as compared to wild type cells was markedly enhanced. Moreover, TH17 cells generated in vitro from CD4 T cells of AV mice in the presence of IL-6, IL-21 and TGF-β exhibited dramatically increased expression of both IL-10 and IL-17 as compared to wild type controls. To investigate putative NFAT binding sites in the IL-10 and IL-17 gene loci, we performed chromatin immunoprecipitation experiments. We show that NFAT1 can bind at the IL-17 locus at 3 out of 9 CNS regions which are accessible specifically during TH17 but not during TH1 and TH2 differentiation. Furthermore, we provide evidence that NFAT1 binds one CNS region in the IL10-locus in TH17 cells. To verify our observations in vivo, we induced experimental autoimmune encephalitis (EAE) in AV mice and wild type controls with the immunodominant myelin antigen MOG33-55 emulsified in complete Freund‘s adjuvant. While wild type animals showed a normal course of disease with development of tail and hind limb paralysis after approximately 10 days, AV mice showed a markedly weaker disease phenotype with less severe degrees of paralysis and accelerated kinetics of remission. Moreover at the peak of the response, there were fewer CD4+CD25- but more CD4+CD25+ T cells in the CNS of AV animals compared to wild type controls. Surprisingly, these cells produced significantly more IL-2, IL-17 and IFN-γ upon restimulation, even though they displayed decreased disease. In summary, our data provide strong evidence that NFAT1 contributes to the regulation of IL-10 and IL-17 expression in TH17 cells and show that increasing NFAT1 activity can ameliorate autoimmune encephalitis. This could occur in part through upregulation of IL-10 expression as observed in vitro, but is also likely to reflect increased infiltration of regulatory T cells into the CNS as well as increased conversion of conventional T cells into Foxp3+ regulatory T cells within the CNS. Disclosures: No relevant conflicts of interest to declare.


Cells ◽  
2019 ◽  
Vol 8 (6) ◽  
pp. 533 ◽  
Author(s):  
Alessia Capone ◽  
Manuela Bianco ◽  
Gabriella Ruocco ◽  
Marco De Bardi ◽  
Luca Battistini ◽  
...  

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). T helper (Th) 17 lymphocytes play a role in the pathogenesis of MS. Indeed, Th17 cells are abundant in the cerebrospinal fluid and peripheral blood of MS patients and promote pathogenesis in the mouse model of MS. To gain insight into the function of Th17 cells in MS, we tested whether Th17 cells polarized from naïve CD4 T cells of healthy donors and MS patients display different features. To this end, we analysed several parameters that typify the Th17 profile during the differentiation process of naïve CD4 T cells obtained from relapsing-remitting (RR)-MS patients (n = 31) and healthy donors (HD) (n = 28). Analysis of an array of cytokines produced by Th17 cells revealed that expression of interleukin (IL)-21, tumour necrosis factor (TNF)-β, IL-2 and IL-1R1 is significantly increased in Th17 cells derived from MS patients compared to healthy donor-derived cells. Interestingly, IL-1R1 expression is also increased in Th17 cells circulating in the blood of MS patients compared to healthy donors. Since IL-2, IL-21, TNF-β, and IL-1R1 play a crucial role in the activation of immune cells, our data indicate that high expression of these molecules in Th17 cells from MS patients could be related to their high inflammatory status.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3482-3482
Author(s):  
Minghui Li ◽  
Kai Sun ◽  
Mark Hubbard ◽  
Doug Redelman ◽  
Angela Panoskaltsis-Mortari ◽  
...  

Abstract IL-17-producing CD4 T cells (Th17) are a recently identified T helper subset that plays a role in mediating host defense to extracellular bacteria infections and is involved in the pathogenesis of many autoimmune diseases. In vitro induction of IL-17 in murine CD4+ T cells has been shown to be dependent on the presence of the proinflammatory cytokines TGF-β and IL-6 whereas IFNγ can suppress the development of Th17 cells. In the current study, we examined the roles of TNFα and IFNγ on IL-17 production by purified T cells in vitro and in vivo after allogeneic bone marrow transplantation (BMT). We present findings that expression of TNFα by the T cell itself is necessary for optimal development of Th17 under in vitro polarizing conditions. A novel role for T cell-derived TNFα in Th17 induction was observed when in vitro polarization of Tnf−/−CD4+ T cells resulted in marked reductions in IL-17+CD4+ T cells compared to Tnf+/+CD4+ T cells. In marked contrast, T cell-derived IFNγ markedly inhibited Th17 development as more IL-17+CD4+ T cells were found in Ifnγ−/−CD4+ T cells than in Ifnγ+/+CD4+ T cells, and of particular interest was the dramatic increase in IL-17+CD8+ cells from Ifnγ−/− mice. To determine if T cell-derived TNFα or IFNγ can regulate Th17 development in vivo we examined the differentiation of alloreactive donor T cells following allogeneic BMT. We have found that donor-derived Th17 cells can be found in lymphoid tissues and GVHD-affected organs after allogeneic BMT. However, transfer of Tnf−/− CD4+ T cells after allogeneic BMT resulted in marked reductions in Th17 cells in the spleen (18×103 vs 7×103, P<0.05). In agreement with the in vitro data and in contrast to what was observed with transfer of Tnf−/− CD4+ T cells, transfer of donor Ifnγ−/− T cells resulted in marked increases in not only IL-17+CD4+ but also IL-17+CD8+ T cells infiltrating the liver (7×103 vs 14×103, P<0.05; 4×104 vs 12.5×104, P<0.05). These results suggest that the donor T cell-derived TNFα and IFNγ opposingly regulate IL-17 induction of both CD4+ and CD8+ T cells in vitro and after allogeneic BMT which correlates with GVHD pathology.


Cells ◽  
2021 ◽  
Vol 10 (11) ◽  
pp. 3039
Author(s):  
Mikołaj Nawrocki ◽  
Niels Lory ◽  
Tanja Bedke ◽  
Friederike Stumme ◽  
Björn-Phillip Diercks ◽  
...  

Nicotinic acid adenine dinucleotide phosphate (NAADP) is the most potent Ca2+ mobilizing agent and its inhibition proved to inhibit T-cell activation. However, the impact of the NAADP signaling on CD4+ T-cell differentiation and plasticity and on the inflammation in tissues other than the central nervous system remains unclear. In this study, we used an antagonist of NAADP signaling, trans-Ned 19, to study the role of NAADP in CD4+ T-cell differentiation and effector function. Partial blockade of NAADP signaling in naïve CD4+ T cells in vitro promoted the differentiation of Th17 cells. Interestingly, trans-Ned 19 also promoted the production of IL-10, co-expression of LAG-3 and CD49b and increased the suppressive capacity of Th17 cells. Moreover, using an IL-17A fate mapping mouse model, we showed that NAADP inhibition promotes conversion of Th17 cells into regulatory T cells in vitro and in vivo. In line with the results, we found that inhibiting NAADP ameliorates disease in a mouse model of intestinal inflammation. Thus, these results reveal a novel function of NAADP in controlling the differentiation and plasticity of CD4+ T cells.


2021 ◽  
Vol 11 ◽  
Author(s):  
Jianping Hu ◽  
Shenglan Yi ◽  
Chaokui Wang ◽  
Yiting Zhang ◽  
Jihong Tang ◽  
...  

A20 is a negative regulator of inflammation and immunity and plays a role in several autoimmune and inflammatory diseases. Here, we demonstrate that A20 overexpression significantly ameliorates severity of EAU by inhibiting the infiltration of Th1 and Th17 cells, and by protecting integrity of the blood retinal barrier. In vitro studies showed that A20 silencing could promote CD4+T cells toward a Th1 and Th17 phenotype. A decreased expression of A20 in CD4+T cells was noticed in active BD patients but not in VKH patients. Furthermore, silencing of A20 in hRPE cells induced the production of IL-6, IL-8, and MCP-1 and downregulated ZO-1 and occludin expression which is mediated by inhibition of MAPK and NF-κB pathways. This study reveals a mechanism by which A20 prevents autoimmune uveitis.


2021 ◽  
Vol 23 (1) ◽  
pp. 177
Author(s):  
Aoi Okubo ◽  
Youhei Uchida ◽  
Yuko Higashi ◽  
Takuya Sato ◽  
Youichi Ogawa ◽  
...  

Th17 cells play an important role in psoriasis. The differentiation of naïve CD4+ T cells into Th17 cells depends on glycolysis as the energy source. CD147/basigin, an integral transmembrane protein belonging to the immunoglobulin superfamily, regulates glycolysis in association with monocarboxylate transporters (MCTs)-1 and -4 in cancer cells and T cells. We examined whether CD147/basigin is involved in the pathogenesis of psoriasis in humans and psoriasis-model mice. The serum level of CD147 was increased in patients with psoriasis, and the expression of CD147 and MCT-1 was elevated in their dermal CD4+ RORγt+ T cells. In vitro, the potential of naïve CD4+ T cells to differentiate into Th17 cells was abrogated in CD147−/− T cells. Imiquimod (IMQ)-induced psoriatic dermatitis was significantly milder in CD147−/− mice and bone marrow chimeric mice lacking CD147 in the hematopoietic cells of myeloid lineage. These findings demonstrate that CD147 is essential for the development of psoriasis via the induction of Th17 cell differentiation.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 452-452
Author(s):  
Jianing Fu ◽  
Dapeng Wang ◽  
Yu Yu ◽  
Kane Kaosaard ◽  
Chen Liu ◽  
...  

Abstract Abstract 452 Background: Allogeneic hematopoietic cell transplantation (HCT) offers great promise for the treatment of hematologic malignancies. However, HCT benefits are frequently offset by graft-versus-host disease (GVHD). Donor T cells can differentiate into Th1 or Th17 subset that contribute to GVHD. The T-box transcription factor T-bet is important for promoting the differentiation of naïve CD4+T cells into Th1 phenotype, while simultaneously inhibiting Th2 and Th17 lineage commitment. Published data indicate that donor T cells deficient for IFN-γ induce exacerbated GVHD. In contrast, our recent study showed that T cells deficient for T-bet were impaired in the induction of GVHD. Given T-bet is a master regulator for the differentiation into Th1 cells that produce IFN-γ, the underlining mechanisms accounted for the distinct outcomes caused by T-bet- versus IFN-γ-deficient donor T cells are not clear. Method: We evaluated the roles of T-bet and IFN-γ in acute GVHD induced by naïve CD4+ T cells or polarized Th17 cells using murine allogeneic bone marrow transplantation (allo-BMT) model. WT, T-bet knockout (KO) and IFN-γ KO mice on C57BL/6 (B6) background were used as donors, and lethally irradiated BALB/c mice were used as recipients. Pathologic analysis and serum cytokine detection were done 14 days after adoptive transfer of WT, T-bet–/–, and IFN-γ–/– CD4+ T cells. Using microarray technology, gene expression profile on donor T cells was analyzed 7 days after adoptive transfer by sorting donor-derived CD4+ T cells from the recipients of WT, T-bet–/– or IFN-γ–/– CD4+ T cells. Results: We compared the ability of WT, T-bet–/–, and IFN-γ–/– CD4 T cells in the induction of acute GVHD. In the comparison with WT cells, IFN-γ–/– CD4 T cells caused similar or even more severe GVHD as expected. In sharp contrast, T-bet–/– CD4 T cells induced much ameliorated GVHD, as significantly higher survival and less body weight loss were observed in the recipients of T-bet–/–T cells. Pathology study on GVHD target organs showed that recipients of T-bet–/– donor T cells had markedly reduced T cell infiltration and tissue damage in liver, gut, and skin, when compared with those of WT or IFN-γ–/– T cells. Reduced GVHD in the recipients of T-bet–/– T cells was consistent with significantly lower levels of pathogenic cytokines IFN-γ, TNF-α, and IL-2 but higher IL-10 (anti-inflammatory), IL-6 (Th17 related) and IL-4 (Th2 related) in serum as compared with those in the recipients of WT T cells. Mechanistic studies in vitro revealed that T-bet–/– CD4 T cells expressed significantly lower levels of IFN-γ, CXCR3 (Th1 specific chemokine receptor) and CD122 (T cell activation marker), but higher levels of IL-17 (Th17 cytokine) and CCR6 (Th17 specific chemokine receptor) compared with WT CD4 T cells, indicating that T-bet–/– T cells impaired in differentiating into Th1 cells and instead into Th17 cells. Given Th17 subset only is capable of causing GVHD and T-bet–/– T cells are prone to Th17-differentiation, we assessed the role of T-bet or IFN-γ in the development of GVHD by comparing the pathogenicity of in vitro polarized WT, T-bet–/– and IFN-γ–/– Th17 cells. While IFN-γ–/– Th17 cells had a comparable ability to cause GVHD compared with WT Th17 cells, T-bet–/– Th17 cells had reduced pathogenicity, and caused ameliorated GVHD. Furthermore, microarray analysis identified genes that are regulated by T-bet but independent of IFN-γ, including Cxcr3, Ccr5, Ccl3, Ccl4, Klrc1, Klrd1, Nkg7 and Pdcd1, which may explain the compromised ability of T-bet−/− not IFN-γ–/–T cells in the induction of GVHD. Conclusions: We conclude that T-bet is required for Th1 differentiation and optimal function of Th17 cells, and it can also control T cell infiltration into GVHD target organs by regulating chemokines and their receptors. Thus, failure in Th1 generation, migration and reduced activity of polarized Th17 cells are likely accounted for impaired ability of T-bet−/− CD4 T cells in the induction of acute GVHD. The current study suggests that targeting T-bet or regulating its downstream effectors independent of IFN-γ may be a promising strategy to control GVHD after allogeneic HCT in clinic. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document