Browsing by Author "Li, Qi-Jing"
Results Per Page
Sort Options
Item Open Access A conjoined universal helper epitope can unveil antitumor effects of a neoantigen vaccine targeting an MHC class I-restricted neoepitope.(NPJ vaccines, 2021-01-18) Swartz, Adam M; Congdon, Kendra L; Nair, Smita K; Li, Qi-Jing; Herndon, James E; Suryadevara, Carter M; Riccione, Katherine A; Archer, Gary E; Norberg, Pamela K; Sanchez-Perez, Luis A; Sampson, John HPersonalized cancer vaccines targeting neoantigens arising from somatic missense mutations are currently being evaluated for the treatment of various cancers due to their potential to elicit a multivalent, tumor-specific immune response. Several cancers express a low number of neoantigens; in these cases, ensuring the immunotherapeutic potential of each neoantigen-derived epitope (neoepitope) is crucial. In this study, we discovered that therapeutic vaccines targeting immunodominant major histocompatibility complex (MHC) I-restricted neoepitopes require a conjoined helper epitope in order to induce a cytotoxic, neoepitope-specific CD8+ T-cell response. Furthermore, we show that the universally immunogenic helper epitope P30 can fulfill this requisite helper function. Remarkably, conjoined P30 was able to unveil immune and antitumor responses to subdominant MHC I-restricted neoepitopes that were, otherwise, poorly immunogenic. Together, these data provide key insights into effective neoantigen vaccine design and demonstrate a translatable strategy using a universal helper epitope that can improve therapeutic responses to MHC I-restricted neoepitopes.Item Open Access Antagonizing the irreversible thrombomodulin-initiated proteolytic signaling alleviates age-related liver fibrosis via senescent cell killing.(Cell research, 2023-07) Pan, Christopher C; Maeso-Díaz, Raquel; Lewis, Tylor R; Xiang, Kun; Tan, Lianmei; Liang, Yaosi; Wang, Liuyang; Yang, Fengrui; Yin, Tao; Wang, Calvin; Du, Kuo; Huang, De; Oh, Seh Hoon; Wang, Ergang; Lim, Bryan Jian Wei; Chong, Mengyang; Alexander, Peter B; Yao, Xuebiao; Arshavsky, Vadim Y; Li, Qi-Jing; Diehl, Anna Mae; Wang, Xiao-FanCellular senescence is a stress-induced, stable cell cycle arrest phenotype which generates a pro-inflammatory microenvironment, leading to chronic inflammation and age-associated diseases. Determining the fundamental molecular pathways driving senescence instead of apoptosis could enable the identification of senolytic agents to restore tissue homeostasis. Here, we identify thrombomodulin (THBD) signaling as a key molecular determinant of the senescent cell fate. Although normally restricted to endothelial cells, THBD is rapidly upregulated and maintained throughout all phases of the senescence program in aged mammalian tissues and in senescent cell models. Mechanistically, THBD activates a proteolytic feed-forward signaling pathway by stabilizing a multi-protein complex in early endosomes, thus forming a molecular basis for the irreversibility of the senescence program and ensuring senescent cell viability. Therapeutically, THBD signaling depletion or inhibition using vorapaxar, an FDA-approved drug, effectively ablates senescent cells and restores tissue homeostasis in liver fibrosis models. Collectively, these results uncover proteolytic THBD signaling as a conserved pro-survival pathway essential for senescent cell viability, thus providing a pharmacologically exploitable senolytic target for senescence-associated diseases.Item Open Access Branched-Chain Amino Acid Accumulation Fuels the Senescence-Associated Secretory Phenotype.(Advanced science (Weinheim, Baden-Wurttemberg, Germany), 2023-11) Liang, Yaosi; Pan, Christopher; Yin, Tao; Wang, Lu; Gao, Xia; Wang, Ergang; Quang, Holly; Huang, De; Tan, Lianmei; Xiang, Kun; Wang, Yu; Alexander, Peter B; Li, Qi-Jing; Yao, Tso-Pang; Zhang, Zhao; Wang, Xiao-FanThe essential branched-chain amino acids (BCAAs) leucine, isoleucine, and valine play critical roles in protein synthesis and energy metabolism. Despite their widespread use as nutritional supplements, BCAAs' full effects on mammalian physiology remain uncertain due to the complexities of BCAA metabolic regulation. Here a novel mechanism linking intrinsic alterations in BCAA metabolism is identified to cellular senescence and the senescence-associated secretory phenotype (SASP), both of which contribute to organismal aging and inflammation-related diseases. Altered BCAA metabolism driving the SASP is mediated by robust activation of the BCAA transporters Solute Carrier Family 6 Members 14 and 15 as well as downregulation of the catabolic enzyme BCAA transaminase 1 during onset of cellular senescence, leading to highly elevated intracellular BCAA levels in senescent cells. This, in turn, activates the mammalian target of rapamycin complex 1 (mTORC1) to establish the full SASP program. Transgenic Drosophila models further indicate that orthologous BCAA regulators are involved in the induction of cellular senescence and age-related phenotypes in flies, suggesting evolutionary conservation of this metabolic pathway during aging. Finally, experimentally blocking BCAA accumulation attenuates the inflammatory response in a mouse senescence model, highlighting the therapeutic potential of modulating BCAA metabolism for the treatment of age-related and inflammatory diseases.Item Open Access Cancer-cell-derived GABA promotes β-catenin-mediated tumour growth and immunosuppression.(Nature cell biology, 2022-02) Huang, De; Wang, Yan; Thompson, J Will; Yin, Tao; Alexander, Peter B; Qin, Diyuan; Mudgal, Poorva; Wu, Haiyang; Liang, Yaosi; Tan, Lianmei; Pan, Christopher; Yuan, Lifeng; Wan, Ying; Li, Qi-Jing; Wang, Xiao-FanMany cancers have an unusual dependence on glutamine. However, most previous studies have focused on the contribution of glutamine to metabolic building blocks and the energy supply. Here, we report that cancer cells with aberrant expression of glutamate decarboxylase 1 (GAD1) rewire glutamine metabolism for the synthesis of γ-aminobutyric acid (GABA)-a prominent neurotransmitter-in non-nervous tissues. An analysis of clinical samples reveals that increased GABA levels predict poor prognosis. Mechanistically, we identify a cancer-intrinsic pathway through which GABA activates the GABAB receptor to inhibit GSK-3β activity, leading to enhanced β-catenin signalling. This GABA-mediated β-catenin activation both stimulates tumour cell proliferation and suppresses CD8+ T cell intratumoural infiltration, such that targeting GAD1 or GABABR in mouse models overcomes resistance to anti-PD-1 immune checkpoint blockade therapy. Our findings uncover a signalling role for tumour-derived GABA beyond its classic function as a neurotransmitter that can be targeted pharmacologically to reverse immunosuppression.Item Open Access Definition of a murine CD8+ MHCII-recognizing effector T cell population(2019) Robins, ElizabethCD4+ and CD8+ T cells are dichotomous lineages in adaptive immunity. While conventionally viewed as distinct fates that are fixed post-thymic development, accumulating evidence indicates that these two populations can exhibit significant lineage plasticity, particularly upon TCR-mediated activation. In this work, I define a novel murine CD4-CD8ab+ MHCII-recognizing population generated from effector CD4+ T cells. Effector CD4-CD8ab+ MHCII-recognizing T cells downregulate expression of T helper cell-associated costimulatory molecules and increase expression of cytotoxic T lymphocyte-associated cytotoxic molecules. TCRb repertoire sequencing and in vivo genetic lineage tracing in acutely-infected wild-type mice demonstrate fundamental lineage reprogramming resulting in an “ex-CD4” T cell phenotype, rather than expansion of an aberrantly-developed CD8+ MHCII-restricted population. Impairing autophagy by functional deletion of the initiating kinase Vps34 or a downstream enzyme Atg7 dramatically enhances the generation of effector CD4-CD8ab+ MHCII-recognizing T cells. These findings suggest that effector CD4+ T cells can exhibit a previously unreported degree of skewing towards the CD8+ T cell lineage, which may point towards a novel direction for vaccine design, particularly against HIV.
Item Open Access Diversity index of mucosal resident T lymphocyte repertoire predicts clinical prognosis in gastric cancer(OncoImmunology, 2015-01-01) Jia, Qingzhu; Zhou, Junfeng; Chen, Gang; Shi, Yan; Yu, Haili; Guan, Peng; Lin, Regina; Jiang, Ning; Yu, Peiwu; Li, Qi-Jing; Wan, Ying© 2015 Taylor & Francis Group, LLC.A characteristic immunopathology of human cancers is the induction of tumor antigen-specific T lymphocyte responses within solid tumor tissues. Current strategies for immune monitoring focus on the quantification of the density and differentiation status of tumor-infiltrating T lymphocytes; however, properties of the TCR repertoire - including antigen specificity, clonality, as well as its prognostic significance β remain elusive. In this study, we enrolled 28 gastric cancer patients and collected tumor tissues, adjacent normal mucosal tissues, and peripheral blood samples to study the landscape and compartmentalization of these patients’ TCR β repertoire by deep sequencing analyses. Our results illustrated antigen-driven expansion within the tumor compartment and the contracted size of shared clonotypes in mucosa and peripheral blood. Most importantly, the diversity of mucosal T lymphocytes could independently predict prognosis, which strongly underscores critical roles of resident mucosal T-cells in executing post-surgery immunosurveillance against tumor relapse.Item Open Access HLA class II-Restricted CD8+ T cells in HIV-1 Virus Controllers.(Scientific reports, 2019-07-15) Nyanhete, Tinashe E; Frisbee, Alyse L; Bradley, Todd; Faison, William J; Robins, Elizabeth; Payne, Tamika; Freel, Stephanie A; Sawant, Sheetal; Weinhold, Kent J; Wiehe, Kevin; Haynes, Barton F; Ferrari, Guido; Li, Qi-Jing; Moody, M Anthony; Tomaras, Georgia DA paradigm shifting study demonstrated that induction of MHC class E and II-restricted CD8+ T cells was associated with the clearance of SIV infection in rhesus macaques. Another recent study highlighted the presence of HIV-1-specific class II-restricted CD8+ T cells in HIV-1 patients who naturally control infection (virus controllers; VCs). However, questions regarding class II-restricted CD8+ T cells ontogeny, distribution across different HIV-1 disease states and their role in viral control remain unclear. In this study, we investigated the distribution and anti-viral properties of HLA-DRB1*0701 and DQB1*0501 class II-restricted CD8+ T cells in different HIV-1 patient cohorts; and whether class II-restricted CD8+ T cells represent a unique T cell subset. We show that memory class II-restricted CD8+ T cell responses were more often detectable in VCs than in chronically infected patients, but not in healthy seronegative donors. We also demonstrate that VC CD8+ T cells inhibit virus replication in both a class I- and class II-dependent manner, and that in two VC patients the class II-restricted CD8+ T cells with an anti-viral gene signature expressed both CD4+ and CD8+ T cell lineage-specific genes. These data demonstrated that anti-viral memory class II-restricted CD8+ T cells with hybrid CD4+ and CD8+ features are present during natural HIV-1 infection.Item Open Access IFI16-STING-NF-κB signaling controls exogenous mitochondrion-induced endothelial activation.(American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, 2022-06) Li, Shu; Xu, He; Song, Mingqing; Shaw, Brian I; Li, Qi-Jing; Kirk, Allan DMitochondria released from injured cells activate endothelial cells (ECs), fostering inflammatory processes, including allograft rejection. The stimulator of interferon genes (STING) senses endogenous mitochondrial DNA, triggering innate immune activation via NF-κB signaling. Here, we show that exogenous mitochondria exposure induces EC STING-NF-κB activation, promoting EC/effector memory T cell adhesion, which is abrogated by NF-κB and STING inhibitors. STING activation in mitochondrion-activated ECs is independent of canonical cGMP-AMP synthetase sensing/signaling, but rather is mediated by interferon gamma-inducible factor 16 (IFI16) and can be inhibited by IFI16 inhibition. Internalized mitochondria undergo mitofusion and STING-dependent mitophagy, leading to selective sequestration of internalized mitochondria. The exposure of donor hearts to exogenous mitochondria activates murine heart ECs in vivo. Collectively, our results suggest that IFI16-STING-NF-κB signaling regulates exogenous mitochondrion-induced EC activation and mitophagy, and exogenous mitochondria foster T cell-mediated CoBRR. These data suggest a novel, donor-directed, therapeutic approach toward mitigating perioperative allograft immunogenicity.Item Open Access MiR-215 Is Induced Post-transcriptionally via HIF-Drosha Complex and Mediates Glioma-Initiating Cell Adaptation to Hypoxia by Targeting KDM1B.(Cancer Cell, 2016-01-11) Hu, Jing; Sun, Tao; Wang, Hui; Chen, Zhengxin; Wang, Shuai; Yuan, Lifeng; Liu, Tingyu; Li, Hai-Ri; Wang, Pingping; Feng, Yukuan; Wang, Qinhong; McLendon, Roger E; Friedman, Allan H; Keir, Stephen T; Bigner, Darell D; Rathmell, Jeff; Fu, Xiang-Dong; Li, Qi-Jing; Wang, Huibo; Wang, Xiao-FanThe hypoxic tumor microenvironment serves as a niche for maintaining the glioma-initiating cells (GICs) that are critical for glioblastoma (GBM) occurrence and recurrence. Here, we report that hypoxia-induced miR-215 is vital for reprograming GICs to fit the hypoxic microenvironment via suppressing the expression of an epigenetic regulator KDM1B and modulating activities of multiple pathways. Interestingly, biogenesis of miR-215 and several miRNAs is accelerated post-transcriptionally by hypoxia-inducible factors (HIFs) through HIF-Drosha interaction. Moreover, miR-215 expression correlates inversely with KDM1B while correlating positively with HIF1α and GBM progression in patients. These findings reveal a direct role of HIF in regulating miRNA biogenesis and consequently activating the miR-215-KDM1B-mediated signaling required for GIC adaptation to hypoxia.Item Open Access Synthetic lethality between HER2 and transaldolase in intrinsically resistant HER2-positive breast cancers.(Nature communications, 2018-10) Ding, Yi; Gong, Chang; Huang, De; Chen, Rui; Sui, Pinpin; Lin, Kevin H; Liang, Gehao; Yuan, Lifeng; Xiang, Handan; Chen, Junying; Yin, Tao; Alexander, Peter B; Wang, Qian-Fei; Song, Er-Wei; Li, Qi-Jing; Wood, Kris C; Wang, Xiao-FanIntrinsic resistance to anti-HER2 therapy in breast cancer remains an obstacle in the clinic, limiting its efficacy. However, the biological basis for intrinsic resistance is poorly understood. Here we performed a CRISPR/Cas9-mediated loss-of-function genetic profiling and identified TALDO1, which encodes the rate-limiting transaldolase (TA) enzyme in the non-oxidative pentose phosphate pathway, as essential for cellular survival following pharmacological HER2 blockade. Suppression of TA increases cell susceptibility to HER2 inhibition in two intrinsically resistant breast cancer cell lines with HER2 amplification. Mechanistically, TA depletion combined with HER2 inhibition significantly reduces cellular NADPH levels, resulting in excessive ROS production and deficient lipid and nucleotide synthesis. Importantly, higher TA expression correlates with poor response to HER2 inhibition in a breast cancer patient cohort. Together, these results pinpoint TA as a novel metabolic enzyme possessing synthetic lethality with HER2 inhibition that can potentially be exploited as a biomarker or target for combination therapy.Item Open Access The Epstein-Barr virus (EBV)-induced tumor suppressor microRNA MiR-34a is growth promoting in EBV-infected B cells.(Journal of virology, 2012-06) Forte, Eleonora; Salinas, Raul E; Chang, Christina; Zhou, Ting; Linnstaedt, Sarah D; Gottwein, Eva; Jacobs, Cassandra; Jima, Dereje; Li, Qi-Jing; Dave, Sandeep S; Luftig, Micah AEpstein-Barr virus (EBV) infection of primary human B cells drives their indefinite proliferation into lymphoblastoid cell lines (LCLs). B cell immortalization depends on expression of viral latency genes, as well as the regulation of host genes. Given the important role of microRNAs (miRNAs) in regulating fundamental cellular processes, in this study, we assayed changes in host miRNA expression during primary B cell infection by EBV. We observed and validated dynamic changes in several miRNAs from early proliferation through immortalization; oncogenic miRNAs were induced, and tumor suppressor miRNAs were largely repressed. However, one miRNA described as a p53-targeted tumor suppressor, miR-34a, was strongly induced by EBV infection and expressed in many EBV and Kaposi's sarcoma-associated herpesvirus (KSHV)-infected lymphoma cell lines. EBV latent membrane protein 1 (LMP1) was sufficient to induce miR-34a requiring downstream NF-κB activation but independent of functional p53. Furthermore, overexpression of miR-34a was not toxic in several B lymphoma cell lines, and inhibition of miR-34a impaired the growth of EBV-transformed cells. This study identifies a progrowth role for a tumor-suppressive miRNA in oncogenic-virus-mediated transformation, highlighting the importance of studying miRNA function in different cellular contexts.Item Open Access The Interplay of Antigen Presenting Cells, Microbiota, and IL-17 Producing T cells in Intestinal Health and Colorectal Cancer(2022) Huang, Hsin-IThe intestine is the home for trillions of microbes, collectively called microbiota. The mutualism of commensal microbes benefits intestinal health. To establish mutualism, intestinal immunity must equip with mechanisms to tolerate innocent microbiota while responding to pathogens. Intestinal immunity is coordinated by specialized and complicated mononuclear phagocytes subsets. The constitution of complex mononuclear phagocytes are thought to be similar in both small intestine and colon, however, these two organs are distinct in anatomy, microbiota abundance and composition, and immunological requirement. However, whether those distinctions require organ-specific mononuclear phagocytes to exert organ-specific immunity is unknown. To address whether there are organ-specific demands, we implement a novel mononuclear phagocyte subsetting strategy in the murine intestine and found two novel colon-specific mononuclear phagocytes: a macrophage population and a Th17-inducing dendritic cell (DC) subset. Those colon-specific DCs and macrophages surprisingly both required the transcription factor IRF4 and co-expressed DC marker CD24 and macrophage marker CD14. Novel IRF4-dependent CD14+CD24+ macrophages were markedly different from conventional macrophages in the way that novel CD14+CD24+ macrophages did not express macrophage markers CX3CR1, CD64, and CD88, and surprisingly not expressed cytokine IL-10, which is known to expressed in intestinal macrophages. Furthermore, we found that novel colon-specific CD14+CD24+ mononuclear phagocytes promote Th17 immunity in the colon and showed definitive evidence of differential requirement of mononuclear phagocytes in small intestine and colon for Th17 immunity. Our research discovered the diversity of mononuclear phagocytes with organ specificity and the organ-specific requirements for Th17 immunity.
While we found the requirement of organ-specific mononuclear phagocytes for Th17 immunity, regulation of innate-like IL-17 producing gd T cells, constituting a large source of IL-17 in intestine, is not addressed. It has been widely known that effector functions of gd T cells were pre-committed in the thymus, however, emerging evidence shows that environmental cues further re-programed gd T cell effectors. In the intestine, the major environmental cues are derived from the microbiota. Nevertheless, the microbiota-dependent regulation is controversial that some reports hold that microbiota provide immune activation signals while other reports conclude the suppressive function of microbiota. It remains unknown whether the impact of microbiota on gd T cells is activated, suppressive, or binary and whether microbiota shape gd T cells immune dynamics in distinct physiological conditions. Here, we identify that microbiota concomitantly upregulate IL-17-production and expression of the inhibitory receptor programmed cell death protein (PD-1) in gd17 T cells. The requirement of microbiota to preferentially upregulate PD-1 is conserved in gd17 T cells across multiple mucosal tissues. We determined the microbiota-driven PD-1 inhibits IL-17 production by endogenous gd17 effectors. We further show that microbiota-driven PD-1 and IL-17 phenotype are dynamics, wherein both PD-1 and IL-17 are downregulated once microbiota is depleted and is concomitantly upregulated during gut inflammation. Additionally, we found subset-specific metabolic shift in gd17 T cells that elevated lipid uptake in gd17 cells in response to intestinal inflammation linking to enhanced IL-17 production.
The coordination between adaptive and innate immune responses is essential for eradicating cancer cells. Among immune cells, DCs are the most powerful cells connecting innate and adaptive immune responses. Especially, Batf3-dependent DCs subset are thought to be key to anti-tumor immunity. The anti-tumor functions of Batf3-dependent DCs have been well reported in skin cancer, however, whether these DCs have the similar anti-tumor functions in colorectal cancer is largely unknown. Here, we investigated the roles of Batf3-dependent DCs in a spontaneous colorectal cancer and surprisingly found that these DCs promoted colon tumorigenesis. We identified an unconventional function of Batf3-dependent DCs to promote tumor infiltrating gd17 T cells in colon tumor instead of expanding anti-tumor IFNg-producing T cells. Proinflammatory cytokine IL-17 is known to promote colon tumor and the pro-tumor role of gd17 T cells have been proposed in several cancers including colorectal cancer. Mechanistically, we determined that Batf3-dependent DCs promote recruitment of gd17 T cells in colon tumor but are not required for gd17 T cells survival or proliferation. As we found that microbiota drives IL-17 production in gd17 T cells, the production of tumor infiltrating endogenous effector gd17 T cells and colon tumor growth were microbiota-dependent. Our study revealed a requirement of Batf3-dependent DCs to regulate tumor infiltrating gd17 T cells and the effector function is modulated by microbiota in colon tumor.
Item Open Access UHRF1 is required for basal stem cell proliferation in response to airway injury.(Cell Discov, 2017) Xiang, Handan; Yuan, Lifeng; Gao, Xia; Alexander, Peter B; Lopez, Omar; Lau, Calvin; Ding, Yi; Chong, Mengyang; Sun, Tao; Chen, Rui; Liu, Si-Qi; Wu, Haiyang; Wan, Ying; Randell, Scott H; Li, Qi-Jing; Wang, Xiao-FanCellular senescence is a cell fate characterized by an irreversible cell cycle arrest, but the molecular mechanism underlying this senescence hallmark remains poorly understood. Through an unbiased search for novel senescence regulators in airway basal cells, we discovered that the epigenetic regulator ubiquitin-like with PHD and ring finger domain-containing protein 1 (UHRF1) is critical for regulating cell cycle progression. Upon injury, basal cells in the mouse airway rapidly induce the expression of UHRF1 in order to stimulate stem cell proliferation and tissue repair. Targeted depletion of Uhrf1 specifically in airway basal cells causes a profound defect in cell cycle progression. Consistently, cultured primary human basal cells lacking UHRF1 do not exhibit cell death or differentiation phenotypes but undergo a spontaneous program of senescence. Mechanistically, UHRF1 loss induces G1 cell cycle arrest by abrogating DNA replication factory formation as evidenced by loss of proliferating cell nuclear antigen (PCNA) puncta and an inability to enter the first cell cycle. This proliferation defect is partially mediated by the p15 pathway. Overall, our study provides the first evidence of an indispensable role of UHRF1 in somatic stem cells proliferation during the process of airway regeneration.Item Open Access Understanding the Role of Circulating and Tissue-Resident T cells in Cancer and Autoimmunity(2021) Christian, Laura SonT cells have long been regarded as essential contributors in cancer surveillance and autoimmune disease, due to their vast abilities to coordinate, execute and monitor the adaptive immune response. Although many studies have focused on the important contributions of circulating memory T cells in cancer, there is a large knowledge gap in the development and functions of the recently characterized tissue-resident memory T cell population in the tumor. In addition, while significant strides have been made in understanding the pathogenesis of certain types of cancer, other types are less well understood. In this dissertation, I seek to understand the roles that tissue-resident memory T cells play in cancer by using high-throughput technologies to understand the clonal relationship of T cells in different tissues and to elucidate their tumor protective mechanisms at the single cell level. In a less characterized model of glioma, I also seek to expand our knowledge of the role of circulating effector T cells and to apply these findings to cancer immunotherapy with a novel form of adoptive T cell transfer.Tissue resident memory T cells (TRMs) develop after initial virus encounter and rapidly clear the pathogen. In the tumor environment, the association between increased TRMs in the tumor and prolonged patient survival has been well documented; however, TRM development and direct functions in tumorigenesis remain elusive. To address this, we used a murine breast cancer model and tracked TRM development in the tumor and in the contralateral distant mammary mucosa tissue. Single-cell RNA-sequencing of both intratumor and distant mucosa TRMs revealed two phenotypically distinct populations of TRMs representing their active and quiescent state. We found that TRMs in different tissue compartments shared the same TCR clonotypes and transcriptomes with a subset of intratumoral effector/effector memory T cells (TEff/EMs), clarifying their developmental ontogeny. At the mechanistic level, we showed that tumor production of CXCL16 maintains CXCR6+ TEff/EMs in the tumor, while the CXCR6- population egressed the tumor to form distant TRMs. We found that releasing CXCR6 mediated retention in the primary tumor led to superior control against lung metastases, indicating a potential direct role of TRMs in protection against tumor metastasis. Furthering our understanding of the circulating T cell response in gliomagenesis, we focused on mutant IDH1 gliomas. Despite the presence of this mutation in most low grade gliomas, there are still many remaining questions about the immune landscape during tumor formation and a lack of therapeutic targets. Using novel compound genetic mouse models that recapitulate the hallmark features of mutant IDH1 glioma, we performed high throughput TCR repertoire sequencing in mutant IDH1 versus WT IDH1 gliomas. Surprisingly, the glioma genotypes showed little differences in T cell infiltration and expansion. Extensive in vitro profiling experiments confirmed this, showing that the metabolite produced by mutant IDH1 tumors, D-2HG, had little effect on the skewing of the immune response. We developed an engineered T Cell Receptor (TCR-T) adoptive cell transfer based approach to identify and clone mutant IDH1 expressing T cells. We hope this collection of T cells will be useful in the immunotherapy of gliomas. Finally, we synthesized and biologically evaluated novel immune-suppressive therapies for the treatment of autoimmune diseases. While treatment options for autoimmune diseases have increased over the past several years, they are limited in their clinical efficacy by high drug toxicity and lack of selectivity. Thus, we sought new immunomodulatory agents with lower toxicity. Our previous work identified subglutinol A as a natural agent that exerts immune-suppressive effects on activated T cells. To provide guiding principles in future therapeutic development and to specifically define the structural requirements of subglutinols that exert their immunomodulatory activity, we prepared and evaluated the immunomodulatory activities of several subglutinol analogs. Though in vitro studies of structure-activity relationship, we identified a new subglutinol analog with reduced structural complexity. It will serve as a potential lead compound for future autoimmune drug development. This new subglutinol A compound, when compared to its parent compound, effectively suppressed inflammatory IFN and IL-17A cytokine production and has reduced T cell toxicity. Using a mouse model of experimental autoimmune encephalitis (EAE), we found that in vivo treatment of this new analog did not ameliorate the symptoms of disease, although it significantly increased the percentage of inflammatory neutrophils. These results suggest that, while not effective in EAE, this compound has potential as a novel therapy in diseases where promoting an influx of inflammatory neutrophils is beneficial.