750 results
Search Results
2. Author Correction: Requirements for the differentiation of innate T-bethigh memory-phenotype CD4+ T lymphocytes under steady state.
- Author
-
Kawabe, Takeshi, Yi, Jaeu, Kawajiri, Akihisa, Hilligan, Kerry, Fang, Difeng, Ishii, Naoto, Yamane, Hidehiro, Zhu, Jinfang, Jankovic, Dragana, Kim, Kwang Soon, Trinchieri, Giorgio, and Sher, Alan
- Subjects
T cells ,TECHNICAL specifications - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
3. Publisher Correction: Metabolic characteristics of CD8+ T cell subsets in young and aged individuals are not predictive of functionality.
- Author
-
Quinn, Kylie M., Hussain, Tabinda, Kraus, Felix, Formosa, Luke E., Lam, Wai K., Dagley, Michael J., Saunders, Eleanor C., Assmus, Lisa M., Wynne-Jones, Erica, Loh, Liyen, van de Sandt, Carolien E., Cooper, Lucy, Good-Jacobson, Kim L., Kedzierska, Katherine, Mackay, Laura K., McConville, Malcolm J., Ramm, Georg, Ryan, Michael T., and La Gruta, Nicole L.
- Subjects
T cells ,OLDER people - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
4. Author Correction: Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma.
- Author
-
Lauss, Martin, Donia, Marco, Harbst, Katja, Andersen, Rikke, Mitra, Shamik, Rosengren, Frida, Salim, Maryem, Vallon-Christersson, Johan, Törngren, Therese, Kvist, Anders, Ringnér, Markus, Svane, Inge Marie, and Jönsson, Göran
- Subjects
CELLULAR therapy ,T cells ,MELANOMA - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
5. Author Correction: In vivo clonal expansion and phenotypes of hypocretin-specific CD4+ T cells in narcolepsy patients and controls.
- Author
-
Jiang, Wei, Birtley, James R., Hung, Shu-Chen, Wang, Weiqi, Chiou, Shin-Heng, Macaubas, Claudia, Kornum, Birgitte, Tian, Lu, Huang, Huang, Adler, Lital, Weaver, Grant, Lu, Liying, Ilstad-Minnihan, Alexandra, Somasundaram, Sriram, Ayyangar, Sashi, Davis, Mark M., Stern, Lawrence J., and Mellins, Elizabeth D.
- Subjects
T cells ,NARCOLEPSY ,OREXINS ,PHENOTYPES - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
6. Transfer of mitochondrial DNA into the nuclear genome during induced DNA breaks.
- Author
-
Wu, Jinchun, Liu, Yang, Ou, Liqiong, Gan, Tingting, Zhangding, Zhengrong, Yuan, Shaopeng, Liu, Xinyi, Liu, Mengzhu, Li, Jiasheng, Yin, Jianhang, Xin, Changchang, Tian, Ye, and Hu, Jiazhi
- Subjects
NUCLEAR DNA ,GENOME editing ,GENOMES ,T cells ,EXONUCLEASES ,MITOCHONDRIAL DNA - Abstract
Mitochondria serve as the cellular powerhouse, and their distinct DNA makes them a prospective target for gene editing to treat genetic disorders. However, the impact of genome editing on mitochondrial DNA (mtDNA) stability remains a mystery. Our study reveals previously unknown risks of genome editing that both nuclear and mitochondrial editing cause discernible transfer of mitochondrial DNA segments into the nuclear genome in various cell types including human cell lines, primary T cells, and mouse embryos. Furthermore, drug-induced mitochondrial stresses and mtDNA breaks exacerbate this transfer of mtDNA into the nuclear genome. Notably, we observe that mitochondrial editors, including mitoTALEN and recently developed base editor DdCBE, can also enhance crosstalk between mtDNA and the nuclear genome. Moreover, we provide a practical solution by co-expressing TREX1 or TREX2 exonucleases during DdCBE editing. These findings imply genome instability of mitochondria during induced DNA breaks and explain the origins of mitochondrial-nuclear DNA segments. Emerging genome editing tools have been reported to possess threats to genome stability, while their impact on mitochondrial DNA (mtDNA) remains unknown. Here, authors report that genome editing of both nuclear and mitochondrial genome triggers transfer of mtDNA fragments into the nuclear genome. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. HIF1α-regulated glycolysis promotes activation-induced cell death and IFN-γ induction in hypoxic T cells.
- Author
-
Shen, Hongxing, Ojo, Oluwagbemiga A., Ding, Haitao, Mullen, Logan J., Xing, Chuan, Hossain, M. Iqbal, Yassin, Abdelrahman, Shi, Vivian Y., Lewis, Zach, Podgorska, Ewa, Andrabi, Shaida A., Antoniewicz, Maciek R., Bonner, James A., and Shi, Lewis Zhichang
- Subjects
IMMUNE checkpoint proteins ,CELL death ,IMMUNE response ,TUMOR microenvironment ,ACETYLCOENZYME A ,T cells - Abstract
Hypoxia is a common feature in various pathophysiological contexts, including tumor microenvironment, and IFN-γ is instrumental for anti-tumor immunity. HIF1α has long been known as a primary regulator of cellular adaptive responses to hypoxia, but its role in IFN-γ induction in hypoxic T cells is unknown. Here, we show that the HIF1α-glycolysis axis controls IFN-γ induction in both human and mouse T cells, activated under hypoxia. Specific deletion of HIF1α in T cells (Hif1α
–/– ) and glycolytic inhibition suppresses IFN-γ induction. Conversely, HIF1α stabilization by hypoxia and VHL deletion in T cells (Vhl–/– ) increases IFN-γ production. Hypoxic Hif1α–/– T cells are less able to kill tumor cells in vitro, and tumor-bearing Hif1α–/– mice are not responsive to immune checkpoint blockade (ICB) therapy in vivo. Mechanistically, loss of HIF1α greatly diminishes glycolytic activity in hypoxic T cells, resulting in depleted intracellular acetyl-CoA and attenuated activation-induced cell death (AICD). Restoration of intracellular acetyl-CoA by acetate supplementation re-engages AICD, rescuing IFN-γ production in hypoxic Hif1α–/– T cells and re-sensitizing Hif1α–/– tumor-bearing mice to ICB. In summary, we identify HIF1α-regulated glycolysis as a key metabolic control of IFN-γ production in hypoxic T cells and ICB response. Anti-cancer immunity relies on the effector functions (e.g., IFNγ production) of T cells that reside in a hypoxic tumor microenvironment. Here, the authors show that HIF1α-controlled glycolysis is an important driver of IFNγ production in hypoxic T cells, governing anti-tumor immunity. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
8. N-acetyltransferase 10 is implicated in the pathogenesis of cycling T cell-mediated autoimmune and inflammatory disorders in mice.
- Author
-
Li, Wen-ping, Mao, Xin-tao, Xie, Jia-huan, Li, Jie-yu, Liu, Bao-qin, Wu, Le-xi, Yang, Bing, Li, Yi-yuan, and Jin, Jin
- Subjects
CELL metabolism ,ENZYME deficiency ,CELLULAR control mechanisms ,AUTOIMMUNE diseases ,CELL proliferation ,T cells - Abstract
T cell expansion has a crucial function in both autoimmune and chronic inflammatory diseases, with cycling T cells contributing to the pathogenesis of autoimmune diseases by causing uncontrolled immune responses and tissue damage. Yet the regulatory mechanisms governing T cell expansion remain incompletely understood. Here we show that the enzyme N-acetyltransferase 10 (NAT10) regulates T cell activation and proliferation upon antigen stimulation. T cell-specific NAT10 deficiency in mice reduces the number of mature T cells in peripheral lymphoid organs. Mechanistically, NAT10 acetylates RACK1 at K185, preventing subsequent RACK1 K48-linked ubiquitination and degradation. The increased RACK1 stability alters ribosome formation and cellular metabolism, leading to enhanced supply of energy and biosynthetic precursors and, eventually, T cell proliferation. Our findings thus highlight the essential function of NAT10 in T cell self-renewal and metabolism and elucidate NAT10 mode of action for the potential development of novel therapies for immune-related disorders. Abnormal T cell proliferation often triggers autoimmune disorders. Here the authors show that T cell-specific deficiency of the enzyme N-acetyltransferase 10 ameliorates experimental autoimmune encephalitis potentially by RACK1-mediated regulation of T cell metabolism and expansion. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Physiological and pathogenic T cell autoreactivity converge in type 1 diabetes.
- Author
-
Eugster, Anne, Lorenc, Anna, Kotrulev, Martin, Kamra, Yogesh, Goel, Manisha, Steinberg-Bains, Katja, Sabbah, Shereen, Dietz, Sevina, Bonifacio, Ezio, Peakman, Mark, and Gomez-Tourino, Iria
- Subjects
GLUTAMATE decarboxylase ,TYPE 1 diabetes ,AUTOIMMUNITY ,AUTOIMMUNE diseases ,GENE expression profiling ,T cells - Abstract
Autoimmune diseases result from autoantigen-mediated activation of adaptive immunity; intriguingly, autoantigen-specific T cells are also present in healthy donors. An assessment of dynamic changes of this autoreactive repertoire in both health and disease is thus warranted. Here we investigate the physiological versus pathogenic autoreactive processes in the context of Type 1 diabetes (T1D) and one of its landmark autoantigens, glutamic acid decarboxylase 65 (GAD65). Using single cell gene expression profiling and tandem T cell receptor (TCR) sequencing, we find that GAD65-specific true naïve cells are present in both health and disease, with GAD65-specific effector and memory responses showing similar ratios in healthy donors and patients. Deeper assessment of phenotype and TCR repertoire uncover differential features in GAD65-specific TCRs, including lower clonal sizes of healthy donor-derived clonotypes in patients. We thus propose a model whereby physiological autoimmunity against GAD65 is needed during early life, and that alterations of these physiological autoimmune processes in predisposed individuals trigger overt Type 1 diabetes. Autoreactive T cells contribute to the onset of autoimmune diseases but are also detectable in healthy individuals. Here, by examining the dynamics of autoreactive T cell responses to a diabetogenic antigen, the authors show that specific phenotype and T cell receptor repertoire changes are associated with health or disease, with additional cues implicated for this transition. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Protective effect of TCR-mediated MAIT cell activation during experimental autoimmune encephalomyelitis.
- Author
-
Walkenhorst, Mark, Sonner, Jana K., Meurs, Nina, Engler, Jan Broder, Bauer, Simone, Winschel, Ingo, Woo, Marcel S., Raich, Lukas, Winkler, Iris, Vieira, Vanessa, Unger, Lisa, Salinas, Gabriela, Lantz, Olivier, Friese, Manuel A., and Willing, Anne
- Subjects
BACTERIAL antigens ,CENTRAL nervous system ,MULTIPLE sclerosis ,ENCEPHALOMYELITIS ,AMPHIREGULIN ,T cells - Abstract
Mucosal-associated invariant T (MAIT) cells express semi-invariant T cell receptors (TCR) for recognizing bacterial and yeast antigens derived from riboflavin metabolites presented on the non-polymorphic MHC class I-related protein 1 (MR1). Neuroinflammation in multiple sclerosis (MS) is likely initiated by autoreactive T cells and perpetuated by infiltration of additional immune cells, but the precise role of MAIT cells in MS pathogenesis remains unknown. Here, we use experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, and find an accumulation of MAIT cells in the inflamed central nervous system (CNS) enriched for MAIT17 (RORγt
+ ) and MAIT1/17 (T-bet+ RORγt+ ) subsets with inflammatory and protective features. Results from transcriptome profiling and Nur77GFP reporter mice show that these CNS MAIT cells are activated via cytokines and TCR. Blocking TCR activation with an anti-MR1 antibody exacerbates EAE, whereas enhancing TCR activation with the cognate antigen, 5-(2-oxopropylideneamino)−6-D-ribitylaminouracil, ameliorates EAE severity, potentially via the induction of amphiregulin (AREG). In summary, our findings suggest that TCR-mediated MAIT cell activation is protective in CNS inflammation, likely involving an induction of AREG. Mucosal-associated invariant T (MAIT) cells mediate protection from pathogens, but their role in autoimmunity is unclear. Here the authors show that, in a mouse experimental autoimmune encephalomyelitis model, MAIT cells accumulate in the inflamed central nervous system and serve protective functions when activated via their T cell receptor. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
11. A splicing isoform of PD-1 promotes tumor progression as a potential immune checkpoint.
- Author
-
Wang, Xuetong, Liu, Tongfeng, Li, Yifei, Ding, Ao, Zhang, Chang, Gu, Yinmin, Zhao, Xujie, Cheng, Shuwen, Cheng, Tianyou, Wu, Songzhe, Duan, Liqiang, Zhang, Jihang, Yin, Rong, Shang, Man, and Gao, Shan
- Subjects
RNA-binding proteins ,ALTERNATIVE RNA splicing ,IMMUNE checkpoint proteins ,TUMOR-infiltrating immune cells ,T cells - Abstract
The immune checkpoint receptor, programmed cell death 1 (PD-1, encoded by PDCD1), mediates the immune escape of cancer, but whether PD-1 splicing isoforms contribute to this process is still unclear. Here, we identify an alternative splicing isoform of human PD-1, which carries a 28-base pairs extension retained from 5′ region of intron 2 (PD-1^28), is expressed in peripheral T cells and tumor infiltrating lymphocytes. PD-1^28 expression is induced on T cells upon activation and is regulated by an RNA binding protein, TAF15. Functionally, PD-1^28 inhibits T cell proliferation, cytokine production, and tumor cell killing in vitro. In vivo, T cell-specific exogenous expression of PD-1^28 promotes tumor growth in both a syngeneic mouse tumor model and humanized NOG mice inoculated with human lung cancer cells. Our study thus demonstrates that PD-1^28 functions as an immune checkpoint, and may contribute to resistance to immune checkpoint blockade therapy. Whether PD-1 splicing isoforms impact T cell anti-tumor capacity has not been fully illustrated. Here the authors identify a human PD-1 isoform, PD-1^28, which functions to suppress anti-cancer immunity in vitro and in both syngeneic and humanized mouse tumor models. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Dominant immune tolerance in the intestinal tract imposed by RelB-dependent migratory dendritic cells regulates protective type 2 immunity.
- Author
-
Geiselhöringer, Anna-Lena, Kolland, Daphne, Patt, Arisha Johanna, Hammann, Linda, Köhler, Amelie, Kreft, Luisa, Wichmann, Nina, Hils, Miriam, Ruedl, Christiane, Riemann, Marc, Biedermann, Tilo, Anz, David, Diefenbach, Andreas, Voehringer, David, Schmidt-Weber, Carsten B., Straub, Tobias, Pasztoi, Maria, and Ohnmacht, Caspar
- Subjects
REGULATORY T cells ,TRANSCRIPTION factors ,DENDRITIC cells ,INTESTINAL infections ,IMMUNOLOGICAL tolerance ,T cells ,HOMEOSTASIS - Abstract
Dendritic cells (DCs) are crucial for initiating protective immune responses and have also been implicated in the generation and regulation of Foxp3
+ regulatory T cells (Treg cells). Here, we show that in the lamina propria of the small intestine, the alternative NF-κB family member RelB is necessary for the differentiation of cryptopatch and isolated lymphoid follicle-associated DCs (CIA-DCs). Moreover, single-cell RNA sequencing reveals a RelB-dependent signature in migratory DCs in mesenteric lymph nodes favoring DC-Treg cell interaction including elevated expression and release of the chemokine CCL22 from RelB-deficient conventional DCs (cDCs). In line with the key role of CCL22 to facilitate DC-Treg cell interaction, RelB-deficient DCs have a selective advantage to interact with Treg cells in an antigen-specific manner. In addition, DC-specific RelB knockout animals show increased total Foxp3+ Treg cell numbers irrespective of inflammatory status. Consequently, DC-specific RelB knockout animals fail to mount protective Th2-dominated immune responses in the intestine after infection with Heligmosomoides polygyrus bakeri. Thus, RelB expression in cDCs acts as a rheostat to establish a tolerogenic set point that is maintained even during strong type 2 immune conditions and thereby is a key regulator of intestinal homeostasis. Dendritic cells play intricate roles in engaging a range of immune cells. Here, the authors establish a role for the transcription factor RelB in dendritic cells as a molecular rheostat that controls the level of immune tolerance by limiting the number of regulatory T cells. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
13. Spatial dynamics of CD39+CD8+ exhausted T cell reveal tertiary lymphoid structures-mediated response to PD-1 blockade in esophageal cancer.
- Author
-
Tanoue, Kenro, Ohmura, Hirofumi, Uehara, Koki, Ito, Mamoru, Yamaguchi, Kyoko, Tsuchihashi, Kenji, Shinohara, Yudai, Lu, Peng, Tamura, Shingo, Shimokawa, Hozumi, Isobe, Taichi, Ariyama, Hiroshi, Shibata, Yoshihiro, Tanaka, Risa, Kusaba, Hitoshi, Esaki, Taito, Mitsugi, Kenji, Kiyozawa, Daisuke, Iwasaki, Takeshi, and Yamamoto, Hidetaka
- Subjects
SQUAMOUS cell carcinoma ,T cells ,CELL populations ,IMMUNE checkpoint proteins ,LYMPHOID tissue - Abstract
Despite the success of immune checkpoint blockade (ICB) therapy for esophageal squamous cell cancer, the key immune cell populations that affect ICB efficacy remain unclear. Here, imaging mass cytometry of tumor tissues from ICB-treated patients identifies a distinct cell population of CD39
+ PD-1+ CD8+ T cells, specifically the TCF1+ subset, precursor exhausted T (CD39+ Tpex) cells, which positively correlate with ICB benefit. CD39+ Tpex cells are predominantly in the stroma, while differentiated CD39+ exhausted T cells are abundantly and proximally within the parenchyma. Notably, CD39+ Tpex cells are concentrated within and around tertiary lymphoid structure (TLS). Accordingly, tumors harboring TLSs have more of these cells in tumor areas than tumors lacking TLSs, suggesting Tpex cell recruitment from TLSs to tumors. In addition, circulating CD39+ Tpex cells are also increased in responders following ICB therapy. Our findings show that this unique subpopulation of CD39+ PD-1+ CD8+ T cells is crucial for ICB benefit, and suggest a key role in TLS-mediated immune responses against tumors. Immune checkpoint blockade (ICB) benefits esophageal squamous cell cancer, but the immune cell mediators remain unclear. Here the author show, by imaging mass cytometry, that CD39+ CD8+ exhausted cells are present abundantly in both tertiary lymphoid tissue and tumor and correlate with responses to ICB. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
14. Pre-operative stereotactic radiosurgery and peri-operative dexamethasone for resectable brain metastases: a two-arm pilot study evaluating clinical outcomes and immunological correlates.
- Author
-
Jansen, Caroline S., Pagadala, Meghana S., Cardenas, Maria A., Prabhu, Roshan S., Goyal, Subir, Zhou, Chengjing, Chappa, Prasanthi, Vo, BaoHan T., Ye, Chengyu, Hopkins, Benjamin, Zhong, Jim, Klie, Adam, Daniels, Taylor, Admassu, Maedot, Green, India, Pfister, Neil T., Neill, Stewart G., Switchenko, Jeffrey M., Prokhnevska, Nataliya, and Hoang, Kimberly B.
- Subjects
ANTIGEN presenting cells ,STEREOTACTIC radiosurgery ,BRAIN metastasis ,TREATMENT effectiveness ,DISEASE relapse ,T cells - Abstract
Enhancing the efficacy of immunotherapy in brain metastases (BrM) requires an improved understanding of the immune composition of BrM and how this is affected by radiation and dexamethasone. Our two-arm pilot study (NCT04895592) allocated 26 patients with BrM to either low (Arm A) or high (Arm B) dose peri-operative dexamethasone followed by pre-operative stereotactic radiosurgery (pSRS) and resection (n= 13 per arm). The primary endpoint, a safety analysis at 4 months, was met. The secondary clinical endpoints of overall survival, distant brain failure, leptomeningeal disease and local recurrence at 12-months were 66%, 37.3%, 6%, and 0% respectively and were not significantly different between arms (p= 0.7739, p= 0.3884, p= 0.3469). Immunological data from two large retrospective BrM datasets and confirmed by correlates from both arms of this pSRS prospective trial revealed that BrM CD8 T cells were composed of predominantly PD1+ TCF1+ stem-like and PD1+ TCF1-TIM3+ effector-like cells. Clustering of TCF1+ CD8 T cells with antigen presenting cells in immune niches was prognostic for local control, even without pSRS. Following pSRS, CD8 T cell and immune niche density were transiently reduced compared to untreated BrM, followed by a rebound 6+ days post pSRS with an increased frequency of TCF1- effector-like cells. In sum, pSRS is safe and therapeutically beneficial, and these data provide a framework for how pSRS may be leveraged to maximize intracranial CD8 T cell responses. Radiation and steroid dosing can affect the immune composition of brain metastasis (BM). The authors have designed a pilot study of pre-operative stereotactic radiosurgery with low or high dose of peri-operative dexamethasone for resectable brain metastases, here reporting clinical outcomes and characterization of intratumor TCF1+ CD8+ stem-like T cell immune niches in the brain. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. TGF-β-mediated crosstalk between TIGIT+ Tregs and CD226+CD8+ T cells in the progression and remission of type 1 diabetes.
- Author
-
Zhong, Ting, Li, Xinyu, Lei, Kang, Tang, Rong, Deng, Qiaolin, Love, Paul E, Zhou, Zhiguang, Zhao, Bin, and Li, Xia
- Subjects
CYTOTOXIC T cells ,TYPE 1 diabetes ,REGULATORY T cells ,CELL populations ,CELL analysis ,T cells - Abstract
Type 1 diabetes (T1D) is a chronic autoimmune condition characterized by hyperglycemia resulting from the destruction of insulin-producing β-cells that is traditionally deemed irreversible, but partial remission (PR) with temporary reversal of hyperglycemia is sometimes observed. Here we use single-cell RNA sequencing to delineate the immune cell landscape across patients in different T1D stages. Together with cohort validation and functional assays, we observe dynamic changes in TIGIT
+ CCR7− Tregs and CD226+ CCR7− CD8+ cytotoxic T cells during the peri-remission phase. Machine learning algorithms further identify TIGIT+ CCR7− Tregs and CD226+ CD8+ T cells as biomarkers for β-cell function decline in a predictive model, while cell communication analysis and in vitro assays suggest that TIGIT+ CCR7− Tregs may inhibit CD226+ CCR7− CD8+ T cells via TGF-β signaling. Lastly, in both cyclophosphamide-induced and streptozotocin (STZ)-induced mouse diabetes models, CD226 inhibition postpones insulitis onset and reduces hyperglycemia severity. Our results thus identify two interrelated immune cell subsets that may serve as biomarkers for monitoring disease progression and targets for therapeutic intervention of T1D. Type 1 diabetes (T1D) manifests as hyperglycemia, with spontaneous remission occasionally observed, but the underneath mechanisms are unclear. Here the authors analyses patients at different stages to find two populations of immune cells correlating with disease progression or remission, while results from mouse diabetes models validate these observations. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
16. Direct recognition of an intact foreign protein by an αβ T cell receptor.
- Author
-
Almeida, Catarina F., Gully, Benjamin S., Jones, Claerwen M., Kedzierski, Lukasz, Gunasinghe, Sachith D., Rice, Michael T., Berry, Richard, Gherardin, Nicholas A., Nguyen, Trang T., Mok, Yee-Foong, Reijneveld, Josephine F., Moody, D. Branch, Van Rhijn, Ildiko, La Gruta, Nicole L., Uldrich, Adam P., Rossjohn, Jamie, and Godfrey, Dale I.
- Subjects
MAJOR histocompatibility complex ,T cells ,MOLECULAR interactions ,CRYSTAL structure ,T cell receptors ,GLOBIN ,THYMUS - Abstract
αβ T cell receptors (αβTCRs) co-recognise antigens when bound to Major Histocompatibility Complex (MHC) or MHC class I-like molecules. Additionally, some αβTCRs can bind non-MHC molecules, but how much intact antigen reactivities are achieved remains unknown. Here, we identify an αβ T cell clone that directly recognises the intact foreign protein, R-phycoerythrin (PE), a multimeric (αβ)
6 γ protein complex. This direct αβTCR–PE interaction occurs in an MHC-independent manner, yet triggers T cell activation and bound PE with an affinity comparable to αβTCR–peptide–MHC interactions. The crystal structure reveals how six αβTCR molecules simultaneously engage the PE hexamer, mediated by the complementarity-determining regions (CDRs) of the αβTCR. Here, the αβTCR mainly binds to two α-helices of the globin fold in the PE α-subunit, which is analogous to the antigen-binding platform of the MHC molecule. Using retrogenic mice expressing this TCR, we show that it supports intrathymic T cell development, maturation, and exit into the periphery as mature CD4/CD8 double negative (DN) T cells with TCR-mediated functional capacity. Accordingly, we show how an αβTCR can recognise an intact foreign protein in an antibody-like manner. Certain specific antigens have been shown to activate T cells in an MHC independent manner. Here the authors show a phycoerythrin reactive mouse TCR which recognises native protein and characterise the molecular nature of this interaction and that this specific TCR can be selected in the thymus. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
17. 68Ga-grazytracer PET for noninvasive assessment of response to immunotherapy in solid tumors and lymphomas: a phase 1/2 clinical trial.
- Author
-
Shen, Xiuling, Zhou, Haoyi, Zhou, Xin, Liu, Zongchao, Meng, Xiangxi, Zhang, Linyu, Song, Yufei, Guo, Rui, Wang, Fei, Li, Kui, Li, Wenqing, Yang, Zhi, Liu, Zhaofei, and Li, Nan
- Subjects
TREATMENT effectiveness ,POSITRON emission tomography ,IMMUNE checkpoint inhibitors ,IMMUNE response ,T cells - Abstract
To tackle the clinical challenge of noninvasively assessing immunotherapy efficacy in patients, here we used positron emission tomography (PET) with
68 Ga-grazytracer, which targets granzyme B, a crucial effector molecule secreted by activated CD8+ T cells. In this phase 1/2 clinical trial (NCT05000372) involving a diverse cohort of 24 patients with solid tumors and lymphomas who received immunotherapies, including immune checkpoint inhibitors (either alone or with chemotherapies) and chimeric antigen receptor-T cell therapy, we examined the in vivo behaviors of68 Ga-grazytracer. Primary endpoints were safety, biodistribution, granzyme B specificity, and the predictive utility of68 Ga-grazytracer, while secondary endpoint was the relationship between68 Ga-grazytracer uptake and tumor immune phenotype.68 Ga-grazytracer exhibited a safe profile and specifically targeted granzyme B in patients.68 Ga-grazytracer PET showed superior predictive value for short-term prognosis and progression-free survival than those of conventional assessment criteria, including RECIST 1.1 and PERCIST. Moreover, the uptake of68 Ga-grazytracer in tumors was significantly higher in those with a "non-desert" immune phenotype than those with an immune "desert" phenotype, thereby meeting the primary and secondary endpoints of this trial. Collectively, we successfully visualized CD8+ T cell effector function in humans using68 Ga-grazytracer PET, offering insights for enhancing immunotherapy assessment, patient stratification and treatment planning. Several approaches based on positron emission tomography (PET) are currently used to evaluate cancer response to immunotherapy. Here the authors report the results of a phase 1/2 trial evaluating a PET radiotracer,68 Ga-grazytracer, for the imaging of granzyme B expression in patients treated with immune checkpoint inhibitors or CAR-T cell therapy. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
18. GLUT1 overexpression in CAR-T cells induces metabolic reprogramming and enhances potency.
- Author
-
Guerrero, Justin A., Klysz, Dorota D., Chen, Yiyun, Malipatlolla, Meena, Lone, Jameel, Fowler, Carley, Stuani, Lucille, May, Audre, Bashti, Malek, Xu, Peng, Huang, Jing, Michael, Basil, Contrepois, Kévin, Dhingra, Shaurya, Fisher, Chris, Svensson, Katrin J., Davis, Kara L., Kasowski, Maya, Feldman, Steven A., and Sotillo, Elena
- Subjects
T-cell exhaustion ,METABOLIC reprogramming ,GLUCOSE transporters ,CELL physiology ,REACTIVE oxygen species ,T cells ,GLYCOLYSIS - Abstract
The intensive nutrient requirements needed to sustain T cell activation and proliferation, combined with competition for nutrients within the tumor microenvironment, raise the prospect that glucose availability may limit CAR-T cell function. Here, we seek to test the hypothesis that stable overexpression (OE) of the glucose transporter GLUT1 in primary human CAR-T cells would improve their function and antitumor potency. We observe that GLUT1OE in CAR-T cells increases glucose consumption, glycolysis, glycolytic reserve, and oxidative phosphorylation, and these effects are associated with decreased T cell exhaustion and increased Th
17 differentiation. GLUT1OE also induces broad metabolic reprogramming associated with increased glutathione-mediated resistance to reactive oxygen species, and increased inosine accumulation. When challenged with tumors, GLUT1OE CAR-T cells secrete more proinflammatory cytokines and show enhanced cytotoxicity in vitro, and demonstrate superior tumor control and persistence in mouse models. Our collective findings support a paradigm wherein glucose availability is rate limiting for effector CAR-T cell function and demonstrate that enhancing glucose availability via GLUT1OE could augment antitumor immune function. T cell activation is a process that requires extra nutrients, which could be difficult to source from the tumor microenvironment in competition with tumor cells. Here authors increase the metabolic fitness of CAR-T cells by stable overexpression of the glucose transporter GLUT1, which allows them to increase their glucose intake and enhances their antitumour function. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
19. Human papillomavirus-encoded circular RNA circE7 promotes immune evasion in head and neck squamous cell carcinoma.
- Author
-
Ge, Junshang, Meng, Yi, Guo, Jiayue, Chen, Pan, Wang, Jie, Shi, Lei, Wang, Dan, Qu, Hongke, Wu, Pan, Fan, Chunmei, Zhang, Shanshan, Liao, Qianjin, Zhou, Ming, Xiang, Bo, Wang, Fuyan, Tan, Ming, Gong, Zhaojian, Xiong, Wei, and Zeng, Zhaoyang
- Subjects
CYTOTOXIC T cells ,IMMUNE checkpoint proteins ,CIRCULAR RNA ,HUMAN papillomavirus ,SQUAMOUS cell carcinoma ,T cells - Abstract
Immune evasion represents a crucial milestone in the progression of cancer and serves as the theoretical foundation for tumor immunotherapy. In this study, we reveal a negative association between Human Papillomavirus (HPV)-encoded circular RNA, circE7, and the infiltration of CD8
+ T cells in head and neck squamous cell carcinoma (HNSCC). Both in vitro and in vivo experiments demonstrate that circE7 suppresses the function and activity of T cells by downregulating the transcription of LGALS9, which encodes the galectin-9 protein. The molecular mechanism involves circE7 binding to acetyl-CoA carboxylase 1 (ACC1), promoting its dephosphorylation and thereby activating ACC1. Activated ACC1 reduces H3K27 acetylation at the LGALS9 gene promoter, leading to decreased galectin-9 expression. Notably, galectin-9 interacts with immune checkpoint molecules TIM-3 and PD-1, inhibiting the secretion of cytotoxic cytokines by T cells and promoting T cell apoptosis. Here, we demonstrate a mechanism by which HPV promotes immune evasion in HNSCC through a circE7-driven epigenetic modification and propose a potential immunotherapy strategy for HNSCC that involves the combined use of anti-PD-1 and anti-TIM-3 inhibitors. Immune evasion is a feature of HPV-positive head and neck squamous cell carcinoma (HNSCC). Here the authors report that a circular RNA, circE7, encoded by HPV promotes immune evasion in HNSCC by promoting downregulation of galectin-9. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
20. Aberrant cytoplasmic expression of UHRF1 restrains the MHC-I-mediated anti-tumor immune response.
- Author
-
Tan, Lianmei, Yin, Tao, Xiang, Handan, Wang, Liuyang, Mudgal, Poorva, Chen, Junying, Ding, Yi, Wang, Guoping, Lim, Bryan Jian Wei, Huang, Yuqi, Huang, De, Liang, Yaosi, Alexander, Peter B., Xiang, Kun, Wang, Ergang, Yan, Chengsong, Ma, Zhehao, Tan, Minjia, Li, Qi-Jing, and Wang, Xiao-Fan
- Subjects
UBIQUITIN ligases ,ANTIGEN presentation ,IMMUNE checkpoint proteins ,IMMUNE response ,TUMOR microenvironment ,UBIQUITINATION ,T cells - Abstract
Immunotherapy successfully complements traditional cancer treatment. However, primary and acquired resistance might limit efficacy. Reduced antigen presentation by MHC-I has been identified as potential resistance factor. Here we show that the epigenetic regulator ubiquitin-like with PHD and ring finger domains 1 (UHRF1), exhibits altered expression and aberrant cytosolic localization in cancerous tissues, where it promotes MHC-I ubiquitination and degradation. Cytoplasmic translocation of UHRF1 is induced by its phosphorylation on a specific serine in response to signals provided by factors present in the tumor microenvironment (TME), such as TGF-β, enabling UHRF1 to bind MHC-I. Downregulation of MHC-I results in suppression of the antigen presentation pathway to establish an immune hostile TME. UHRF1 inactivation by genetic deletion synergizes with immune checkpoint blockade (ICB) treatment and induces an anti-tumour memory response by evoking low-affinity T cells. Our study adds to the understanding of UHRF1 in cancer immune evasion and provides a potential target to synergize with immunotherapy and overcome immunotherapeutic resistance. MHC-I mediated antigen presentation is an important element of the anti-tumour immune response. Here authors identify a tumour immune escape mechanism by which the cancer cells express the ubiquitin E3 ligase UHRF1 in the cytoplasm instead of the nuclear expression pattern observed in normal tissues, and this results in degradation of MHC-I and thus diminished antigen presentation and anti-tumour T cell response. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Quantifiable blood TCR repertoire components associate with immune aging.
- Author
-
Hu, Jing, Pan, Mingyao, Reid, Brett, Tworoger, Shelley, and Li, Bo
- Subjects
T-cell exhaustion ,OLDER people ,BONE marrow transplantation ,CELL populations ,T cells ,BONE marrow - Abstract
T cell senescence alters the homeostasis of distinct T cell populations and results in decayed adaptive immune protection in older individuals, but a link between aging and dynamic T cell clone changes has not been made. Here, using a newly developed computational framework, Repertoire Functional Units (RFU), we investigate over 6500 publicly available TCR repertoire sequencing samples from multiple human cohorts and identify age-associated RFUs consistently across different cohorts. Quantification of RFU reduction with aging reveals accelerated loss under immunosuppressive conditions. Systematic analysis of age-associated RFUs in clinical samples manifests a potential link between these RFUs and improved clinical outcomes, such as lower ICU admission and reduced risk of complications, during acute viral infections. Finally, patients receiving bone marrow transplantation show a secondary expansion of the age-associated clones upon stem cell transfer from younger donors. Together, our results suggest the existence of a 'TCR clock' that could reflect the immune functions in aging populations. Immune aging is associated with altered homeostasis of distinct T cell populations, but a link to clonal dynamic is still not made. Here the authors develop a new framework, Repertoire Functional Units (RFU), and use public TCR sequences to find specific TCR clonal changes that correlate with aged immune repertoires or outcomes of acute viral infection. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Immune profiling-based targeting of pathogenic T cells with ustekinumab in ANCA-associated glomerulonephritis.
- Author
-
Engesser, Jonas, Khatri, Robin, Schaub, Darius P., Zhao, Yu, Paust, Hans-Joachim, Sultana, Zeba, Asada, Nariaki, Riedel, Jan-Hendrik, Sivayoganathan, Varshi, Peters, Anett, Kaffke, Anna, Jauch-Speer, Saskia-Larissa, Goldbeck-Strieder, Thiago, Puelles, Victor G., Wenzel, Ulrich O., Steinmetz, Oliver M., Hoxha, Elion, Turner, Jan-Eric, Mittrücker, Hans-Willi, and Wiech, Thorsten
- Subjects
ANTINEUTROPHIL cytoplasmic antibodies ,KIDNEY failure ,KIDNEY physiology ,DISEASE relapse ,IMMUNOSUPPRESSIVE agents ,T cells - Abstract
Antineutrophil cytoplasmic antibody (ANCA)–associated vasculitis is a life-threatening autoimmune disease that often results in kidney failure caused by crescentic glomerulonephritis (GN). To date, treatment of most patients with ANCA-GN relies on non-specific immunosuppressive agents, which may have serious adverse effects and be only partially effective. Here, using spatial and single-cell transcriptome analysis, we characterize inflammatory niches in kidney samples from 34 patients with ANCA-GN and identify proinflammatory, cytokine-producing CD4
+ and CD8+ T cells as a pathogenic signature. We then utilize these transcriptomic profiles for digital pharmacology and identify ustekinumab, a monoclonal antibody targeting IL-12 and IL-23, as the strongest therapeutic drug to use. Moreover, four patients with relapsing ANCA-GN are treated with ustekinumab in combination with low-dose cyclophosphamide and steroids, with ustekinumab given subcutaneously (90 mg) at weeks 0, 4, 12, and 24. Patients are followed up for 26 weeks to find this treatment well-tolerated and inducing clinical responses, including improved kidney function and Birmingham Vasculitis Activity Score, in all ANCA-GN patients. Our findings thus suggest that targeting of pathogenic T cells in ANCA-GN patients with ustekinumab might represent a potential approach and warrants further investigation in clinical trials. Antineutrophil cytoplasmic antibody (ANCA) is currently treated with broad-spectrum immune suppressive drugs. Here the authors decipher inflammatory niches in the kidney of patients with ANCA-GN by combining spatial and single-cell transcriptomics to identify ustekinumab as a promising treatment option and successfully treat four ANCA-GN patients. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
23. Tuning the fluidity and protein corona of ultrasound-responsive liposomal nanovaccines to program T cell immunity in mice.
- Author
-
He, Jia, Wang, Chaoyu, Fang, Xiao, Li, Junyao, Shen, Xueying, Zhang, Junxia, Peng, Cheng, Li, Hongjian, Li, Sai, Karp, Jeffrey M., and Kuai, Rui
- Subjects
CANCER vaccines ,INTRAVENOUS injections ,CERVICAL cancer ,IMMUNE response ,SPLEEN ,T cells - Abstract
Inducing high levels of antigen-specific CD8α
+ T cells in the tumor is beneficial for cancer immunotherapy, but achieving this in a safe and effective manner remains challenging. Here, we have developed a designer liposomal nanovaccine containing a sonosensitizer (LNVS) to efficiently program T cell immunity in mice. Following intravenous injection, LNVS accumulates in the spleen in a protein corona and fluidity-dependent manner, leading to greater frequencies of antigen-specific CD8α+ T cells than soluble vaccines (the mixture of antigens and adjuvants). Meanwhile, some LNVS passively accumulates in the tumor, where it responds to ultrasound (US) to increase the levels of chemokines and adhesion molecules that are beneficial for recruiting CD8α+ T cells to the tumor. LNVS + US induces higher levels of intratumoral antitumor T cells than traditional sonodynamic therapy, regresses established mouse MC38 tumors and orthotopic cervical cancer, and protects cured mice from relapse. Our platform sheds light on the importance of tuning the fluidity and protein corona of naovaccines to program T cell immunity in mice and may inspire new strategies for cancer immunotherapy. Delivery and fluidity of cancer nanoparticle vaccines alters efficiency and immune priming. Here the authors show a sonosensitive nanovaccine which depending on fluidity of the vaccine improves vaccine targeting and subsequent anti-tumour immune responses. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
24. HIF-2α-dependent induction of miR-29a restrains TH1 activity during T cell dependent colitis.
- Author
-
Czopik, Agnieszka K., McNamee, Eóin N., Vaughn, Victoria, Huang, Xiangsheng, Bang, In Hyuk, Clark, Trent, Wang, Yanyu, Ruan, Wei, Nguyen, Tom, Masterson, Joanne C., Tak, Eunyoung, Frank, Sandra, Collins, Colm B., Li, Howard, Rodriguez-Aguayo, Cristian, Lopez-Berestein, Gabriel, Gerich, Mark E., Furuta, Glenn T., Yuan, Xiaoyi, and Sood, Anil K.
- Subjects
INFLAMMATORY bowel diseases ,HYPOXIA-inducible factors ,CELLULAR control mechanisms ,CELL physiology ,TRANSCRIPTION factors ,T cells - Abstract
Metabolic imbalance leading to inflammatory hypoxia and stabilization of hypoxia-inducible transcription factors (HIFs) is a hallmark of inflammatory bowel diseases. We hypothesize that HIF could be stabilized in CD4
+ T cells during intestinal inflammation and alter the functional responses of T cells via regulation of microRNAs. Our assays reveal markedly increased T cell-intrinsic hypoxia and stabilization of HIF protein during experimental colitis. microRNA screen in primary CD4+ T cells points us towards miR-29a and our subsequent studies identify a selective role for HIF-2α in CD4-cell-intrinsic induction of miR-29a during hypoxia. Mice with T cell-intrinsic HIF-2α deletion display elevated T-bet (target of miR-29a) levels and exacerbated intestinal inflammation. Mice with miR-29a deficiency in T cells show enhanced intestinal inflammation. T cell-intrinsic overexpression of HIF-2α or delivery of miR-29a mimetic dampen TH 1-driven colitis. In this work, we show a previously unrecognized function for hypoxia-dependent induction of miR-29a in attenuating TH 1-mediated inflammation. Inflammatory intestinal lesions are often hypoxic, which results in the stabilization and activation of hypoxia-inducible-factors (HIF). Here authors show that in a mouse model of colitis, HIF-2α is specifically stabilized in CD4+ type 1T helper (TH 1) cells, leading to the upregulation of miR-29a expression and suppression of TH 1 cell function, which pathway is potentially targetable for therapeutic purposes. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
25. IL-2 delivery to CD8+ T cells during infection requires MRTF/SRF-dependent gene expression and cytoskeletal dynamics.
- Author
-
Maurice, Diane, Costello, Patrick, Diring, Jessica, Gualdrini, Francesco, Frederico, Bruno, and Treisman, Richard
- Subjects
TRANSCRIPTION factors ,T cell differentiation ,T cells ,REGULATOR genes ,GENE expression - Abstract
Paracrine IL-2 signalling drives the CD8 + T cell expansion and differentiation that allow protection against viral infections, but the underlying molecular events are incompletely understood. Here we show that the transcription factor SRF, a master regulator of cytoskeletal gene expression, is required for effective IL-2 signalling during L. monocytogenes infection. Acting cell-autonomously with its actin-regulated cofactors MRTF-A and MRTF-B, SRF is dispensible for initial TCR-mediated CD8
+ T cell proliferation, but is required for sustained IL-2 dependent CD8+ effector T cell expansion, and persistence of memory cells. Following TCR activation, Mrtfab-null CD8+ T cells produce IL-2 normally, but homotypic clustering is impaired both in vitro and in vivo. Expression of cytoskeletal structural and regulatory genes, most notably actins, is defective in Mrtfab-null CD8+ T cells. Activation-induced cell clustering in vitro requires F-actin assembly, and Mrtfab-null cell clusters are small, contain less F-actin, and defective in IL-2 retention. Clustering of Mrtfab-null cells can be partially restored by exogenous actin expression. IL-2 mediated CD8+ T cell proliferation during infection thus depends on the control of cytoskeletal dynamics and actin gene expression by MRTF-SRF signalling. The transcription factor SRF, together with its co-factors MRTF-A and MRTF-B, controls cytoskeletal gene expression. Here authors show that MRTF/SRF inactivation leads to decreased IL-2 mediated CD8 + T cell proliferation during infection, resulting from disrupted homotypic cell clustering and reduced retention of IL-2. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
26. Targeting PRMT3 impairs methylation and oligomerization of HSP60 to boost anti-tumor immunity by activating cGAS/STING signaling.
- Author
-
Shi, Yunxing, Wu, Zongfeng, Liu, Shaoru, Zuo, Dinglan, Niu, Yi, Qiu, Yuxiong, Qiao, Liang, He, Wei, Qiu, Jiliang, Yuan, Yunfei, Wang, Guocan, and Li, Binkui
- Subjects
IMMUNE checkpoint proteins ,PROTEIN arginine methyltransferases ,HOMEOSTASIS ,HEPATOCELLULAR carcinoma ,LEAKAGE ,T cells ,MITOCHONDRIAL DNA - Abstract
Immune checkpoint blockade (ICB) has emerged as a promising therapeutic option for hepatocellular carcinoma (HCC), but resistance to ICB occurs and patient responses vary. Here, we uncover protein arginine methyltransferase 3 (PRMT3) as a driver for immunotherapy resistance in HCC. We show that PRMT3 expression is induced by ICB-activated T cells via an interferon-gamma (IFNγ)-STAT1 signaling pathway, and higher PRMT3 expression levels correlate with reduced numbers of tumor-infiltrating CD8
+ T cells and poorer response to ICB. Genetic depletion or pharmacological inhibition of PRMT3 elicits an influx of T cells into tumors and reduces tumor size in HCC mouse models. Mechanistically, PRMT3 methylates HSP60 at R446 to induce HSP60 oligomerization and maintain mitochondrial homeostasis. Targeting PRMT3-dependent HSP60 methylation disrupts mitochondrial integrity and increases mitochondrial DNA (mtDNA) leakage, which results in cGAS/STING-mediated anti-tumor immunity. Lastly, blocking PRMT3 functions synergize with PD-1 blockade in HCC mouse models. Our study thus identifies PRMT3 as a potential biomarker and therapeutic target to overcome immunotherapy resistance in HCC. Resistance to immune checkpoint blockade (ICB) is a challenge in hepatocellular carcinoma therapy. Here, the authors show that targeting PRMT3 markedly improves the anticancer efficacy of ICB via interrupting HSP60 methylation and oligomerization, damaging mitochondrial integrity, and thereby activating cGAS/STING-mediated anti-tumor immunity. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
27. Genomic and immune heterogeneity of multiple synchronous lung adenocarcinoma at different developmental stages.
- Author
-
Zhao, Yue, Gao, Jian, Wang, Jun, Fan, Fanfan, Cheng, Chao, Qian, Danwen, Guo, Ran, Zhang, Yang, Ye, Ting, Augustine, Marcellus, Lin, Yicong, Shang, Jun, Li, Hang, Pan, Yunjian, Huang, Qingyuan, Chen, Haiqing, Han, Han, Gao, Zhendong, Wang, Qiming, and Zhang, Shiyue
- Subjects
REGULATORY T cells ,LUNG cancer ,T cells ,RNA sequencing ,CONVERGENT evolution - Abstract
Multiple synchronous lung cancers (MSLCs) constitute a unique subtype of lung cancer. To explore the genomic and immune heterogeneity across different pathological stages of MSLCs, we analyse 16 MSLCs from 8 patients using single-cell RNA-seq, single-cell TCR sequencing, and bulk whole-exome sequencing. Our investigation indicates clonally independent tumours with convergent evolution driven by shared driver mutations. However, tumours from the same individual exhibit few shared mutations, indicating independent origins. During the transition from pre-invasive to invasive adenocarcinoma, we observe a shift in T cell phenotypes characterized by increased Treg cells and exhausted CD8
+ T cells, accompanied by diminished cytotoxicity. Additionally, invasive adenocarcinomas exhibit greater neoantigen abundance and a more diverse TCR repertoire, indicating heightened heterogeneity. In summary, despite having a common genetic background and environmental exposure, our study emphasizes the individuality of MSLCs at different stages, highlighting their unique genomic and immune characteristics. Multiple synchronous lung cancers (MSLCs) are a subtype of lung cancer. Here the authors characterise MSLCs using single cell RNA sequencing, single cell TCR sequencing and bulk whole-exome sequencing to investigate the mutations that arise in and are associated with invasive adenocarcinoma development, and immune microenvironment changes in this process. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
28. Enhanced tumor response to adoptive T cell therapy with PHD2/3-deficient CD8 T cells.
- Author
-
Dvorakova, Tereza, Finisguerra, Veronica, Formenti, Matteo, Loriot, Axelle, Boudhan, Loubna, Zhu, Jingjing, and Van den Eynde, Benoit J.
- Subjects
CELL metabolism ,CELL physiology ,HYPOXIA-inducible factors ,IMMUNOSUPPRESSION ,TUMOR microenvironment ,T cells - Abstract
While adoptive cell therapy has shown success in hematological malignancies, its potential against solid tumors is hindered by an immunosuppressive tumor microenvironment (TME). In recent years, members of the hypoxia-inducible factor (HIF) family have gained recognition as important regulators of T-cell metabolism and function. The role of HIF signalling in activated CD8 T cell function in the context of adoptive cell transfer, however, has not been explored in full depth. Here we utilize CRISPR-Cas9 technology to delete prolyl hydroxylase domain-containing enzymes (PHD) 2 and 3, thereby stabilizing HIF-1 signalling, in CD8 T cells that have already undergone differentiation and activation, modelling the T cell phenotype utilized in clinical settings. We observe a significant boost in T-cell activation and effector functions following PHD2/3 deletion, which is dependent on HIF-1α, and is accompanied by an increased glycolytic flux. This improvement in CD8 T cell performance translates into an enhancement in tumor response to adoptive T cell therapy in mice, across various tumor models, even including those reported to be extremely resistant to immunotherapeutic interventions. These findings hold promise for advancing CD8 T-cell based therapies and overcoming the immune suppression barriers within challenging tumor microenvironments. The hypoxia inducible factor HIF-1α has been shown to regulate T cell metabolism and function. Here authors deleted the prolyl hydroxylase domain-containing enzymes PHD2 and 3, thereby stabilizing HIF-1α, in therapeutic CD8 T cells to achieve better functionality upon adoptive transfer to tumour-bearing mice. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. Discovery of an unconventional lamprey lymphocyte lineage highlights divergent features in vertebrate adaptive immune system evolution.
- Author
-
Huang, Yingyi, Liu, Xiang, Li, Shuo, Li, Chen, Wang, Hong-Yan, Liu, Qun, Chen, Jian-Yang, Zhang, Yingying, Li, Yanan, Zhang, Xianghui, Wang, Qian, Liu, Kaiqiang, Liu, Yu-Yan, Pang, Yue, Liu, Shanshan, Fan, Guangyi, and Shao, Changwei
- Subjects
HEMATOPOIETIC growth factors ,BIOLOGICAL evolution ,LYMPHOCYTE subsets ,T cells ,RNA sequencing - Abstract
Lymphocyte receptors independently evolved in both jawed and jawless vertebrates with similar adaptive immune responses. However, the diversity of functional subtypes and molecular architecture in jawless vertebrate lymphocytes, comparable to jawed species, is not well defined. Here, we profile the gills, intestines, and blood of the lamprey, Lampetra morii, with single-cell RNA sequencing, using a full-length transcriptome as a reference. Our findings reveal higher tissue-specific heterogeneity among T-like cells in contrast to B-like cells. Notably, we identify a unique T-like cell subtype expressing a homolog of the nonlymphoid hematopoietic growth factor receptor, MPL-like (MPL-L). These MPL-L+ T-like cells exhibit features distinct from T cells of jawed vertebrates, particularly in their elevated expression of hematopoietic genes. We further discovered that MPL-L
+ VLRA+ T-like cells are widely present in the typhlosole, gill, liver, kidney, and skin of lamprey and they proliferate in response to both a T cell mitogen and recombinant human thrombopoietin. These findings provide new insights into the adaptive immune response in jawless vertebrates, shedding new light on the evolution of adaptive immunity. Lymphocyte subsets remain inadequately understood in jawless vertebrates, relative to jawed vertebrates. In this study, the authors combine single-cell RNA and whole-transcriptome sequencing to demonstrate that the lamprey, Lampreta morii, harbours specialised T-like cells that are distinct from those found in jawed vertebrates. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
30. Androgens contribute to sex bias of autoimmunity in mice by T cell-intrinsic regulation of Ptpn22 phosphatase expression.
- Author
-
Lee, Jean, Yurkovetskiy, Leonid A., Reiman, Derek, Frommer, Lara, Strong, Zoe, Chang, Anthony, Kahaly, George J., Khan, Aly A., and Chervonsky, Alexander V.
- Subjects
TYPE 1 diabetes ,T cell receptors ,SYSTEMIC lupus erythematosus ,T cells ,AUTOIMMUNE diseases - Abstract
Autoimmune diseases such as systemic lupus erythematosus (SLE) display a strong female bias. Although sex hormones have been associated with protecting males from autoimmunity, the molecular mechanisms are incompletely understood. Here we report that androgen receptor (AR) expressed in T cells regulates genes involved in T cell activation directly, or indirectly via controlling other transcription factors. T cell-specific deletion of AR in mice leads to T cell activation and enhanced autoimmunity in male mice. Mechanistically, Ptpn22, a phosphatase and negative regulator of T cell receptor signaling, is downregulated in AR-deficient T cells. Moreover, a conserved androgen-response element is found in the regulatory region of Ptpn22 gene, and the mutation of this transcription element in non-obese diabetic mice increases the incidence of spontaneous and inducible diabetes in male mice. Lastly, Ptpn22 deficiency increases the disease severity of male mice in a mouse model of SLE. Our results thus implicate AR-regulated genes such as PTPN22 as potential therapeutic targets for autoimmune diseases. Androgen is a sex hormone that may contribute to sex biases in autoimmunity. Here the authors show that in T cells androgen induces the expression of Ptpn22 phosphatase to negatively regulate T cell activation, thereby contributing to protecting males from major autoimmune diseases such as systemic lupus erythematosus and type 1 diabetes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Publisher Correction: ER stress-induced mediator C/EBP homologous protein thwarts effector T cell activity in tumors through T-bet repression.
- Author
-
Cao, Yu, Trillo-Tinoco, Jimena, Sierra, Rosa A., Anadon, Carmen, Dai, Wenjie, Mohamed, Eslam, Cen, Ling, Costich, Tara L., Magliocco, Anthony, Marchion, Douglas, Klar, Richard, Michel, Sven, Jaschinski, Frank, Reich, Richard R., Mehrotra, Shikhar, Cubillos-Ruiz, Juan R., Munn, David H., Conejo-Garcia, Jose R., and Rodriguez, Paulo C.
- Subjects
ENDOPLASMIC reticulum ,T cells ,TUMORS - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
32. Author Correction: Human FCHO1 deficiency reveals role for clathrin-mediated endocytosis in development and function of T cells.
- Author
-
Łyszkiewicz, Marcin, Ziętara, Natalia, Frey, Laura, Pannicke, Ulrich, Stern, Marcel, Liu, Yanshan, Fan, Yanxin, Puchałka, Jacek, Hollizeck, Sebastian, Somekh, Ido, Rohlfs, Meino, Yilmaz, Tuğba, Ünal, Ekrem, Karakukcu, Musa, Patiroğlu, Türkan, Kellerer, Christina, Karasu, Ebru, Sykora, Karl-Walter, Lev, Atar, and Simon, Amos
- Subjects
T cells ,ENDOCYTOSIS ,CELL physiology - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
33. CAR T-cell-mediated delivery of bispecific innate immune cell engagers for neuroblastoma.
- Author
-
Pascual-Pasto, Guillem, McIntyre, Brendan, Hines, Margaret G., Giudice, Anna M., Garcia-Gerique, Laura, Hoffmann, Jennifer, Mishra, Pamela, Matlaga, Stephanie, Lombardi, Simona, Shraim, Rawan, Schürch, Patrick M., Yarmarkovich, Mark, Hofmann, Ted J., Alikarami, Fatemeh, Martinez, Daniel, Tsang, Matthew, Gil-de-Gómez, Luis, Spear, Timothy T., Bernt, Kathrin M., and Wolpaw, Adam J.
- Subjects
CHIMERIC antigen receptors ,ENGINEERS ,CYTOTOXINS ,TREATMENT effectiveness ,NEUROBLASTOMA ,T cells - Abstract
Novel chimeric antigen receptor (CAR) T-cell approaches are needed to improve therapeutic efficacy in solid tumors. High-risk neuroblastoma is an aggressive pediatric solid tumor that expresses cell-surface GPC2 and GD2 with a tumor microenvironment infiltrated by CD16a-expressing innate immune cells. Here we engineer T-cells to express a GPC2-directed CAR and simultaneously secrete a bispecific innate immune cell engager (BiCE) targeting both GD2 and CD16a. In vitro, GPC2.CAR-GD2.BiCE T-cells induce GPC2-dependent cytotoxicity and secrete GD2.BiCE that promotes GD2-dependent activation of antitumor innate immunity. In vivo, GPC2.CAR-GD2.BiCE T-cells locally deliver GD2.BiCE and increase intratumor retention of NK-cells. In mice bearing neuroblastoma patient-derived xenografts and reconstituted with human CD16a-expressing immune cells, GD2.BiCEs enhance GPC2.CAR antitumor efficacy. A CAR.BiCE strategy should be considered for tumor histologies where antigen escape limits CAR efficacy, especially for solid tumors like neuroblastoma that are infiltrated by innate immune cells. GPC2 and GD2 have been described as immunotherapeutic targets in neuroblastoma. Here the authors engineer T cells to simultaneously express a GPC2-directed CAR and a bispecific innate immune cell engager targeting GD2 and CD16a, showing antitumor activity in neuroblastoma preclinical models. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. ITGB6 modulates resistance to anti-CD276 therapy in head and neck cancer by promoting PF4+ macrophage infiltration.
- Author
-
Zhang, Caihua, Li, Kang, Zhu, Hongzhang, Cheng, Maosheng, Chen, Shuang, Ling, Rongsong, Wang, Cheng, and Chen, Demeng
- Subjects
HEAD & neck cancer ,IMMUNE checkpoint inhibitors ,GENE expression ,SQUAMOUS cell carcinoma ,RNA sequencing ,T cells - Abstract
Enoblituzumab, an immunotherapeutic agent targeting CD276, shows both safety and efficacy in activating T cells and oligodendrocyte-like cells against various cancers. Preclinical studies and mouse models suggest that therapies targeting CD276 may outperform PD1/PD-L1 blockade. However, data from mouse models indicate a significant non-responsive population to anti-CD276 treatment, with the mechanisms of resistance still unclear. In this study, we evaluate the activity of anti-CD276 antibodies in a chemically-induced murine model of head and neck squamous cell carcinoma. Using models of induced and orthotopic carcinogenesis, we identify ITGB6 as a key gene mediating differential responses to anti-CD276 treatment. Through single-cell RNA sequencing and gene-knockout mouse models, we find that ITGB6 regulates the expression of the tumor-associated chemokine CX3CL1, which recruits and activates PF4
+ macrophages that express high levels of CX3CR1. Inhibition of the CX3CL1-CX3CR1 axis suppresses the infiltration and secretion of CXCL16 by PF4+ macrophages, thereby reinvigorating cytotoxic CXCR6+ CD8+ T cells and enhancing sensitivity to anti-CD276 treatment. Further investigations demonstrate that inhibiting ITGB6 restores sensitivity to PD1 antibodies in mice resistant to anti-PD1 treatment. In summary, our research reveals a resistance mechanism associated with immune checkpoint inhibitor therapy and identifies potential targets to overcome resistance in cancer treatment. Response rate to anti-CD276 based immunotherapy remains suboptimal in patients with cancer. Here, in a chemically-induced murine model of head and neck squamous cell carcinoma, the authors show that expression of ITGB6 in tumor cells promotes resistance to anti-CD276 therapy, associated with accumulation of PF4+ macrophages and T cell dysfunction. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
35. Targeting ERK-MYD88 interaction leads to ERK dysregulation and immunogenic cancer cell death.
- Author
-
Virard, François, Giraud, Stéphane, Bonnet, Mélanie, Magadoux, Léa, Martin, Laetitia, Pham, Thuy Ha, Skafi, Najwa, Deneuve, Sophie, Frem, Rita, Villoutreix, Bruno O., Sleiman, Nawal Hajj, Reboulet, Jonathan, Merabet, Samir, Chaptal, Vincent, Chaveroux, Cédric, Hussein, Nader, Aznar, Nicolas, Fenouil, Tanguy, Treilleux, Isabelle, and Saintigny, Pierre
- Subjects
CELL transformation ,MITOGEN-activated protein kinases ,ADAPTOR proteins ,CANCER cells ,CELL death ,T cells - Abstract
The quest for targeted therapies is critical in the battle against cancer. The RAS/MAP kinase pathway is frequently implicated in neoplasia, with ERK playing a crucial role as the most distal kinase in the RAS signaling cascade. Our previous research demonstrated that the interaction between ERK and MYD88, an adaptor protein in innate immunity, is crucial for RAS-dependent transformation and cancer cell survival. In this study, we examine the biological consequences of disrupting the ERK-MYD88 interaction through the ERK D-recruitment site (DRS), while preserving ERK's kinase activity. Our results indicate that EI-52, a small-molecule benzimidazole targeting ERK-MYD88 interaction induces an HRI-mediated integrated stress response (ISR), resulting in immunogenic apoptosis specific to cancer cells. Additionally, EI-52 exhibits anti-tumor efficacy in patient-derived tumors and induces an anti-tumor T cell response in mice in vivo. These findings suggest that inhibiting the ERK-MYD88 interaction may be a promising therapeutic approach in cancer treatment. The interaction between MyD88 and ERK is necessary for RAS-dependent transformation and cancer cell survival. Here, the authors identify benzimidazole compound EI-52 as a disrupter of this interaction and demonstrate its therapeutic efficacy in tumours via ERK dysregulation and the induction of immunogenic cell death. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Inhibition of glycosphingolipid synthesis with eliglustat in combination with immune checkpoint inhibitors in advanced cancers: preclinical evidence and phase I clinical trial.
- Author
-
Dong, Liang, Cao, Zhi, Chen, Meixia, Liu, Yang, Ma, Xinran, Lu, Yuting, Zhang, Yan, Feng, Kaichao, Zhang, Yang, Meng, Zhenzhen, Yang, Qingming, Wang, Yao, Wu, Zhiqiang, and Han, Weidong
- Subjects
IMMUNE checkpoint inhibitors ,MAJOR histocompatibility complex ,CANCER cells ,IMMUNE response ,GLYCOSPHINGOLIPIDS ,T cell receptors ,T cells - Abstract
Glycosphingolipids (GSLs) are abundantly expressed in cancer cells. The effects of GSL-targeted immunotherapies are not fully understood. Here, we show that the inhibition of GSL synthesis with the UDP-glucose ceramide glucosyltransferase inhibitor eliglustat can increase the exposure of the major histocompatibility complex (MHC) and tumour antigen peptides, enhancing the antitumour response of CD8
+ T cells in a range of tumour models. We therefore conducted a proof-of-concept phase I trial on the combination of eliglustat and an anti-PD-1 antibody for the treatment of advanced cancers (NCT04944888). The primary endpoints were safety and feasibility, and the secondary endpoint was antitumor activity. All prespecified endpoints were met. Among the 31 enrolled patients, only 1 patient experienced a grade 3 adverse event (AE), and no grade 4 AEs were observed. The objective response rate was 22.6% and the disease control rate reached 71%. Of the 8 patients with proficient mismatch repair/microsatellite stable (pMMR/MSS) colorectal cancer, one achieved complete response and two each had partial response and stable disease. In summary, inhibiting the synthesis of GSLs might represent an effective immunotherapy approach. Glycosphingolipids (GSLs) are abnormally expressed in cancer cells. Here, in addition to showing that inhibition of the synthesis of GSLs with eliglustat promotes anti-tumor immune responses in preclinical models, the authors report the results of a phase I trial of eliglustat and anti-PD-1 antibodies for the treatment of advanced cancers. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
37. TFEB activation hallmarks antigenic experience of B lymphocytes and directs germinal center fate decisions.
- Author
-
Münchhalfen, Matthias, Görg, Richard, Haberl, Michael, Löber, Jens, Willenbrink, Jakob, Schwarzt, Laura, Höltermann, Charlotte, Ickes, Christian, Hammermann, Leonard, Kus, Jan, Chapuy, Björn, Ballabio, Andrea, Reichardt, Sybille D., Flügel, Alexander, Engels, Niklas, and Wienands, Jürgen
- Subjects
TRANSCRIPTION factors ,B cells ,IMMUNOLOGIC memory ,T cells ,GERMINAL centers ,B cell receptors ,CHEMOKINE receptors - Abstract
Ligation of the B cell antigen receptor (BCR) initiates humoral immunity. However, BCR signaling without appropriate co-stimulation commits B cells to death rather than to differentiation into immune effector cells. How BCR activation depletes potentially autoreactive B cells while simultaneously primes for receiving rescue and differentiation signals from cognate T lymphocytes remains unknown. Here, we use a mass spectrometry-based proteomic approach to identify cytosolic/nuclear shuttling elements and uncover transcription factor EB (TFEB) as a central BCR-controlled rheostat that drives activation-induced apoptosis, and concurrently promotes the reception of co-stimulatory rescue signals by supporting B cell migration and antigen presentation. CD40 co-stimulation prevents TFEB-driven cell death, while enhancing and prolonging TFEB's nuclear residency, which hallmarks antigenic experience also of memory B cells. In mice, TFEB shapes the transcriptional landscape of germinal center B cells. Within the germinal center, TFEB facilitates the dark zone entry of light-zone-residing centrocytes through regulation of chemokine receptors and, by balancing the expression of Bcl-2/BH3-only family members, integrates antigen-induced apoptosis with T cell-provided CD40 survival signals. Thus, TFEB reprograms antigen-primed germinal center B cells for cell fate decisions. B cell receptor activation leads to contrary outcomes in the absence and presence of co-stimulation. Here, the authors show transcription factor EB acts as a B cell receptor-controlled rheostat that balances activation-induced cell death with co-stimulatory rescue signals, collectively reprograming antigen-primed germinal center B cells for fate decisions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Dysfunctional tumor-infiltrating Vδ1 + T lymphocytes in microsatellite-stable colorectal cancer.
- Author
-
Stary, Victoria, Pandey, Ram V., List, Julia, Kleissl, Lisa, Deckert, Florian, Kabiljo, Julijan, Laengle, Johannes, Gerakopoulos, Vasileios, Oehler, Rudolf, Watzke, Lukas, Farlik, Matthias, Lukowski, Samuel W., Vogt, Anne B., Stary, Georg, Stockinger, Hannes, Bergmann, Michael, and Pilat, Nina
- Subjects
T cells ,IMMUNOSUPPRESSION ,IMMUNE recognition ,TUMOR-infiltrating immune cells ,COLORECTAL cancer - Abstract
Although γδ T cells are known to participate in immune dysregulation in solid tumors, their relevance to human microsatellite-stable (MSS) colorectal cancer (CRC) is still undefined. Here, using integrated gene expression analysis and T cell receptor sequencing, we characterized γδ T cells in MSS CRC, with a focus on Vδ1 + T cells. We identified Vδ1
+ T cells with shared motifs in the third complementarity-determining region of the δ-chain, reflective of antigen recognition. Changes in gene and protein expression levels suggested a dysfunctional effector state of Vδ1+ T cells in MSS CRC, distinct from Vδ1+ T cells in microsatellite-instable (MSI). Interaction analysis highlighted an immunosuppressive role of fibroblasts in the dysregulation of Vδ1+ T cells in MSS CRC via the TIGIT-NECTIN2 axis. Blocking this pathway with a TIGIT antibody partially restored cytotoxicity of the dysfunctional Vδ1 phenotype. These results define an operative pathway in γδ T cells in MSS CRC. Although γδ T cells are known to participate in immune dysregulation in solid tumors, their relevance to human microsatellite-stable (MSS) colorectal cancer (CRC) is less well-studied. Here, using single-cell RNA-sequencing, the authors identify a Vδ1 + T cell subset, which are functionally impaired in MSS CRC via a TIGIT-NECTIN2 interaction. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
39. Single-cell profiling identifies a CD8bright CD244bright Natural Killer cell subset that reflects disease activity in HLA-A29-positive birdshot chorioretinopathy.
- Author
-
Nath, Pulak R., Maclean, Mary, Nagarajan, Vijay, Lee, Jung Wha, Yakin, Mehmet, Kumar, Aman, Nadali, Hadi, Schmidt, Brian, Kaya, Koray D., Kodati, Shilpa, Young, Alice, Caspi, Rachel R., Kuiper, Jonas J. W., and Sen, H. Nida
- Subjects
KILLER cells ,T cells ,CELL receptors ,CELLULAR recognition ,CELL populations - Abstract
Birdshot chorioretinopathy is an inflammatory eye condition strongly associated with MHC-I allele HLA-A29. The striking association with MHC-I suggests involvement of T cells, whereas natural killer (NK) cell involvement remains largely unstudied. Here we show that HLA-A29-positive birdshot chorioretinopathy patients have a skewed NK cell pool containing expanded CD16 positive NK cells which produce more proinflammatory cytokines. These NK cells contain populations that express CD8A which is involved in MHC-I recognition on target cells, display gene signatures indicative of high cytotoxic activity (GZMB, PRF1 and ISG15), and signaling through NK cell receptor CD244 (SH2D1B). Long-term monitoring of a cohort of birdshot chorioretinopathy patients with active disease identifies a population of CD8
bright CD244bright NK cells, which rapidly declines to normal levels upon clinical remission following successful treatment. Collectively, these studies implicate CD8bright CD244bright NK cells in birdshot chorioretinopathy. Birdshot chorioretinopathy is an inflammatory eye condition strongly associated with HLA-A29. Here the authors use a single cell RNA sequencing approach to characterise NK cell involvement in this disease and show that CD8bright CD244bright NK cells are associated with active disease. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
40. Targeting myeloid-derived suppressor cells promotes antiparasitic T-cell immunity and enhances the efficacy of PD-1 blockade (15 words).
- Author
-
Zhang, Chuanshan, Wang, Hui, Aji, Tuerganaili, Li, Zhide, Li, Yinshi, Ainiwaer, Abidan, Rousu, Zibigu, Li, Jing, Wang, Maolin, Deng, Bingqing, duolikun, Adilai, Kang, Xuejiao, Zheng, Xuran, Yu, Qian, Shao, Yingmei, Zhang, Wenbao, Vuitton, Dominique A., Tian, Zhigang, Sun, Haoyu, and Wen, Hao
- Subjects
MYELOID-derived suppressor cells ,SUPPRESSOR cells ,T cells ,PROGRAMMED cell death 1 receptors ,ECHINOCOCCUS multilocularis ,IMMUNE response - Abstract
Immune exhaustion corresponds to a loss of effector function of T cells that associates with cancer or chronic infection. Here, our objective was to decipher the mechanisms involved in the immune suppression of myeloid-derived suppressor cells (MDSCs) and to explore the potential to target these cells for immunotherapy to enhance checkpoint blockade efficacy in a chronic parasite infection. We demonstrated that programmed cell-death-1 (PD-1) expression was significantly upregulated and associated with T-cell dysfunction in advanced alveolar echinococcosis (AE) patients and in Echinococcus multilocularis-infected mice. PD-1 blockade ex vivo failed to reverse AE patients' peripheral blood T-cell dysfunction. PD-1/PD-L1 blockade or PD-1 deficiency had no significant effects on metacestode in mouse model. This was due to the inhibitory capacities of immunosuppressive granulocytic MDSCs (G-MDSCs), especially in the liver surrounding the parasite pseudotumor. MDSCs suppressed T-cell function in vitro in an indoleamine 2, 3 dioxygenase 1 (IDO1)-dependent manner. Although depleting MDSCs alone restored T-cell effector functions and led to some limitation of disease progression in E. multilocularis-infected mice, combination with PD-1 blockade was better to induce antiparasitic efficacy. Our findings provide preclinical evidence in support of targeting MDSC or combining such an approach with checkpoint blockade in patients with advanced AE. (200 words) Myeloid-derived suppressor cells play an important role in immune suppression caused by Echinococcus multilocularis infection. This study demonstrates that the removal of this cell type effectively restores T-cell function and potentiates PD-1 blockade therapy in chronic liver infection. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. Ca2+ transients on the T cell surface trigger rapid integrin activation in a timescale of seconds.
- Author
-
Li, Yue, Wang, ShiHui, Zhang, YouHua, Liu, ZhaoYuan, Zheng, YunZhe, Zhang, Kun, Chen, ShiYang, Lv, XiaoYing, Huang, MengWen, Pan, XingChao, Zheng, YaJuan, Yuan, MengYa, Ge, GaoXiang, Zeng, Yi Arial, Lin, ChangDong, and Chen, JianFeng
- Subjects
T cells ,LYMPHOCYTE transformation ,SKIN inflammation ,LYMPHOCYTES ,INTEGRINS ,CHEMOKINE receptors ,LABORATORY mice - Abstract
One question in lymphocyte homing is how integrins are rapidly activated to enable immediate arrest of fast rolling lymphocytes upon encountering chemokines at target vascular beds given the slow chemokine-induced integrin inside-out activation. Herein we demonstrate that chemokine CCL25-triggered Ca
2+ influx induces T cell membrane-proximal external Ca2+ concentration ([Ca2+ ]ex ) drop in 6 s from physiological concentration 1.2 mM to 0.3 mM, a critical extracellular Ca2+ threshold for inducing αLβ2 activation, triggering rapid αLβ2 activation and T cell arrest before occurrence of αLβ2 inside-out activation. Talin knockdown inhibits the slow inside-out activation of αLβ2 but not [Ca2+ ]ex drop-triggered αLβ2 quick activation. Blocking Ca2+ influx significantly suppresses T cell rolling-to-arrest transition and homing to skin lesions in a mouse psoriasis model, thus alleviating skin inflammation. [Ca2+ ]ex decrease-triggered rapid integrin activation bridges the gap between initial chemokine stimulation and slow integrin inside-out activation, ensuring immediate lymphocyte arrest and subsequent diapedesis on the right location. Lymphocytes need to be slowed down rapidly to enter tissues. Here the authors characterise the arrest of lymphocytes and using a calcium biosensor propose that a rapid drop in extracellular calcium concentration results in integrin activation and lymphocyte arrest. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
42. Type II innate lymphoid cell plasticity contributes to impaired reconstitution after allogeneic hematopoietic stem cell transplantation.
- Author
-
Laurie, Sonia J., Foster II, Joseph P., Bruce, Danny W., Bommiasamy, Hemamalini, Kolupaev, Oleg V., Yazdimamaghani, Mostafa, Pattenden, Samantha G., Chao, Nelson J., Sarantopoulos, Stefanie, Parker, Joel S., Davis, Ian J., and Serody, Jonathan S.
- Subjects
HEMATOPOIETIC stem cell transplantation ,INNATE lymphoid cells ,T cells ,HEMATOPOIETIC stem cells ,MUCOUS membranes ,GRAFT versus host disease ,SMALL intestine ,HOMEOSTASIS ,IMMUNOPRECIPITATION - Abstract
Type II innate lymphoid cells (ILC2s) maintain homeostasis and barrier integrity in mucosal tissues. In both mice and humans, ILC2s poorly reconstitute after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Determining the mechanisms involved in their impaired reconstitution could improve transplant outcomes. By integrating single-cell chromatin and transcriptomic analyses of transplanted ILC2s, we identify a previously unreported population of converted ILC1-like cells in the mouse small intestine post-transplant. Exposure of ILC2s to proinflammatory cytokines resulted in a mixed ILC1-ILC2 phenotype but was able to convert only a small population of ILC2s to ILC1s, which were found post-transplant. Whereas ILC2s protected against acute graft-versus-host disease (aGVHD) mediated mortality, infusion of proinflammatory cytokine-exposed ILC2s accelerated aGvHD. Interestingly, murine ILC2 reconstitution post-HSCT is decreased in the presence of alloreactive T cells. Finally, peripheral blood cells from human patients with aGvHD have an altered ILC2-associated chromatin landscape compared to transplanted controls. These data demonstrate that following transplantation ILC2s convert to a pro-pathogenic population with an ILC1-like chromatin state and provide insights into the contribution of ILC plasticity to the impaired reconstitution of ILC2 cells, which is one of several potential mechanisms for the poor reconstitution of these important cells after allo-HSCT. Allogeneic hematopoietic stem cell transplantation restores the whole hematopoietic compartment of the recipient, but some cell types, such as Type II innate lymphoid cells (ILC2) are not reconstituted efficiently. Here authors show that ILC2s could be converted to ILC1-like cells in the mouse small intestine post-transplantation, which might contribute to the lower than physiological ILC2 numbers. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
43. Terminal deoxynucleotidyl transferase and CD84 identify human multi-potent lymphoid progenitors.
- Author
-
Kim, YeEun, Calderon, Ariel A., Favaro, Patricia, Glass, David R., Tsai, Albert G., Ho, Daniel, Borges, Luciene, Greenleaf, William J., and Bendall, Sean C.
- Subjects
TRANSCRIPTION factors ,BONE marrow ,TRANSFERASES ,T cells ,HEMATOPOIESIS ,HUMAN beings ,DNA polymerases - Abstract
Lymphoid specification in human hematopoietic progenitors is not fully understood. To better associate lymphoid identity with protein-level cell features, we conduct a highly multiplexed single-cell proteomic screen on human bone marrow progenitors. This screen identifies terminal deoxynucleotidyl transferase (TdT), a specialized DNA polymerase intrinsic to VDJ recombination, broadly expressed within CD34
+ progenitors prior to B/T cell emergence. While these TdT+ cells coincide with granulocyte-monocyte progenitor (GMP) immunophenotype, their accessible chromatin regions show enrichment for lymphoid-associated transcription factor (TF) motifs. TdT expression on GMPs is inversely related to the SLAM family member CD84. Prospective isolation of CD84lo GMPs demonstrates robust lymphoid potentials ex vivo, while still retaining significant myeloid differentiation capacity, akin to LMPPs. This multi-omic study identifies human bone marrow lymphoid-primed progenitors, further defining the lympho-myeloid axis in human hematopoiesis. How lymphoid and myeloid specification occurs in human haematopoietic progenitors is not fully understood. Here the authors perform a proteomic screen on human bone marrow progenitors and suggest TdT+ and CD84- progenitors as lymphoid-primed progenitors with residual myeloid potentials. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
44. Adeno-associated virus delivered CXCL9 sensitizes glioblastoma to anti-PD-1 immune checkpoint blockade.
- Author
-
von Roemeling, Christina A., Patel, Jeet A., Carpenter, Savannah L., Yegorov, Oleg, Yang, Changlin, Bhatia, Alisha, Doonan, Bently P., Russell, Rylynn, Trivedi, Vrunda S., Klippel, Kelena, Ryu, Daniel H., Grippin, Adam, Futch, Hunter S., Ran, Yong, Hoang-Minh, Lan B., Weidert, Frances L., Golde, Todd E., and Mitchell, Duane A.
- Subjects
IMMUNE checkpoint proteins ,ADENO-associated virus ,GLIOBLASTOMA multiforme ,TREATMENT effectiveness ,LYMPHOKINES ,T cells ,CHEMOKINE receptors - Abstract
There are numerous mechanisms by which glioblastoma cells evade immunological detection, underscoring the need for strategic combinatorial treatments to achieve appreciable therapeutic effects. However, developing combination therapies is difficult due to dose-limiting toxicities, blood-brain-barrier, and suppressive tumor microenvironment. Glioblastoma is notoriously devoid of lymphocytes driven in part by a paucity of lymphocyte trafficking factors necessary to prompt their recruitment and activation. Herein, we develop a recombinant adeno-associated virus (AAV) gene therapy that enables focal and stable reconstitution of the tumor microenvironment with C-X-C motif ligand 9 (CXCL9), a powerful call-and-receive chemokine for lymphocytes. By manipulating local chemokine directional guidance, AAV-CXCL9 increases tumor infiltration by cytotoxic lymphocytes, sensitizing glioblastoma to anti-PD-1 immune checkpoint blockade in female preclinical tumor models. These effects are accompanied by immunologic signatures evocative of an inflamed tumor microenvironment. These findings support AAV gene therapy as an adjuvant for reconditioning glioblastoma immunogenicity given its safety profile, tropism, modularity, and off-the-shelf capability. The limited infiltration and migration of T cells in the brain can hinder the success of immune checkpoint blockade in glioblastoma (GBM). Here the authors show that an adeno-associated virus-based gene therapy for the intratumor delivery of CXCL9 promotes T cell infiltration and enhances response to anti-PD1 in GBM preclinical models. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. Heterogeneity in lung macrophage control of Mycobacterium tuberculosis is modulated by T cells.
- Author
-
Lai, Rocky, Williams, Travis, Rakib, Tasfia, Lee, Jinhee, and Behar, Samuel M.
- Subjects
MYCOBACTERIUM tuberculosis ,T cells ,CELLULAR recognition ,ALVEOLAR macrophages ,LUNGS ,MACROPHAGES ,T cell receptors ,IMMUNOSENESCENCE - Abstract
Following Mycobacterium tuberculosis infection, alveolar macrophages are initially infected but ineffectively restrict bacterial replication. The distribution of M. tuberculosis among different cell types in the lung changes with the onset of T cell immunity when the dominant infected cellular niche shifts from alveolar to monocyte-derived macrophages (MDM). We hypothesize that changes in bacterial distribution among different cell types is driven by differences in T cell recognition of infected cells and their subsequent activation of antimicrobial effector mechanisms. We show that CD4 and CD8 T cells efficiently eliminate M. tuberculosis infection in alveolar macrophages, but they have less impact on suppressing infection in MDM, which may be a bacterial niche. Importantly, CD4 T cell responses enhance MDM recruitment to the lung. Thus, the outcome of infection depends on the interaction between the T cell subset and the infected cell; both contribute to the resolution and persistence of the infection. Both T cells and lung macrophages are involved in the control of lung tuberculosis infection. Here the authors examine the dependence of the lung macrophage response upon the presence of T cells and show that CD4 and CD8 T cells promote the elimination of M.Tb in alveolar macrophages but have less impact on monocyte-derived macrophages. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Using a pan-cancer atlas to investigate tumour associated macrophages as regulators of immunotherapy response.
- Author
-
Coulton, Alexander, Murai, Jun, Qian, Danwen, Thakkar, Krupa, Lewis, Claire E., and Litchfield, Kevin
- Subjects
MACROPHAGES ,T cells ,IMMUNE checkpoint proteins ,REFERENCE sources ,TUMORS - Abstract
The paradigm for macrophage characterization has evolved from the simple M1/M2 dichotomy to a more complex model that encompasses the broad spectrum of macrophage phenotypic diversity, due to differences in ontogeny and/or local stimuli. We currently lack an in-depth pan-cancer single cell RNA-seq (scRNAseq) atlas of tumour-associated macrophages (TAMs) that fully captures this complexity. In addition, an increased understanding of macrophage diversity could help to explain the variable responses of cancer patients to immunotherapy. Our atlas includes well established macrophage subsets as well as a number of additional ones. We associate macrophage composition with tumour phenotype and show macrophage subsets can vary between primary and metastatic tumours growing in sites like the liver. We also examine macrophage-T cell functional cross talk and identify two subsets of TAMs associated with T cell activation. Analysis of TAM signatures in a large cohort of immune checkpoint inhibitor-treated patients (CPI1000 +) identify multiple TAM subsets associated with response, including the presence of a subset of TAMs that upregulate collagen-related genes. Finally, we demonstrate the utility of our data as a resource and reference atlas for mapping of novel macrophage datasets using projection. Overall, these advances represent an important step in both macrophage classification and overcoming resistance to immunotherapies in cancer. Single cell sequencing can be used to examine tumour associated macrophages (TAM) and comparison between studies has been a challenge. Here the authors show a comparison tool to compare and contrast TAMs from different human tumour types and how these cells associate with T cells exploring further macrophage heterogeneity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
47. Three SARS-CoV-2 spike protein variants delivered intranasally by measles and mumps vaccines are broadly protective.
- Author
-
Zhang, Yuexiu, Chamblee, Michelle, Xu, Jiayu, Qu, Panke, Shamseldin, Mohamed M., Yoo, Sung J., Misny, Jack, Thongpan, Ilada, KC, Mahesh, Hall, Jesse M., Gupta, Yash A., Evans, John P., Lu, Mijia, Ye, Chengjin, Hsu, Cheng Chih, Liang, Xueya, Martinez-Sobrido, Luis, Yount, Jacob S., Boyaka, Prosper N., and Liu, Shan-Lu
- Subjects
SARS-CoV-2 Omicron variant ,MEASLES vaccines ,COMBINED vaccines ,COVID-19 vaccines ,SARS-CoV-2 ,T cells - Abstract
As the new SARS-CoV-2 Omicron variants and subvariants emerge, there is an urgency to develop intranasal, broadly protective vaccines. Here, we developed highly efficacious, intranasal trivalent SARS-CoV-2 vaccine candidates (TVC) based on three components of the MMR vaccine: measles virus (MeV), mumps virus (MuV) Jeryl Lynn (JL1) strain, and MuV JL2 strain. Specifically, MeV, MuV-JL1, and MuV-JL2 vaccine strains, each expressing prefusion spike (preS-6P) from a different variant of concern (VoC), were combined to generate TVCs. Intranasal immunization of IFNAR1
−/− mice and female hamsters with TVCs generated high levels of S-specific serum IgG antibodies, broad neutralizing antibodies, and mucosal IgA antibodies as well as tissue-resident memory T cells in the lungs. The immunized female hamsters were protected from challenge with SARS-CoV-2 original WA1, B.1.617.2, and B.1.1.529 strains. The preexisting MeV and MuV immunity does not significantly interfere with the efficacy of TVC. Thus, the trivalent platform is a promising next-generation SARS-CoV-2 vaccine candidate. In this study, the authors developed intranasal measles virus and mumps virus-based trivalent vaccines, each expressing three distinct SARS-CoV-2 stabilized prefusion spike proteins. They show that the intranasal vaccines provide protection against infection of SARS-CoV-2 variants in small animal models. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
48. Multi-modal generative modeling for joint analysis of single-cell T cell receptor and gene expression data.
- Author
-
Drost, Felix, An, Yang, Bonafonte-Pardàs, Irene, Dratva, Lisa M., Lindeboom, Rik G. H., Haniffa, Muzlifah, Teichmann, Sarah A., Theis, Fabian, Lotfollahi, Mohammad, and Schubert, Benjamin
- Subjects
GENE expression ,DEEP learning ,CELL physiology ,T cell receptors ,T cells ,KNOWLEDGE transfer - Abstract
Recent advances in single-cell immune profiling have enabled the simultaneous measurement of transcriptome and T cell receptor (TCR) sequences, offering great potential for studying immune responses at the cellular level. However, integrating these diverse modalities across datasets is challenging due to their unique data characteristics and technical variations. Here, to address this, we develop the multimodal generative model mvTCR to fuse modality-specific information across transcriptome and TCR into a shared representation. Our analysis demonstrates the added value of multimodal over unimodal approaches to capture antigen specificity. Notably, we use mvTCR to distinguish T cell subpopulations binding to SARS-CoV-2 antigens from bystander cells. Furthermore, when combined with reference mapping approaches, mvTCR can map newly generated datasets to extensive T cell references, facilitating knowledge transfer. In summary, we envision mvTCR to enable a scalable analysis of multimodal immune profiling data and advance our understanding of immune responses. Although single-cell RNA sequencing analysis now allows simultaneous examination of transcriptome and T cell receptor repertoire sequences, integrating these two modalities remains a challenge. Here, the authors develop mvTCR, a generative deep learning model that integrates transcriptome and T cell receptor data into a joint representation capturing cell functions and phenotypes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
49. HIV-1 Vpr combats the PU.1-driven antiviral response in primary human macrophages.
- Author
-
Virgilio, Maria C., Ramnani, Barkha, Chen, Thomas, Disbennett, W. Miguel, Lubow, Jay, Welch, Joshua D., and Collins, Kathleen L.
- Subjects
TRANSCRIPTION factors ,HIV ,MACROPHAGES ,VIRAL transmission ,IMMUNE response ,PROTEOLYSIS ,T cells - Abstract
HIV-1 Vpr promotes efficient spread of HIV-1 from macrophages to T cells by transcriptionally downmodulating restriction factors that target HIV-1 Envelope protein (Env). Here we find that Vpr induces broad transcriptomic changes by targeting PU.1, a transcription factor necessary for expression of host innate immune response genes, including those that target Env. Consistent with this, we find silencing PU.1 in infected macrophages lacking Vpr rescues Env. Vpr downmodulates PU.1 through a proteasomal degradation pathway that depends on physical interactions with PU.1 and DCAF1, a component of the Cul4A E3 ubiquitin ligase. The capacity for Vpr to target PU.1 is highly conserved across primate lentiviruses. In addition to impacting infected cells, we find that Vpr suppresses expression of innate immune response genes in uninfected bystander cells, and that virion-associated Vpr can degrade PU.1. Together, we demonstrate Vpr counteracts PU.1 in macrophages to blunt antiviral immune responses and promote viral spread. Virgilio et al show that HIV Vpr promotes the degradation of the myeloid transcription factor, PU.1, to prevent the expression of PU.1-regulated antiviral factors that would otherwise target HIV Env and inhibit viral spread in macrophages. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
50. CD4+ T cells display a spectrum of recall dynamics during re-infection with malaria parasites.
- Author
-
Lee, Hyun Jae, Moreira, Marcela L., Li, Shihan, Asatsuma, Takahiro, Williams, Cameron G., Skinner, Oliver P., Asad, Saba, Bramhall, Michael, Jiang, Zhe, Liu, Zihan, Kerr, Ashlyn S., Engel, Jessica A., Soon, Megan S. F., Straube, Jasmin, Barrera, Irving, Murray, Evan, Chen, Fei, Nideffer, Jason, Jagannathan, Prasanna, and Haque, Ashraful
- Subjects
T cells ,REINFECTION ,PLASMODIUM ,GRAPHICAL user interfaces ,T cell receptors ,TRANSCRIPTOMES - Abstract
Children in malaria-endemic regions can experience repeated Plasmodium infections over short periods of time. Effects of re-infection on multiple co-existing CD4
+ T cell subsets remain unresolved. Here, we examine antigen-experienced CD4+ T cells during re-infection in mice, using scRNA-seq/TCR-seq and spatial transcriptomics. TCR transgenic TEM cells initiate rapid Th1/Tr1 recall responses prior to proliferating, while GC Tfh counterparts are refractory, with TCM /Tfh-like cells exhibiting modest non-proliferative responses. Th1-recall is a partial facsimile of primary Th1-responses, with no upregulated effector-associated genes being unique to recall. Polyclonal, TCR-diverse, CD4+ T cells exhibit similar recall dynamics, with individual clones giving rise to multiple effectors including highly proliferative Th1/Tr1 cells, as well as GC Tfh and Tfh-like cells lacking proliferative capacity. Thus, we show substantial diversity in recall responses mounted by multiple co-existing CD4+ T cell subsets in the spleen, and present graphical user interfaces for studying gene expression dynamics and clonal relationships during re-infection. CD4+ T cells are known to be important in Plasmodium infection. Here the authors use mouse models to track antigen-experienced TCR transgenic and polyclonal CD4+ T cells during Plasmodium re-infection, and show different T cell phenotypes and varied responses in different areas of the spleen. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.