
Local TSH/TSHR signaling promotes CD8+ T cell exhaustion and immune evasion in colorectal carcinoma
Sisi Zeng, Huiling Hu, Zhiyang Li, Qi Hu, Rong Shen, Mingzhou Li, Yunshi Liang, Zuokang Mao, Yandong Zhang, Wanqi Zhan, Qin Zhu, Feifei Wang, Jianbiao Xiao, Bohan Xu, Guanglong Liu, Yanan Wang, Bingsong Li, Shaowan Xu, Zhaowen Zhang, Ceng Zhang, Zhizhang Wang, Li Liang
Cancer Communications ›› 2024, Vol. 44 ›› Issue (11) : 1287-1310.
Local TSH/TSHR signaling promotes CD8+ T cell exhaustion and immune evasion in colorectal carcinoma
Background: Dysfunction of CD8+ T cells in the tumor microenvironment (TME) contributes to tumor immune escape and immunotherapy tolerance. The effects of hormones such as leptin, steroid hormones, and glucocorticoids on T cell function have been reported previously. However, the mechanism underlying thyroid-stimulating hormone (TSH)/thyroid-stimulating hormone receptor (TSHR) signaling in CD8+ T cell exhaustion and tumor immune evasion remain poorly understood. This study was aimed at investigating the effects of TSH/TSHR signaling on the function of CD8+ T cells and immune evasion in colorectal cancer (CRC).
Methods: TSHR expression levels in CD8+ T cells were assessed with immunofluorescence and flow cytometry. Functional investigations involved manipulation of TSHR expression in cellular and mouse models to study its role in CD8+ T cells. Mechanistic insights were mainly gained through RNA-sequencing, Western blotting, chromatin immunoprecipitation and luciferase activity assay. Immunofluorescence, flow cytometry and Western blotting were used to investigate the source of TSH and TSHR in CRC tissues.
Results: TSHR was highly expressed in cancer cells and CD8+ T cells in CRC tissues. TSH/TSHR signaling was identified as the intrinsic pathway promoting CD8+ T cell exhaustion. Conditional deletion of TSHR in CD8+ tumor-infiltrating lymphocytes (TILs) improved effector differentiation and suppressed the expression of immune checkpoint receptors such as programmed cell death 1 (PD-1) and hepatitis A virus cellular receptor 2 (HAVCR2 or TIM3) through the protein kinase A (PKA)/cAMP-response element binding protein (CREB) signaling pathway. CRC cells secreted TSHR via exosomes to increase the TSHR level in CD8+ T cells, resulting in immunosuppression in the TME. Myeloid-derived suppressor cells (MDSCs) was the main source of TSH within the TME. Low expression of TSHR in CRC was a predictor of immunotherapy response.
Conclusions: The present findings highlighted the role of endogenous TSH/TSHR signaling in CD8+ T cell exhaustion and immune evasion in CRC. TSHR may be suitable as a predictive and therapeutic biomarker in CRC immunotherapy.
Thyroid stimulating hormone / Thyroid stimulating hormone receptor / Colorectal carcinoma / CD8+ T cell exhaustion / Immune evasion
[1] |
Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 2019; 16(6): 361-75.
CrossRef
Google scholar
|
[2] |
Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015; 15(8): 486-99.
CrossRef
Google scholar
|
[3] |
Dolina JS, Van Braeckel-Budimi. N, Thomas GD, Salek-Ardakani S. CD8(+) T Cell Exhaustion in Cancer. Front Immunol. 2021; 12: 715234.
CrossRef
Google scholar
|
[4] |
Budimir N, Thomas GD, Dolina JS, Salek-Ardakani S. Reversing T-cell Exhaustion in Cancer: Lessons Learned from PD-1/PD-L1 Immune Checkpoint Blockade. Cancer Immunol Res. 2022; 10(2): 146-53.
CrossRef
Google scholar
|
[5] |
Shi M, Ye L, Zhao L, He L, Chen J, Zhang J, et al. Tumor derived exosomal ENTPD2 impair CD8(+) T cell function in colon cancer through ATP-adenosine metabolism reprogramming. Cell Commun Signal. 2024; 22(1): 274.
CrossRef
Google scholar
|
[6] |
Stelekati E, Cai Z, Manne S, Chen Z, Beltra JC, Buchness LA, et al. MicroRNA-29a attenuates CD8 T cell exhaustion and induces memory-like CD8 T cells during chronic infection. Proc Natl Acad Sci U S A. 2022; 119(17): e2106083119.
CrossRef
Google scholar
|
[7] |
Bishop EL, Gudgeon N, Dimeloe S. Control of T Cell Metabolism by Cytokines and Hormones. Front Immunol. 2021; 12: 653605.
CrossRef
Google scholar
|
[8] |
Acharya N, Madi A, Zhang H, Klapholz M, Escobar G, Dulberg S, et al. Endogenous Glucocorticoid Signaling Regulates CD8(+) T Cell Differentiation and Development of Dysfunction in the Tumor Microenvironment. Immunity. 2020; 53(3): 658-71.e6.
CrossRef
Google scholar
|
[9] |
Scofield VL, Montufar-Solis D. Cheng E, Estes MK, Klein JR. Intestinal TSH production is localized in crypt enterocytes and in villus ‘hotblocks’ and is coupled to IL-7 production: evidence for involvement of TSH during acute enteric virus infection. Immunol Lett. 2005; 99(1): 36-44.
CrossRef
Google scholar
|
[10] |
Liu CR, Miao J, Zhao ZK, Li LY, Liu YM, Zhang YL, et al. Functional human TSHβ splice variant produced by plasma cell may be involved in the immunologic injury of thyroid in the patient with Hashimoto’s thyroiditis. Mol Cell Endocrinol. 2015; 414: 132-42.
CrossRef
Google scholar
|
[11] |
Ellerhorst JA, Sendi-Naderi A. Johnson MK, Cooke CP, Dang SM, Diwan AH. Human melanoma cells express functional receptors for thyroid-stimulating hormone. Endocr Relat Cancer. 2006; 13(4): 1269-77.
CrossRef
Google scholar
|
[12] |
Vastrad B, Vastrad C, Godavarthi A, Chandrashekar R. Molecular mechanisms underlying gliomas and glioblastoma pathogenesis revealed by bioinformatics analysis of microarray data. Med Oncol. 2017; 34(11): 182.
CrossRef
Google scholar
|
[13] |
Kim JW, Lee S, Lui N, Choi H, Mulvihill M, Fang LT, et al. A somatic TSHR mutation in a patient with lung adenocarcinoma with bronchioloalveolar carcinoma, coronary artery disease and severe chronic obstructive pulmonary disease. Oncol Rep. 2012; 28(4): 1225-30.
CrossRef
Google scholar
|
[14] |
Govindaraj V, Yaduvanshi NS, Krishnamachar H, Rao AJ. Expression of thyroid-stimulating hormone receptor, octamer-binding transcriptio. factor 4, and intracisternal A particle-promoted polypeptide in human breast cancer tissues. Horm Mol Biol Clin Investig. 2012; 9(3): 173-8.
CrossRef
Google scholar
|
[15] |
Gyftaki R, Liacos C, Politi E, Liontos M, Saltiki K, Papageorgiou T, et al. Differential transcriptional and protein expression of thyroid-stimulating hormone receptor in ovarian carcinomas. Int J Gynecol Cancer. 2014; 24(5): 851-6.
|
[16] |
Shih YL, Huang YH, Lin KH, Chu YD, Yeh CT. Identification of Functional Thyroid Stimulating Hormone Receptor and TSHR Gene Mutations in Hepatocellular Carcinoma. Anticancer Res. 2018; 38(5): 2793-802.
CrossRef
Google scholar
|
[17] |
Wang J, Klein JR. Hormonal regulation of extrathymic gut T cell development: involvement of thyroid stimulating hormone. Cell Immunol. 1995; 161(2): 299-302.
CrossRef
Google scholar
|
[18] |
Wang J, Whetsell M, Klein JR. Local hormone networks and intestinal T cell homeostasis. Science. 1997; 275(5308): 1937-9.
CrossRef
Google scholar
|
[19] |
Klein JR. The immune system as a regulator of thyroid hormone activity. Exp Biol Med (Maywood). 2006; 231(3): 229-36.
CrossRef
Google scholar
|
[20] |
Wang X, He Q, Shen H, Xia A, Tian W, Yu W, et al. TOX promotes the exhaustion of antitumor CD8(+) T cells by preventing PD1 degradation in hepatocellular carcinoma. J Hepatol. 2019; 71(4): 731-41.
CrossRef
Google scholar
|
[21] |
Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010; 207(10): 2187-94.
CrossRef
Google scholar
|
[22] |
Chu YD, Yeh CT. The Molecular Function and Clinical Role of Thyroid Stimulating Hormone Receptor in Cancer Cells. Cells. 2020; 9(7): 1730.
CrossRef
Google scholar
|
[23] |
Wen AY, Sakamoto KM, Miller LS. The role of the transcription factor CREB in immune function. J Immunol. 2010; 185(11): 6413-9.
CrossRef
Google scholar
|
[24] |
Xu Y, Yan J, Tao Y, Qian X, Zhang C, Yin L, et al. Pituitary hormone α-MSH promotes tumor-induced myelopoiesis and immunosuppression. Science. 2022; 377(6610): 1085-91.
CrossRef
Google scholar
|
[25] |
Sanin DE, Prendergast CT, Mountford AP. IL-10 Production in Macrophages Is Regulated by a TLR-Driven CREB-Mediated Mechanism That Is Linked to Genes Involved in Cell Metabolism. J Immunol. 2015; 195(3): 1218-32.
CrossRef
Google scholar
|
[26] |
Li J, Lin KW, Murray F, Nakajima T, Zhao Y, Perkins DL, et al. Regulation of cytotoxic T lymphocyte antigen 4 by cyclic AMP. Am J Respir Cell Mol Biol. 2013; 48(1): 63-70.
CrossRef
Google scholar
|
[27] |
Alvarez Y, Municio C, Alonso S, Sánchez Crespo M, Fernández N. The induction of IL-10 by zymosan in dendritic cells depends on CREB activation by the coactivators CREB-binding protein and TORC2 and autocrine PGE2. J Immunol. 2009; 183(2): 1471-9.
CrossRef
Google scholar
|
[28] |
Yun SJ, Lee B, Komori K, Lee MJ, Lee BG, Kim K, et al. Regulation of TIM-3 expression in a human T cell line by tumor-conditioned media and cyclic AMP-dependent signaling. Mol Immunol. 2019; 105: 224-32.
CrossRef
Google scholar
|
[29] |
Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, et al. Suppression of Exosomal PD-L1 Induces Systemic Anti-tumor Immunity and Memory. Cell. 2019; 177(2): 414-27.e13.
CrossRef
Google scholar
|
[30] |
Boursi B, Haynes K, Mamtani R, Yang YX. Thyroid dysfunction, thyroid hormone replacement and colorectal cancer risk. J Natl Cancer Inst. 2015; 107(6): djv084.
CrossRef
Google scholar
|
[31] |
Rostkowska O, Spychalski P, Dobrzycka M, Wilczyński M, Łachiński AJ, Obołończyk Ł, et al. Effects of thyroid hormone imbalance on colorectal cancer carcinogenesis and risk -a systematic review. Endokrynol Pol. 2019; 70(2): 190-7.
|
/
〈 |
|
〉 |