1
|
Bhat A, Shah R, Sharma M, Mahajan K, Kumar R. The current status and future trends in immunotoxicogenomics. IMMUNOTOXICOGENOMICS 2025:261-277. [DOI: 10.1016/b978-0-443-18502-1.00013-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
2
|
Liu X, Xi X, Xu S, Chu H, Hu P, Li D, Zhang B, Liu H, Jiang T, Lu Z. Targeting T cell exhaustion: emerging strategies in non-small cell lung cancer. Front Immunol 2024; 15:1507501. [PMID: 39726592 PMCID: PMC11669709 DOI: 10.3389/fimmu.2024.1507501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Lung cancer continues to be a major contributor to cancer-related deaths globally. Recent advances in immunotherapy have introduced promising treatments targeting T cell functionality. Central to the efficacy of these therapies is the role of T cells, which are often rendered dysfunctional due to continuous antigenic stimulation in the tumor microenvironment-a condition referred to as T cell exhaustion. This review addresses the critical challenge of T cell exhaustion in non-small cell lung cancer (NSCLC), offering a detailed examination of its molecular underpinnings and the resultant therapeutic ineffectiveness. We synthesize current knowledge on the drivers of T cell exhaustion, evaluate emerging strategies for its reversal, and explore the potential impact of these insights for enhancing the clinical efficacy of immunotherapies. By consolidating reported clinical trials and preclinical studies, this article highlights innovative approaches to modulate immune responses and improve patient outcomes, thus providing a roadmap for future research and therapeutic development in lung cancer immunotherapy.
Collapse
Affiliation(s)
- Xianqiang Liu
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Xiaowei Xi
- Technical University of Munich (TUM) School of Medicine and Health, Munich, Germany
| | - Shengshan Xu
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Hongyu Chu
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Penghui Hu
- Scientific Research and Education Department, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Dong Li
- Department of Intensive Care Unit and Clinical Experimental Center, Jiangmen Central Hospital, Jiangmen, China
| | - Bin Zhang
- Department of Cardiovascular Disease and Clinical Experimental Center, Jiangmen Central Hospital, Jiangmen, China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hejie Liu
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Tianxiao Jiang
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Zhuming Lu
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| |
Collapse
|
3
|
Wang Y, Torres-García D, Mostert TP, Reinalda L, Van Kasteren SI. A Bioorthogonal Dual Fluorogenic Probe for the Live-Cell Monitoring of Nutrient Uptake by Mammalian Cells. Angew Chem Int Ed Engl 2024; 63:e202401733. [PMID: 38716701 DOI: 10.1002/anie.202401733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Indexed: 06/21/2024]
Abstract
Cells rely heavily on the uptake of exogenous nutrients for survival, growth, and differentiation. Yet quantifying the uptake of small molecule nutrients at the single cell level is difficult. Here we present a new approach to studying the nutrient uptake in live single cells using Inverse Electron-Demand Diels Alder (IEDDA) chemistry. We have modified carboxyfluorescein-diacetate-succinimidyl esters (CFSE)-a quenched fluorophore that can covalently react with proteins and is only turned on in the cytosol of a cell following esterase activity-with a tetrazine. This tetrazine serves as a second quencher for the pendant fluorophore. Upon reaction with nutrients modified with an electron-rich or strained dienophile in an IEDDA reaction, this quenching group is destroyed, thereby enabling the probe to fluoresce. This has allowed us to monitor the uptake of a variety of dienophile-containing nutrients in live primary immune cell populations using flow cytometry and live-cell microscopy.
Collapse
Affiliation(s)
- Yixuan Wang
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Diana Torres-García
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Thijmen P Mostert
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Luuk Reinalda
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Sander I Van Kasteren
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| |
Collapse
|
4
|
Yang T, Zhao G, Zhu W, Yu W, Jiang Y, Zhou Y, Li Y. Effect on the splenocyte function of weaned piglets induced by continuous lipopolysaccharide injections. J Vet Res 2024; 68:295-302. [PMID: 38947147 PMCID: PMC11210365 DOI: 10.2478/jvetres-2024-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 04/22/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction When piglets are exposed to pathogens for a long period, the immune system organs, among them the spleen, play a major role in combating the stress caused by those pathogens. In the present study, the effect on splenocyte function was investigated in a model of weaned piglets in which stress was induced by multiple low doses of lipopolysaccharide (LPS). Material and Methods Forty-eight 28-day-old piglets were divided into two groups: the LPS group and the control group. During the experimental period of thirteen days, the LPS group was intraperitoneally injected with LPS (100 μg/kg) once per day, and the control group was injected with the same volume of 0.9% sterile saline. On the 1st, 5th, 9th and 13th days, the piglets' spleens were collected for isolating splenocytes. The proliferation ability of splenocytes was evaluated by the cell-counting-kit 8 method. Flow cytometry was used to detect cell cycle stage and apoptosis, and the nitric oxide level of cell supernatant was also tested. Results In the experimental group, the proliferation ability of splenocytes was enhanced, the proportion of cells in the G0/G1 phase was smaller, and cells were promoted to the S and G2/M phases. Meanwhile, apoptosis was suppressed and nitric oxide release upregulated. The results were significantly different between the LPS group and the control group on the 5th and 9th days. Conclusion The difference between the results of one group and those of the other suggest that after the 5th LPS injection, multiple low doses of LPS activated splenocytes and restored the number of splenocytes, which maintained and possibly enhanced the regulation of the immune function of the spleen.
Collapse
Affiliation(s)
- Tingyu Yang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang330045, Jiangxi, China
| | - Guotong Zhao
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang330045, Jiangxi, China
| | - Wenlu Zhu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang330045, Jiangxi, China
| | - Wanting Yu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang330045, Jiangxi, China
| | - Yijie Jiang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang330045, Jiangxi, China
| | - Yunxiao Zhou
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang330045, Jiangxi, China
| | - Yong Li
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang330045, Jiangxi, China
| |
Collapse
|
5
|
Barberis M, Rojas López A. Metabolic imbalance driving immune cell phenotype switching in autoimmune disorders: Tipping the balance of T- and B-cell interactions. Clin Transl Med 2024; 14:e1626. [PMID: 38500390 PMCID: PMC10948951 DOI: 10.1002/ctm2.1626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/18/2024] [Accepted: 02/25/2024] [Indexed: 03/20/2024] Open
Abstract
The interplay between the immune system and the metabolic state of a cell is intricate. In all phases of an immune response, the corresponding metabolic changes shall occur to support its modulation, in addition to the signalling through the cytokine environment and immune receptor stimulation. While autoimmune disorders may develop because of a metabolic imbalance that modulates switching between T-cell phenotypes, the effects that the interaction between T and B cells have on one another's cellular metabolism are yet to be understood in disease context. Here, we propose a perspective which highlights the potential of targeting metabolism to modulate T- and B-cell subtypes populations as well as T-B and B-T cell interactions to successfully treat autoimmune disorders. Specifically, we envision how metabolic changes can tip the balance of immune cells interactions, through definite mechanisms in both health and disease, to explain phenotype switches of B and T cells. Within this scenario, we highlight targeting metabolism that link inflammation, immunometabolism, epigenetics and ageing, is critical to understand inflammatory disorders. The combination of treatments targeting immune cells that cause (T/B) cell phenotype imbalances, and the metabolic pathways involved, may increase the effectiveness of treatment of autoimmune disorders, and/or ameliorate their symptoms to improve patients' quality of life.
Collapse
Affiliation(s)
- Matteo Barberis
- Molecular Systems BiologySchool of BiosciencesFaculty of Health and Medical SciencesUniversity of SurreyGuildfordSurreyUK
- Centre for Mathematical and Computational Biology, CMCBUniversity of SurreyGuildfordSurreyUK
- Synthetic Systems Biology and Nuclear OrganizationSwammerdam Institute for Life SciencesUniversity of AmsterdamAmsterdamThe Netherlands
| | - Alejandra Rojas López
- Molecular Systems BiologySchool of BiosciencesFaculty of Health and Medical SciencesUniversity of SurreyGuildfordSurreyUK
- Centre for Mathematical and Computational Biology, CMCBUniversity of SurreyGuildfordSurreyUK
| |
Collapse
|
6
|
Wang Y, Mei X, Lin Z, Yang X, Cao J, Zhong J, Wang J, Cheng L, Wang Z. Virus infection pattern imprinted and diversified the differentiation of T-cell memory in transcription and function. Front Immunol 2024; 14:1334597. [PMID: 38264657 PMCID: PMC10803622 DOI: 10.3389/fimmu.2023.1334597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/14/2023] [Indexed: 01/25/2024] Open
Abstract
Introduction Memory T (Tm) cells are a subpopulation of immune cells with great heterogeneity. Part of this diversity came from T cells that were primed with different viruses. Understanding the differences among different viral-specific Tms will help develop new therapeutic strategies for viral infections. Methods In this study, we compared the transcriptome of Tm cells that primed with CMV, EBV and SARS-CoV-2 with single-cell sequencing and studied the similarities and differences in terms of subpopulation composition, activation, metabolism and transcriptional regulation. Results We found that CMV is marked by plentiful cytotoxic Temra cells, while EBV is more abundant in functional Tem cells. More importantly, we found that CD28 and CTLA4 can be used as continuous indicators to interrogate the antiviral ability of T cells. Furthermore, we proposed that REL is a main regulatory factor for CMV-specific T cells producing cytokines and plays an antiviral role. Discussion Our data gives deep insight into molecular characteristics of Tm subsets from different viral infection, which is important to understand T cell immunization. Furthermore, our results provide basic background knowledges for T cell based vaccine development in future.
Collapse
Affiliation(s)
- Yuan Wang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| | - Xinyue Mei
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhengfang Lin
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoyun Yang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| | - Jinpeng Cao
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| | - Jiaying Zhong
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junxiang Wang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Li Cheng
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhongfang Wang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Wei P, Kou W, Riaz F, Zhang K, Fu J, Pan F. Combination therapy of HIFα inhibitors and Treg depletion strengthen the anti-tumor immunity in mice. Eur J Immunol 2023; 53:e2250182. [PMID: 37615189 DOI: 10.1002/eji.202250182] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 07/12/2023] [Accepted: 08/22/2023] [Indexed: 08/25/2023]
Abstract
Hypoxia-inducible factor 1 alpha (HIF1α), under hypoxic conditions, is known to play an oxygen sensor stabilizing role by exerting context- and cell-dependent stimulatory and inhibitory functions in immune cells. Nevertheless, how HIF1α regulates T cell differentiation and functions in tumor settings has not been elucidated. Herein, we demonstrated that T-cell-specific deletion of HIF1α improves the inflammatory potential and memory phenotype of CD8+ T cells. We validated that T cell-specific HIF1α ablation reduced the B16 melanomas development with the indication of ameliorated antitumor immune response with enhanced IFN-γ+ CD8+ T cells despite the increase in the Foxp3+ regulatory T-cell population. This was further verified by treating tumor-bearing mice with a HIF1α inhibitor. Results indicated that HIF1α inhibitor also recapitulates HIF1α ablation effects by declining tumor growth and enhancing the memory and inflammatory potential of CD8+ T cells. Furthermore, a combination of Treg inhibitor with HIF1α inhibitor can substantially reduce tumor size. Collectively, these findings highlight the notable roles of HIF1α in distinct CD8+ T-cell subsets. This study suggests the significant implications for enhancing the potential of T cell-based antitumor immunity by combining HIF1α and Tregs inhibitors.
Collapse
Affiliation(s)
- Ping Wei
- Department of Otolaryngology, Ministry of Education Key Laboratory of Child Development, and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Kou
- Department of Otolaryngology, Ministry of Education Key Laboratory of Child Development, and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Farooq Riaz
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, P. R. China
| | - Kaimin Zhang
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, P. R. China
| | - Juan Fu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fan Pan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, P. R. China
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
Gressler AE, Leng H, Zinecker H, Simon AK. Proteostasis in T cell aging. Semin Immunol 2023; 70:101838. [PMID: 37708826 PMCID: PMC10804938 DOI: 10.1016/j.smim.2023.101838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023]
Abstract
Aging leads to a decline in immune cell function, which leaves the organism vulnerable to infections and age-related multimorbidities. One major player of the adaptive immune response are T cells, and recent studies argue for a major role of disturbed proteostasis contributing to reduced function of these cells upon aging. Proteostasis refers to the state of a healthy, balanced proteome in the cell and is influenced by synthesis (translation), maintenance and quality control of proteins, as well as degradation of damaged or unwanted proteins by the proteasome, autophagy, lysosome and cytoplasmic enzymes. This review focuses on molecular processes impacting on proteostasis in T cells, and specifically functional or quantitative changes of each of these upon aging. Importantly, we describe the biological consequences of compromised proteostasis in T cells, which range from impaired T cell activation and function to enhancement of inflamm-aging by aged T cells. Finally, approaches to improve proteostasis and thus rejuvenate aged T cells through pharmacological or physical interventions are discussed.
Collapse
Affiliation(s)
- A Elisabeth Gressler
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Houfu Leng
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Heidi Zinecker
- Ascenion GmbH, Am Zirkus 1, Bertold-Brecht-Platz 3, 10117 Berlin, Germany
| | - Anna Katharina Simon
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom.
| |
Collapse
|
9
|
Panahi Meymandi AR, Akbari B, Soltantoyeh T, Hadjati J, Klionsky DJ, Badie B, Mirzaei HR. Crosstalk between autophagy and metabolic regulation of (CAR) T cells: therapeutic implications. Front Immunol 2023; 14:1212695. [PMID: 37675121 PMCID: PMC10477670 DOI: 10.3389/fimmu.2023.1212695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/04/2023] [Indexed: 09/08/2023] Open
Abstract
Despite chimeric antigen receptor (CAR) T cell therapy's extraordinary success in subsets of B-cell lymphoma and leukemia, various barriers restrict its application in solid tumors. This has prompted investigating new approaches for producing CAR T cells with superior therapeutic potential. Emerging insights into the barriers to CAR T cell clinical success indicate that autophagy shapes the immune response via reprogramming cellular metabolism and vice versa. Autophagy, a self-cannibalization process that includes destroying and recycling intracellular components in the lysosome, influences T cell biology, including development, survival, memory formation, and cellular metabolism. In this review, we will emphasize the critical role of autophagy in regulating and rewiring metabolic circuits in CAR T cells, as well as how the metabolic status of CAR T cells and the tumor microenvironment (TME) alter autophagy regulation in CAR T cells to restore functional competence in CAR Ts traversing solid TMEs.
Collapse
Affiliation(s)
- Ahmad Reza Panahi Meymandi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnia Akbari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahereh Soltantoyeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamshid Hadjati
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Behnam Badie
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, United States
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
10
|
Lee S, Kim S, Kim SD, Oh SJ, Kong SK, Lee HM, Kim S, Choi SW. Differences in the metabolomic profile of the human palatine tonsil between pediatrics and adults. PLoS One 2023; 18:e0288871. [PMID: 37523386 PMCID: PMC10389742 DOI: 10.1371/journal.pone.0288871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 07/05/2023] [Indexed: 08/02/2023] Open
Abstract
Palatine tonsils (PT) are B cell-predominant lymphoid organs that provide primary immune responses to airborne and dietary pathogens. Numerous histopathological and immunological studies have been conducted on PT, yet no investigations have been conducted on its metabolic profile. We performed high-resolution magic angle spinning nuclear magnetic resonance spectroscopy-based metabolic profiling in 35 pediatric and 28 adult human palatine tonsillar tissue samples. A total of 36 metabolites were identified, and the levels of 10 metabolites were significantly different depending on age. Among them, partial correlation analysis shows that glucose levels increased with age, whereas glycine, phosphocholine, phosphoethanolamine, and ascorbate levels decreased with age. We confirmed the decrease in immunometabolic activity in adults through metabolomic analysis, which had been anticipated from previous histological and immunological studies on the PT. These results improve our understanding of metabolic changes in the PT with aging and serve as a basis for future tonsil-related metabolomic studies.
Collapse
Affiliation(s)
- Seokhwan Lee
- Department of Otorhinolaryngology, Inje University Haeundae Paik Hospital, Busan, Republic of Korea
| | - Seonghye Kim
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan, Republic of Korea
| | - Sung-Dong Kim
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Se-Joon Oh
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Soo-Keun Kong
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Hyun-Min Lee
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Suhkmann Kim
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan, Republic of Korea
| | - Sung-Won Choi
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| |
Collapse
|
11
|
Ganjoo S, Gupta P, Corbali HI, Nanez S, Riad TS, Duong LK, Barsoumian HB, Masrorpour F, Jiang H, Welsh JW, Cortez MA. The role of tumor metabolism in modulating T-Cell activity and in optimizing immunotherapy. Front Immunol 2023; 14:1172931. [PMID: 37180129 PMCID: PMC10169689 DOI: 10.3389/fimmu.2023.1172931] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
Immunotherapy has revolutionized cancer treatment and revitalized efforts to harness the power of the immune system to combat a variety of cancer types more effectively. However, low clinical response rates and differences in outcomes due to variations in the immune landscape among patients with cancer continue to be major limitations to immunotherapy. Recent efforts to improve responses to immunotherapy have focused on targeting cellular metabolism, as the metabolic characteristics of cancer cells can directly influence the activity and metabolism of immune cells, particularly T cells. Although the metabolic pathways of various cancer cells and T cells have been extensively reviewed, the intersections among these pathways, and their potential use as targets for improving responses to immune-checkpoint blockade therapies, are not completely understood. This review focuses on the interplay between tumor metabolites and T-cell dysfunction as well as the relationship between several T-cell metabolic patterns and T-cell activity/function in tumor immunology. Understanding these relationships could offer new avenues for improving responses to immunotherapy on a metabolic basis.
Collapse
Affiliation(s)
- Shonik Ganjoo
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Priti Gupta
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Halil Ibrahim Corbali
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Selene Nanez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Thomas S. Riad
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lisa K. Duong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B. Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Fatemeh Masrorpour
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hong Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James W. Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
12
|
Liu S, Liao S, Liang L, Deng J, Zhou Y. The relationship between CD4 + T cell glycolysis and their functions. Trends Endocrinol Metab 2023; 34:345-360. [PMID: 37061430 DOI: 10.1016/j.tem.2023.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 04/17/2023]
Abstract
CD4+ T cells are effector T cells (Teffs) produced by the differentiation of initial T cells in peripheral lymphoid tissue after being attacked by antigens, and have an indispensable role in the development and activation of B cells and CD8+ T cells to regulate and assist immunity. In this review, we provide a new perspective on the relationship between CD4+ T cell glycolysis and its function. We summarize the effects of changes in the glycolysis level of CD4+ T cells on their activation, differentiation, proliferation, and survival. In addition, we emphasize that regulation of the glycolysis level of CD4+ T cells changes their inflammatory phenotypes and function. The study of immune metabolism has received more attention recently, but more work is needed to answer many open questions.
Collapse
Affiliation(s)
- Siyi Liu
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Shan Liao
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Lin Liang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Jun Deng
- Department of Early Clinical Trial Center, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, Hunan 410013, China.
| | - Yanhong Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
13
|
Pei W, Kuchroo VK. tRNA-m 1A modification: a translational checkpoint for T cell expansion. Cell Res 2023; 33:271-272. [PMID: 36550269 PMCID: PMC10066370 DOI: 10.1038/s41422-022-00764-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Weike Pei
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
14
|
Beumer-Chuwonpad A, van Alphen FPJ, Kragten NAM, Freen-van Heeren JJ, Rodriguez Gomez M, Verhoeven AJ, van den Biggelaar M, van Gisbergen KPJM. Memory CD8 + T cells upregulate glycolysis and effector functions under limiting oxygen conditions. Eur J Immunol 2023; 53:e2249918. [PMID: 36482267 PMCID: PMC10108084 DOI: 10.1002/eji.202249918] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 11/01/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
Memory CD8+ T cells are indispensable for maintaining long-term immunity against intracellular pathogens and tumors. Despite their presence at oxygen-deprived infected tissue sites or in tumors, the impact of local oxygen pressure on memory CD8+ T cells remains largely unclear. We sought to elucidate how oxygen pressure impacts memory CD8+ T cells arising after infection with Listeria monocytogenes-OVA. Our data revealed that reduced oxygen pressure during in vitro culture switched CD8+ T cell metabolism from oxidative phosphorylation to a glycolytic phenotype. Quantitative proteomic analysis showed that limiting oxygen conditions increased the expression of glucose transporters and components of the glycolytic pathway, while decreasing TCA cycle and mitochondrial respiratory chain proteins. The altered CD8+ T cell metabolism did not affect the expansion potential, but enhanced the granzyme B and IFN-γ production capacity. In vivo, memory CD8+ T cells cultured under low oxygen pressure provided protection against bacterial rechallenge. Taken together, our study indicates that strategies of cellular immune therapy may benefit from reducing oxygen during culture to develop memory CD8+ T cells with superior effector functions.
Collapse
Affiliation(s)
- Ammarina Beumer-Chuwonpad
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Floris P J van Alphen
- Department of Research Facilities, Sanquin Research and Laboratory Services, Amsterdam, The Netherlands
| | - Natasja A M Kragten
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Julian J Freen-van Heeren
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Rodriguez Gomez
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Arthur J Verhoeven
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Huang Y, Jia A, Wang Y, Liu G. CD8 + T cell exhaustion in anti-tumour immunity: The new insights for cancer immunotherapy. Immunology 2023; 168:30-48. [PMID: 36190809 DOI: 10.1111/imm.13588] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/29/2022] [Indexed: 12/27/2022] Open
Abstract
CD8+ T cells play a crucial role in anti-tumour immunity, but they often undergo exhaustion, which affects the anti-tumour activity of CD8+ T cells. The effect and mechanism of exhausted CD8+ T cells have become the focus of anti-tumour immunity research. Recently, a large number of studies have confirmed that long-term antigen exposure can induce exhaustion. Cytokines previously have identified their effects (such as IL-2 and IL-10) may play a dual role in the exhaustion process of CD8+ T cells, suggesting a new mechanism of inducing exhaustion. This review just focuses our current understanding of the biology of exhausted CD8+ T cells, including differentiation pathways, cellular characteristics and signalling pathways involved in inducing exhaustion, and summarizes how these can be applied to tumour immunotherapy.
Collapse
Affiliation(s)
- Yijin Huang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Anna Jia
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yufei Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
16
|
Yu L, Ding L, Wang ZY, Zhao XZ, Wang YH, Liang C, Li J. Hybrid Metabolic Activity-Related Prognostic Model and Its Effect on Tumor in Renal Cell Carcinoma. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:1147545. [PMID: 36591111 PMCID: PMC9797315 DOI: 10.1155/2022/1147545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
Background Tumor cells with a hybrid metabolic state, in which glycolysis and oxidative phosphorylation (OXPHOS) can be used, usually have a strong ability to adapt to different stress environments due to their metabolic plasticity. However, few studies on tumor cells with this phenotype have been conducted in the field of renal cell carcinoma (RCC). Methods The metabolic pathway (glycolysis, OXPHOS) related gene sets were obtained from the Molecular Signatures Database (V7.5.1). The gene expression matrix, clinical information, and mutation data were obtained by Perl programming language (5.32.0) mining, the Cancer Genome Atlas and International Cancer Genome Consortium database. Gene Set Enrichment Analysis (GSEA) software (4.0.3) was utilised to analyse glycolysis-related gene sets. Analysis of survival, immune infiltration, mutation, etc. was performed using the R programming language (4.1.0). Results Eight genes that are highly associated with glycolysis and OXHPOS were used to construct the cox proportional hazards model, and risk scores were calculated based on this to predict the prognosis of clear cell RCC patients and to classify patients into risk groups. Gene Ontology, the Kyoto Encyclopaedia of Genes and Genomes, and GSEA were analysed according to the differential genes to investigate the signal pathways related to the hybrid metabolic state. Immunoinfiltration analysis revealed that CD8+T cells, M2 macrophages, etc., had significant differences in infiltration. In addition, the analysis of mutation data showed significant differences in the number of mutations of PBRM1, SETD2, and BAP1 between groups. Cell experiments demonstrated that the DLD gene expression was abnormally high in various tumor cells and is associated with the strong migration ability of RCC. Conclusions We successfully constructed a risk score system based on glycolysis and OXPHOS-related genes to predict the prognosis of RCC patients. Bioinformatics analysis and cell experiments also revealed the effect of the hybrid metabolic activity on the migration ability and immune activity of RCC and the possible therapeutic targets for patients.
Collapse
Affiliation(s)
- Lei Yu
- Department of Urology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Ding
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhong-Yuan Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xing-Zhi Zhao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Hao Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chao Liang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Li
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
Liu Y, Zhou J, Li X, Zhang X, Shi J, Wang X, Li H, Miao S, Chen H, He X, Dong L, Lee GR, Zheng J, Liu RJ, Su B, Ye Y, Flavell RA, Yi C, Wu Y, Li HB. tRNA-m 1A modification promotes T cell expansion via efficient MYC protein synthesis. Nat Immunol 2022; 23:1433-1444. [PMID: 36138184 DOI: 10.1038/s41590-022-01301-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 08/04/2022] [Indexed: 02/04/2023]
Abstract
Naive T cells undergo radical changes during the transition from dormant to hyperactive states upon activation, which necessitates de novo protein production via transcription and translation. However, the mechanism whereby T cells globally promote translation remains largely unknown. Here, we show that on exit from quiescence, T cells upregulate transfer RNA (tRNA) m1A58 'writer' proteins TRMT61A and TRMT6, which confer m1A58 RNA modification on a specific subset of early expressed tRNAs. These m1A-modified early tRNAs enhance translation efficiency, enabling rapid and necessary synthesis of MYC and of a specific group of key functional proteins. The MYC protein then guides the exit of naive T cells from a quiescent state into a proliferative state and promotes rapid T cell expansion after activation. Conditional deletion of the Trmt61a gene in mouse CD4+ T cells causes MYC protein deficiency and cell cycle arrest, disrupts T cell expansion upon cognate antigen stimulation and alleviates colitis in a mouse adoptive transfer colitis model. Our study elucidates for the first time, to our knowledge, the in vivo physiological roles of tRNA-m1A58 modification in T cell-mediated pathogenesis and reveals a new mechanism of tRNA-m1A58-controlled T cell homeostasis and signal-dependent translational control of specific key proteins.
Collapse
Affiliation(s)
- Yongbo Liu
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Zhou
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaoyu Li
- Department of Biochemistry and Department of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoting Zhang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Jintong Shi
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuefei Wang
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shan Miao
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huifang Chen
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Chongqing International Institute for Immunology, Chongqing, China
| | - Xiaoxiao He
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liting Dong
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Gap Ryol Lee
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ru-Juan Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Bing Su
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youqiong Ye
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA. .,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA.
| | - Chengqi Yi
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China. .,Department of Chemical Biology and Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
| | - Yuzhang Wu
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Hua-Bing Li
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Chongqing International Institute for Immunology, Chongqing, China. .,Department of Geriatrics, Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Chye A, Allen I, Barnet M, Burnett DL. Insights Into the Host Contribution of Endocrine Associated Immune-Related Adverse Events to Immune Checkpoint Inhibition Therapy. Front Oncol 2022; 12:894015. [PMID: 35912205 PMCID: PMC9329613 DOI: 10.3389/fonc.2022.894015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Blockade of immune checkpoints transformed the paradigm of systemic cancer therapy, enabling substitution of a cytotoxic chemotherapy backbone to one of immunostimulation in many settings. Invigorating host immune cells against tumor neo-antigens, however, can induce severe autoimmune toxicity which in many cases requires ongoing management. Many immune-related adverse events (irAEs) are clinically and pathologically indistinguishable from inborn errors of immunity arising from genetic polymorphisms of immune checkpoint genes, suggesting a possible shared driver for both conditions. Many endocrine irAEs, for example, have analogous primary genetic conditions with varied penetrance and severity despite consistent genetic change. This is akin to onset of irAEs in response to immune checkpoint inhibitors (ICIs), which vary in timing, severity and nature despite a consistent drug target. Host contribution to ICI response and irAEs, particularly those of endocrine origin, such as thyroiditis, hypophysitis, adrenalitis and diabetes mellitus, remains poorly defined. Improved understanding of host factors contributing to ICI outcomes is essential for tailoring care to an individual’s unique genetic predisposition to response and toxicity, and are discussed in detail in this review.
Collapse
Affiliation(s)
- Adrian Chye
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
| | - India Allen
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
| | - Megan Barnet
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
- *Correspondence: Megan Barnet, ; Deborah L. Burnett,
| | - Deborah L. Burnett
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
- *Correspondence: Megan Barnet, ; Deborah L. Burnett,
| |
Collapse
|
19
|
Daniel CJ, Pelz C, Wang X, Munks MW, Ko A, Murugan D, Byers SA, Juarez E, Taylor KL, Fan G, Coussens LM, Link JM, Sears RC. T-cell Dysfunction upon Expression of MYC with Altered Phosphorylation at Threonine 58 and Serine 62. Mol Cancer Res 2022; 20:1151-1165. [PMID: 35380701 PMCID: PMC9262837 DOI: 10.1158/1541-7786.mcr-21-0560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 03/01/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022]
Abstract
As a transcription factor that promotes cell growth, proliferation, and apoptosis, c-MYC (MYC) expression in the cell is tightly controlled. Disruption of oncogenic signaling pathways in human cancers can increase MYC protein stability, due to altered phosphorylation ratios at two highly conserved sites, Threonine 58 (T58) and Serine 62 (S62). The T58 to Alanine mutant (T58A) of MYC mimics the stabilized, S62 phosphorylated, and highly oncogenic form of MYC. The S62A mutant is also stabilized, lacks phosphorylation at both Serine 62 and Threonine 58, and has been shown to be nontransforming in vitro. However, several regulatory proteins are reported to associate with MYC lacking phosphorylation at S62 and T58, and the role this form of MYC plays in MYC transcriptional output and in vivo oncogenic function is understudied. We generated conditional c-Myc knock-in mice in which the expression of wild-type MYC (MYCWT), the T58A mutant (MYCT58A), or the S62A mutant (MYCS62A) with or without expression of endogenous Myc is controlled by the T-cell-specific Lck-Cre recombinase. MYCT58A expressing mice developed clonal T-cell lymphomas with 100% penetrance and conditional knock-out of endogenous Myc accelerated this lymphomagenesis. In contrast, MYCS62A mice developed clonal T-cell lymphomas at a much lower penetrance, and the loss of endogenous MYC reduced the penetrance while increasing the appearance of a non-transgene driven B-cell lymphoma with splenomegaly. Together, our study highlights the importance of regulated phosphorylation of MYC at T58 and S62 for T-cell transformation. IMPLICATIONS Dysregulation of phosphorylation at conserved T58 and S62 residues of MYC differentially affects T-cell development and lymphomagenesis.
Collapse
Affiliation(s)
- Colin J. Daniel
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Carl Pelz
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Xiaoyan Wang
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Michael W. Munks
- Department of Molecular Microbiology and Immunology, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Aaron Ko
- Department of Molecular Microbiology and Immunology, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Dhaarini Murugan
- Department of Cell, Developmental and Cancer Biology, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Sarah A. Byers
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Eleonora Juarez
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Karyn L. Taylor
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Guang Fan
- Department of Pathology, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Lisa M. Coussens
- Department of Cell, Developmental and Cancer Biology, School of Medicine, Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Jason M. Link
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
20
|
Chen X, Sunkel B, Wang M, Kang S, Wang T, Gnanaprakasam JNR, Liu L, Cassel TA, Scott DA, Muñoz-Cabello AM, Lopez-Barneo J, Yang J, Lane AN, Xin G, Stanton B, Fan TWM, Wang R. Succinate dehydrogenase/complex II is critical for metabolic and epigenetic regulation of T cell proliferation and inflammation. Sci Immunol 2022; 7:eabm8161. [PMID: 35486677 PMCID: PMC9332111 DOI: 10.1126/sciimmunol.abm8161] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Effective T cell-mediated immune responses require the proper allocation of metabolic resources to sustain growth, proliferation, and cytokine production. Epigenetic control of the genome also governs T cell transcriptome and T cell lineage commitment and maintenance. Cellular metabolic programs interact with epigenetic regulation by providing substrates for covalent modifications of chromatin. By using complementary genetic, epigenetic, and metabolic approaches, we revealed that tricarboxylic acid (TCA) cycle flux fueled biosynthetic processes while controlling the ratio of succinate/α-ketoglutarate (α-KG) to modulate the activities of dioxygenases that are critical for driving T cell inflammation. In contrast to cancer cells, where succinate dehydrogenase (SDH)/complex II inactivation drives cell transformation and growth, SDH/complex II deficiency in T cells caused proliferation and survival defects when the TCA cycle was truncated, blocking carbon flux to support nucleoside biosynthesis. Replenishing the intracellular nucleoside pool partially relieved the dependence of T cells on SDH/complex II for proliferation and survival. SDH deficiency induced a proinflammatory gene signature in T cells and promoted T helper 1 and T helper 17 lineage differentiation. An increasing succinate/α-KG ratio in SDH-deficient T cells promoted inflammation by changing the pattern of the transcriptional and chromatin accessibility signatures and consequentially increasing the expression of the transcription factor, PR domain zinc finger protein 1. Collectively, our studies revealed a role of SDH/complex II in allocating carbon resources for anabolic processes and epigenetic regulation in T cell proliferation and inflammation.
Collapse
Affiliation(s)
- Xuyong Chen
- Center for Childhood Cancer & Blood Diseases, Hematology/Oncology & BMT, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Benjamin Sunkel
- Center for Childhood Cancer & Blood Diseases, Hematology/Oncology & BMT, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Meng Wang
- Center for Childhood Cancer & Blood Diseases, Hematology/Oncology & BMT, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Siwen Kang
- Center for Childhood Cancer & Blood Diseases, Hematology/Oncology & BMT, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Tingting Wang
- Center for Childhood Cancer & Blood Diseases, Hematology/Oncology & BMT, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - JN Rashida Gnanaprakasam
- Center for Childhood Cancer & Blood Diseases, Hematology/Oncology & BMT, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Lingling Liu
- Center for Childhood Cancer & Blood Diseases, Hematology/Oncology & BMT, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Teresa A. Cassel
- Center for Environmental and Systems Biochemistry, Dept. of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - David A. Scott
- Cancer Metabolism Core, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ana M. Muñoz-Cabello
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario "Virgen del Rocío"/CSIC/Universidad de Sevilla, Spain
| | - Jose Lopez-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario "Virgen del Rocío"/CSIC/Universidad de Sevilla, Spain
| | - Jun Yang
- Department of Surgery, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Andrew N. Lane
- Center for Environmental and Systems Biochemistry, Dept. of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Gang Xin
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Benjamin Stanton
- Center for Childhood Cancer & Blood Diseases, Hematology/Oncology & BMT, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Teresa W.-M. Fan
- Center for Environmental and Systems Biochemistry, Dept. of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Ruoning Wang
- Center for Childhood Cancer & Blood Diseases, Hematology/Oncology & BMT, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
21
|
Kabiraj P, Grund EM, Clarkson BDS, Johnson RK, LaFrance-Corey RG, Lucchinetti CF, Howe CL. Teriflunomide shifts the astrocytic bioenergetic profile from oxidative metabolism to glycolysis and attenuates TNFα-induced inflammatory responses. Sci Rep 2022; 12:3049. [PMID: 35197552 PMCID: PMC8866412 DOI: 10.1038/s41598-022-07024-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/27/2022] [Indexed: 12/13/2022] Open
Abstract
Astrocytes utilize both glycolytic and mitochondrial pathways to power cellular processes that are vital to maintaining normal CNS functions. These cells also mount inflammatory and acute phase reactive programs in response to diverse stimuli. While the metabolic functions of astrocytes under homeostatic conditions are well-studied, the role of cellular bioenergetics in astrocyte reactivity is poorly understood. Teriflunomide exerts immunomodulatory effects in diseases such as multiple sclerosis by metabolically reprogramming lymphocytes and myeloid cells. We hypothesized that teriflunomide would constrain astrocytic inflammatory responses. Purified murine astrocytes were grown under serum-free conditions to prevent acquisition of a spontaneous reactive state. Stimulation with TNFα activated NFκB and increased secretion of Lcn2. TNFα stimulation increased basal respiration, maximal respiration, and ATP production in astrocytes, as assessed by oxygen consumption rate. TNFα also increased glycolytic reserve and glycolytic capacity of astrocytes but did not change the basal glycolytic rate, as assessed by measuring the extracellular acidification rate. TNFα specifically increased mitochondrial ATP production and secretion of Lcn2 required ATP generated by oxidative phosphorylation. Inhibition of dihydroorotate dehydrogenase via teriflunomide transiently increased both oxidative phosphorylation and glycolysis in quiescent astrocytes, but only the increased glycolytic ATP production was sustained over time, resulting in a bias away from mitochondrial ATP production even at doses down to 1 μM. Preconditioning with teriflunomide prevented the TNFα-induced skew toward oxidative phosphorylation, reduced mitochondrial ATP production, and reduced astrocytic inflammatory responses, suggesting that this drug may limit neuroinflammation by acting as a metabolomodulator.
Collapse
Affiliation(s)
- Parijat Kabiraj
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Translational Neuroimmunology Lab, Mayo Clinic, Guggenheim 1542C, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ethan M Grund
- Translational Neuroimmunology Lab, Mayo Clinic, Guggenheim 1542C, 200 First Street SW, Rochester, MN, 55905, USA
- Mayo Graduate School Neuroscience PhD Program and Medical Scientist Training Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, 55905, USA
| | - Benjamin D S Clarkson
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Translational Neuroimmunology Lab, Mayo Clinic, Guggenheim 1542C, 200 First Street SW, Rochester, MN, 55905, USA
- Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Renee K Johnson
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Translational Neuroimmunology Lab, Mayo Clinic, Guggenheim 1542C, 200 First Street SW, Rochester, MN, 55905, USA
| | - Reghann G LaFrance-Corey
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Translational Neuroimmunology Lab, Mayo Clinic, Guggenheim 1542C, 200 First Street SW, Rochester, MN, 55905, USA
| | - Claudia F Lucchinetti
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Charles L Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA.
- Translational Neuroimmunology Lab, Mayo Clinic, Guggenheim 1542C, 200 First Street SW, Rochester, MN, 55905, USA.
- Division of Experimental Neurology, Mayo Clinic, Rochester, MN, 55905, USA.
- Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
22
|
Zhou L, Fu F, Wang Y, Yang L. Interlocked feedback loops balance the adaptive immune response. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:4084-4100. [PMID: 35341288 DOI: 10.3934/mbe.2022188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Adaptive immune responses can be activated by harmful stimuli. Upon activation, a cascade of biochemical events ensues the proliferation and the differentiation of T cells, which can remove the stimuli and undergo cell death to maintain immune cell homeostasis. However, normal immune processes can be disrupted by certain dysregulations, leading to pathological responses, such as cytokine storms and immune escape. In this paper, a qualitative mathematical model, composed of key feedback loops within the immune system, was developed to study the dynamics of various response behaviors. First, simulation results of the model well reproduce the results of several immune response processes, particularly pathological immune responses. Next, we demonstrated how the interaction of positive and negative feedback loops leads to irreversible bistable, reversible bistable and monostable, which characterize different immune response processes: cytokine storm, normal immune response, immune escape. The stability analyses suggest that the switch-like behavior is the basis of rapid activation of the immune system, and a balance between positive and negative regulation loops is necessary to prevent pathological responses. Furthermore, we have shown how the treatment moves the system back to a healthy state from the pathological immune response. The bistable mechanism that revealed in this work is helpful to understand the dynamics of different immune response processes.
Collapse
Affiliation(s)
- Lingli Zhou
- School of Mathematical Sciences, Soochow University, Suzhou 215006, China
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| | - Fengqing Fu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Yao Wang
- School of Mathematical Sciences, Soochow University, Suzhou 215006, China
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| | - Ling Yang
- School of Mathematical Sciences, Soochow University, Suzhou 215006, China
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| |
Collapse
|
23
|
Han X, Hu S, Yang Q, Sang X, Tang D, Cao G. Paeoniflorin ameliorates airway inflammation and immune response in ovalbumin induced asthmatic mice: From oxidative stress to autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 96:153835. [PMID: 34799185 DOI: 10.1016/j.phymed.2021.153835] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/14/2021] [Accepted: 10/27/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Asthma characterized by airway remodeling is a multiple pulmonary disease, which is associated with various physiological processes including inflammation reaction, immune response, oxidative stress and autophagy. PURPOSE This study aimed to investigate whether these processes are modulated by the total glucosides of Paeonia lactiflora Pall (TGP), and its active compound paeoniflorin (PF) with anti-inflammatory and immune-regulatory effects could alleviate ovalbumin (OVA)-induced mouse asthma. METHODS In vivo, models of mouse asthma were established by intraperitoneally with a mixture of OVA and aluminum hydroxide, plus a single nasal injected with OVA to female C57BL/6 mice. The results were observed with PET imaging, TEM, RT-PCR, western blotting. In vitro, CD4+ T cells were isolated and detected with flow cytometry. RESULTS TGP, either in its crude or processed form, and PF effectively ameliorated lung injury in mice induced by OVA, regulated immune/inflammatory response by inhibiting the release of pro-inflammatory cytokines, thereby decreasing Th2 cell proportion, inhibited oxidative stress by recovering mitochondrial membrane potential and regulating metabolic activity in dose-dependent manner. Moreover, PF could inhibit autophagy by regulating mitochondrial function. In addition, the therapeutic effects of TGP and PF on pulmonary injury in asthmatic mice were not affected by processing. CONCLUSION PF may be a valuable agent in ameliorating inflammation and immune response in asthmatic mice, and the possible mechanism involved in this response rang may from oxidative stress to autophagy.
Collapse
Affiliation(s)
- Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shaoqi Hu
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dongxin Tang
- First Affiliated Hospital of Guizhou Universit of Traditional Chinese Medicine (TCM), Guiyang, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
24
|
Aggarwal V, Rathod S, Vashishth K, Upadhyay A. Immune Cell Metabolites as Fuel for Cancer Cells. IMMUNO-ONCOLOGY CROSSTALK AND METABOLISM 2022:153-186. [DOI: 10.1007/978-981-16-6226-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
25
|
Alves RW, da Silva EM, Doretto-Silva L, Andrade-Oliveira V. Metabolic Pathways in Immune Cells Commitment and Fate. ESSENTIAL ASPECTS OF IMMUNOMETABOLISM IN HEALTH AND DISEASE 2022:53-82. [DOI: 10.1007/978-3-030-86684-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Chen D, Tang TX, Deng H, Yang XP, Tang ZH. Interleukin-7 Biology and Its Effects on Immune Cells: Mediator of Generation, Differentiation, Survival, and Homeostasis. Front Immunol 2021; 12:747324. [PMID: 34925323 PMCID: PMC8674869 DOI: 10.3389/fimmu.2021.747324] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
Interleukin-7 (IL-7), a molecule known for its growth-promoting effects on progenitors of B cells, remains one of the most extensively studied cytokines. It plays a vital role in health maintenance and disease prevention, and the congenital deficiency of IL-7 signaling leads to profound immunodeficiency. IL-7 contributes to host defense by regulating the development and homeostasis of immune cells, including T lymphocytes, B lymphocytes, and natural killer (NK) cells. Clinical trials of recombinant IL-7 have demonstrated safety and potent immune reconstitution effects. In this article, we discuss IL-7 and its functions in immune cell development, drawing on a substantial body of knowledge regarding the biology of IL-7. We aim to answer some remaining questions about IL-7, providing insights essential for designing new strategies of immune intervention.
Collapse
Affiliation(s)
- Deng Chen
- Division of Trauma and Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting-Xuan Tang
- Class 1901, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Hai Deng
- Division of Trauma and Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang-Ping Yang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Tang
- Division of Trauma and Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
La Rosa F, Guiducci L, Guzzardi MA, Cacciato Insilla A, Burchielli S, Brunetto MR, Bonino F, Campani D, Iozzo P. Maternal High-Fat Feeding Affects the Liver and Thymus Metabolic Axis in the Offspring and Some Effects Are Attenuated by Maternal Diet Normalization in a Minipig Model. Metabolites 2021; 11:800. [PMID: 34940559 PMCID: PMC8703533 DOI: 10.3390/metabo11120800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 11/30/2022] Open
Abstract
Maternal high-fat diet (HFD) affects metabolic and immune development. We aimed to characterize the effects of maternal HFD, and the subsequent diet-normalization of the mothers during a second pregnancy, on the liver and thymus metabolism in their offspring, in minipigs. Offspring born to high-fat (HFD) and normal diet (ND) fed mothers were studied at week 1 and months 1, 6, 12 of life. Liver and thymus glucose uptake (GU) was measured with positron emission tomography during hyperinsulinemic-isoglycemia. Histological analyses were performed to quantify liver steatosis, inflammation, and hepatic hematopoietic niches (HHN), and thymocyte size and density in a subset. The protocol was repeated after maternal-diet-normalization in the HFD group. At one week, HFDoff were characterized by hyperglycemia, hyperinsulinemia, severe insulin resistance (IR), and high liver and thymus GU, associating with thymocyte size and density, with elevated weight-gain, liver IR, and steatosis in the first 6 months of life. Maternal diet normalization reversed thymus and liver hypermetabolism, and increased HHN at one week. It also normalized systemic insulin-sensitivity and liver fat content at all ages. Instead, weight-gain excess, hyperglycemia, and hepatic IR were still observed at 1 month, i.e., end-lactation. We conclude that intra-uterine HFD exposure leads to time-changing metabolic and immune-correlated abnormalities. Maternal diet-normalization reversed most of the effects in the offspring.
Collapse
Affiliation(s)
- Federica La Rosa
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (L.G.); (M.A.G.)
| | - Letizia Guiducci
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (L.G.); (M.A.G.)
| | - Maria Angela Guzzardi
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (L.G.); (M.A.G.)
| | - Andrea Cacciato Insilla
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, Division of Pathology, Pisa University Hospital, 56124 Pisa, Italy; (A.C.I.); (D.C.)
| | | | - Maurizia Rossana Brunetto
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy;
- Hepatology Unit, Department of Medical Specialties, Laboratory of Molecular Genetics and Pathology of Hepatitis Viruses, Pisa University Hospital, 56124 Pisa, Italy
- Institute of Biostructure and Bioimaging (IBB), National Research Council (CNR), 80145 Napoli, Italy;
| | - Ferruccio Bonino
- Institute of Biostructure and Bioimaging (IBB), National Research Council (CNR), 80145 Napoli, Italy;
| | - Daniela Campani
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, Division of Pathology, Pisa University Hospital, 56124 Pisa, Italy; (A.C.I.); (D.C.)
| | - Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (L.G.); (M.A.G.)
| |
Collapse
|
28
|
Metabolic Disorders in Multiple Myeloma. Int J Mol Sci 2021; 22:ijms222111430. [PMID: 34768861 PMCID: PMC8584036 DOI: 10.3390/ijms222111430] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/08/2021] [Accepted: 10/21/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy and is attributed to monoclonal proliferation of plasma cells in the bone marrow. Cancer cells including myeloma cells deregulate metabolic pathways to ensure proliferation, growth, survival and avoid immune surveillance, with glycolysis and glutaminolysis being the most identified procedures involved. These disorders are considered a hallmark of cancer and the alterations performed ensure that enough energy is available for rapid cell proliferation. An association between metabolic syndrome, inflammatory cytokinesand incidence of MM has been also described, while the use of metformin and statins has been identified as a positive prognostic factor for the disease course. In this review, we aim to present the metabolic disorders that occur in multiple myeloma, the potential defects on the immune system and the potential advantage of targeting the dysregulated pathways in order to enhance antitumor therapeutics.
Collapse
|
29
|
Zhou X, Li Y, Ji Y, Liu T, Zhao N, He J, Yao J. PD-1 Involvement in Peripheral Blood CD8 + T Lymphocyte Dysfunction in Patients with Acute-on-chronic Liver Failure. J Clin Transl Hepatol 2021; 9:283-290. [PMID: 34221914 PMCID: PMC8237147 DOI: 10.14218/jcth.2020.00142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/09/2021] [Accepted: 03/07/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND AIMS Programmed cell death-1 (PD-1) plays an important role in downregulating T lymphocytes but the mechanisms are still poorly understood. This study aimed to explore the role of PD-1 in CD8+ T lymphocyte dysfunction in hepatitis B virus (HBV)-related acute-on-chronic liver failure (ACLF). METHODS Thirty patients with HBV-ACLF and 30 healthy controls (HCs) were recruited. The differences in the numbers and functions of CD8+ T lymphocytes, PD-1 and glucose transporter-1 (Glut1) expression from the peripheral blood of patients with HBV-ACLF and HCs were analyzed. In vitro, the CD8+ T lymphocytes from HCs were cultured (HC group) and the CD8+ T lymphocytes from ACLF patients were cultured with PD-L1-IgG (ACLF+PD-1 group) or IgG (ACLF group). The numbers and functions of CD8+ T lymphocytes, PD-1 expression, glycogen uptake capacity, and Glut1, hexokinase-2 (HK2), and pyruvate kinase (PKM2) expression were analyzed among the HC group, ACLF group and ACLF+ PD-1group. RESULTS The absolute numbers of CD8+ T lymphocytes in the peripheral blood from patients with HBV-ACLF were lower than in the HCs (p<0.001). The expression of PD-1 in peripheral blood CD8+ T lymphocytes was lower in HCs than in patients with HBV-ACLF (p=0.021). Compared with HCs, PD-1 expression was increased (p=0.021) and Glut1 expression was decreased (p=0.016) in CD8+ T lymphocytes from the HBV-ACLF group. In vitro, glycogen uptake and functions of ACLF CD8+ T lymphocytes were significantly lower than that in HCs (p=0.017; all p<0.001). When PD-1/PD-L1 was activated, the glycogen uptake rate and expression levels of Glut1, HK2, and PKM2 showed a decreasing trend (ACLF+PD-1 group compared to ACLF group , all p<0.05). The functions of CD8+ T lymphocytes in the ACLF+PD-1 group [using biomarkers of Ki67, CD69, IL-2, interferon-gamma, and tumor necrosis factor-alpha- were lower than in the ACLF group (all p<0.05). CONCLUSIONS CD8+ T lymphocyte dysfunction is observed in patients with HBV-ACLF. PD-1-induced T lymphocyte dysfunction might involve glycolysis inhibition.
Collapse
Affiliation(s)
- Xiaoshuang Zhou
- Department of Nephrology, Shanxi Provincial People’s Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yidong Li
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaqiu Ji
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tian Liu
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ninghui Zhao
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Correspondence to: Jia Yao and Ninghui Zhao, Department of Gastroenterology, Shanxi Baiqiuen Hospital, Shanxi Medical University, No. 99 Longcheng Street, Taiyuan, Shanxi 030001, China. ORCID: https://orcid.org/0000-0003-2210-7717 (JY), https://orcid.org/0000-0002-9715-9303 (NZ). Tel/Fax: +86-199-3491-1619, E-mail: (JY) and (NZ); Jiefeng He, Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, China. ORCID: https://orcid.org/0000-0003-2958-0232. E-mail:
| | - Jiefeng He
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Correspondence to: Jia Yao and Ninghui Zhao, Department of Gastroenterology, Shanxi Baiqiuen Hospital, Shanxi Medical University, No. 99 Longcheng Street, Taiyuan, Shanxi 030001, China. ORCID: https://orcid.org/0000-0003-2210-7717 (JY), https://orcid.org/0000-0002-9715-9303 (NZ). Tel/Fax: +86-199-3491-1619, E-mail: (JY) and (NZ); Jiefeng He, Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, China. ORCID: https://orcid.org/0000-0003-2958-0232. E-mail:
| | - Jia Yao
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Institute of Liver Disease and Organ Transplantation, Shanxi Medical University, Taiyuan, Shanxi, China
- Correspondence to: Jia Yao and Ninghui Zhao, Department of Gastroenterology, Shanxi Baiqiuen Hospital, Shanxi Medical University, No. 99 Longcheng Street, Taiyuan, Shanxi 030001, China. ORCID: https://orcid.org/0000-0003-2210-7717 (JY), https://orcid.org/0000-0002-9715-9303 (NZ). Tel/Fax: +86-199-3491-1619, E-mail: (JY) and (NZ); Jiefeng He, Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, China. ORCID: https://orcid.org/0000-0003-2958-0232. E-mail:
| |
Collapse
|
30
|
Liu X, Zhao J, Wang H, Wang W, Su X, Liao X, Zhang S, Sun J, Zhang Z. Metabolic Defects of Peripheral T Cells in COVID-19 Patients. THE JOURNAL OF IMMUNOLOGY 2021; 206:2900-2908. [PMID: 34049969 DOI: 10.4049/jimmunol.2100068] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/05/2021] [Indexed: 11/19/2022]
Abstract
The relatively low partial pressure of oxygen, reduced oxygen saturation, and aberrant plasma metabolites in COVID-19 may alter energy metabolism in peripheral immune cells. However, little is known regarding the immunometabolic defects of T cells in COVID-19 patients, which may contribute to the deregulated immune functions of these cells. In this study, we longitudinally characterized the metabolic profiles of resting and activated T cells from acutely infected and convalescent COVID-19 patients by flow cytometry and confirmed the metabolic profiles with a Seahorse analyzer. Non-COVID-19 and healthy subjects were enrolled as controls. We found that ex vivo T cells from acutely infected COVID-19 patients were highly activated and apoptotic and displayed more extensive mitochondrial metabolic dysfunction, especially cells in CD8+ T cell lineages, than those from convalescent COVID-19 patients or healthy controls, but slightly disturbed mitochondrial metabolic activity was observed in non-COVID-19 patients. Importantly, plasma IL-6 and C-reactive protein (CRP) levels positively correlated with mitochondrial mass and negatively correlated with fatty acid uptake in T cells from COVID-19 patients. Additionally, compared with those from healthy controls, in vitro-activated T cells from acutely infected COVID-19 patients showed signs of lower glycolysis, a reduced glycolytic capacity, and a decreased glycolytic reserve, accompanied by lower activation of mTOR signaling. Thus, newly identified defects in T cell mitochondrial metabolic functions and metabolic reprogramming upon activation might contribute to immune deficiency in COVID-19.
Collapse
Affiliation(s)
- Xiaoju Liu
- Department of Infectious Diseases, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, South University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Juanjuan Zhao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, South University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Haiyan Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, South University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Wan Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, South University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Xu Su
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, South University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Xuejiao Liao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, South University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Shuye Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; and
| | - Jian Sun
- Department of Infectious Diseases, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China;
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, South University of Science and Technology, Shenzhen, Guangdong Province, China; .,Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, Guangdong Province, China
| |
Collapse
|
31
|
Wang L, Das JK, Kumar A, Peng HY, Ren Y, Xiong X, Yang JM, Song J. Autophagy in T-cell differentiation, survival and memory. Immunol Cell Biol 2021; 99:351-360. [PMID: 33141986 DOI: 10.1111/imcb.12422] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/23/2020] [Accepted: 10/30/2020] [Indexed: 01/22/2023]
Abstract
Over the past decade, autophagy has emerged as a critical regulatory mechanism of the immune system through critically controlling various aspects of T cell biology and determining the fate of different T cell subsets. Autophagy maintains T cell development and survival by regulating the degradation of organelles and apoptotic proteins. The autophagic process also impacts the formation of memory T cells. Alteration of autophagy in T cells may lead to a variety of pathological conditions such as inflammation, autoimmune diseases and cancer. In this review, we discuss how autophagy impacts T cell differentiation, survival and memory, and its implication in immunotherapy for various diseases.
Collapse
Affiliation(s)
- Liqing Wang
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, USA
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
| | - Jugal Kishore Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Anil Kumar
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Hao-Yun Peng
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, USA
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
| | - Yijie Ren
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Xiaofang Xiong
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Jin-Ming Yang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, USA
| |
Collapse
|
32
|
Wu S, Kuang H, Ke J, Pi M, Yang DH. Metabolic Reprogramming Induces Immune Cell Dysfunction in the Tumor Microenvironment of Multiple Myeloma. Front Oncol 2021; 10:591342. [PMID: 33520703 PMCID: PMC7845572 DOI: 10.3389/fonc.2020.591342] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor cells rewire metabolism to meet their increased nutritional demands, allowing the maintenance of tumor survival, proliferation, and expansion. Enhancement of glycolysis and glutaminolysis is identified in most, if not all cancers, including multiple myeloma (MM), which interacts with a hypoxic, acidic, and nutritionally deficient tumor microenvironment (TME). In this review, we discuss the metabolic changes including generation, depletion or accumulation of metabolites and signaling pathways, as well as their relationship with the TME in MM cells. Moreover, we describe the crosstalk among metabolism, TME, and changing function of immune cells during cancer progression. The overlapping metabolic phenotype between MM and immune cells is discussed. In this sense, targeting metabolism of MM cells is a promising therapeutic approach. We propose that it is important to define the metabolic signatures that may regulate the function of immune cells in TME in order to improve the response to immunotherapy.
Collapse
Affiliation(s)
- Shaojie Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huixian Kuang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Ke
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Medical Center of Assessment of Bone & Joint Diseases, Orthopaedic Hospital, General Hospital of Southern Theater Command, Guangzhou, China
| | - Manfei Pi
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dong-Hua Yang
- College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| |
Collapse
|
33
|
Gaissmaier L, Christopoulos P. Immune Modulation in Lung Cancer: Current Concepts and Future Strategies. Respiration 2020; 99:1-27. [PMID: 33291116 DOI: 10.1159/000510385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer immunotherapy represents the most dynamic field of biomedical research currently, with thoracic immuno-oncology as a forerunner. PD-(L)1 inhibitors are already part of standard first-line treatment for both non-small-cell and small-cell lung cancer, while unprecedented 5-year survival rates of 15-25% have been achieved in pretreated patients with metastatic disease. Evolving strategies are mainly aiming for improvement of T-cell function, increase of immune activation in the tumor microenvironment (TME), and supply of tumor-reactive lymphocytes. Several novel therapeutics have demonstrated preclinical efficacy and are increasingly used in rational combinations within clinical trials. Two overarching trends dominate: extension of immunotherapy to earlier disease stages, mainly as neoadjuvant treatment, and a shift of focus towards multivalent, individualized, mutatome-based antigen-specific modalities, mainly adoptive cell therapies and cancer vaccines. The former ensures ample availability of treated and untreated patient samples, the latter facilitates deeper mechanistic insights, and both in combination build an overwhelming force that is accelerating progress and driving the greatest revolution cancer medicine has seen so far. Today, immune modulation represents the most potent therapeutic modality in oncology, the most important topic in clinical and translational cancer research, and arguably our greatest, meanwhile justified hope for achieving cure of pulmonary neoplasms and other malignancies in the next future.
Collapse
Affiliation(s)
- Lena Gaissmaier
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany,
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Heidelberg, Germany,
| |
Collapse
|
34
|
Jiang Y, Tu X, Zhang X, Liao H, Han S, Jiang W, Zheng Y, Zhao P, Tong Z, Fu Q, Qi Q, Shen J, Zhong L, Pan Y, Fang W. Nutrition and metabolism status alteration in advanced hepatocellular carcinoma patients treated with anti-PD-1 immunotherapy. Support Care Cancer 2020; 28:5569-5579. [PMID: 32361828 DOI: 10.1007/s00520-020-05478-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/17/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE The aim of this study was to evaluate the nutrition and metabolism status alteration during immunotherapy in advanced hepatocellular carcinoma (HCC) patients. METHODS Patients with advanced HCC who participated in the clinical trials of single-agent anti-PD-1 immunotherapy or sorafenib were retrospectively included. We analyzed self-comparison of the nutritional and metabolic indices of patients in the anti-PD-1 and sorafenib treatment group. We conducted mutual-comparison of the mentioned indices between the disease progression group and disease control group among anti-PD-1 treatment patients. We further analyzed those indices with statistical differences by partial correlation and survival analysis. RESULTS Both self-comparison before and after treatment in the anti-PD-1 group and mutual-comparison of disease progression and the control group showed significant differences in multiple indices, but we did not observe significant differences in the sorafenib group. Strikingly, albumin (ALB)/prognostic nutritional index (PNI, calculated by serum albumin and lymphocyte count) decreased distinctly in the immunotherapy disease progression group patients. However, changes in ALB/PNI were not significant in disease progression patients from the sorafenib group or in the disease control patients with immunotherapy. Partial correlation analysis suggested that ALB and PNI were positively correlated with the efficacy of immunotherapy. Furthermore, survival analysis showed that the median progression-free survival and median overall survival of patients in the ALB/PNI decreased group were significantly shorter than those of patients from the ALB/PNI increased group. CONCLUSION Anti-PD-1 immunotherapy might alter the nutritional and metabolic status in advanced HCC patients. We also should pay attention to the nutritional and metabolic status of patients when drug resistance is detected.
Collapse
Affiliation(s)
- Yizhen Jiang
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Xiaoxuan Tu
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China
| | - Xiangying Zhang
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China
| | - Haihong Liao
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Shuwen Han
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Weiqin Jiang
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China
| | - Yi Zheng
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China
| | - Peng Zhao
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China
| | - Zhou Tong
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China
| | - Qihan Fu
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China
| | - Quan Qi
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Junjun Shen
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Liping Zhong
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Yuefen Pan
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, 313000, Zhejiang, People's Republic of China.
| | - Weijia Fang
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
35
|
Koss B, Shields BD, Taylor EM, Storey AJ, Byrum SD, Gies AJ, Washam CL, Choudhury SR, Hyun Ahn J, Uryu H, Williams JB, Krager KJ, Chiang TC, Mackintosh SG, Edmondson RD, Aykin-Burns N, Gajewski TF, Wang GG, Tackett AJ. Epigenetic Control of Cdkn2a.Arf Protects Tumor-Infiltrating Lymphocytes from Metabolic Exhaustion. Cancer Res 2020; 80:4707-4719. [PMID: 33004350 PMCID: PMC7642172 DOI: 10.1158/0008-5472.can-20-0524] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/04/2020] [Accepted: 08/28/2020] [Indexed: 01/06/2023]
Abstract
T-cell exhaustion in cancer is linked to poor clinical outcomes, where evidence suggests T-cell metabolic changes precede functional exhaustion. Direct competition between tumor-infiltrating lymphocytes (TIL) and cancer cells for metabolic resources often renders T cells dysfunctional. Environmental stress produces epigenome remodeling events within TIL resulting from loss of the histone methyltransferase EZH2. Here, we report an epigenetic mechanism contributing to the development of metabolic exhaustion in TIL. A multiomics approach revealed a Cdkn2a.Arf-mediated, p53-independent mechanism by which EZH2 inhibition leads to mitochondrial dysfunction and the resultant exhaustion. Reprogramming T cells to express a gain-of-function EZH2 mutant resulted in an enhanced ability of T cells to inhibit tumor growth in vitro and in vivo. Our data suggest that manipulation of T-cell EZH2 within the context of cellular therapies may yield lymphocytes that are able to withstand harsh tumor metabolic environments and collateral pharmacologic insults. SIGNIFICANCE: These findings demonstrate that manipulation of T-cell EZH2 in cellular therapies may yield cellular products able to withstand solid tumor metabolic-deficient environments. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/21/4707/F1.large.jpg.
Collapse
Affiliation(s)
- Brian Koss
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Bradley D Shields
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Erin M Taylor
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Aaron J Storey
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | - Allen J Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | - Charity L Washam
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | - Samrat Roy Choudhury
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jeong Hyun Ahn
- Lineberger Comprehensive Cancer Center, Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina
| | - Hidetaka Uryu
- Lineberger Comprehensive Cancer Center, Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina
| | - Jason B Williams
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Kimberly J Krager
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Tung-Chin Chiang
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Rick D Edmondson
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Nukhet Aykin-Burns
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Thomas F Gajewski
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Gang Greg Wang
- Lineberger Comprehensive Cancer Center, Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Arkansas Children's Research Institute, Little Rock, Arkansas
| |
Collapse
|
36
|
Balyan R, Gautam N, Gascoigne NR. The Ups and Downs of Metabolism during the Lifespan of a T Cell. Int J Mol Sci 2020; 21:E7972. [PMID: 33120978 PMCID: PMC7663011 DOI: 10.3390/ijms21217972] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/16/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023] Open
Abstract
Understanding the various mechanisms that govern the development, activation, differentiation, and functions of T cells is crucial as it could provide opportunities for therapeutic interventions to disrupt immune pathogenesis. Immunometabolism is one such area that has garnered significant interest in the recent past as it has become apparent that cellular metabolism is highly dynamic and has a tremendous impact on the ability of T cells to grow, activate, and differentiate. In each phase of the lifespan of a T-cell, cellular metabolism has to be tailored to match the specific functional requirements of that phase. Resting T cells rely on energy-efficient oxidative metabolism but rapidly shift to a highly glycolytic metabolism upon activation in order to meet the bioenergetically demanding process of growth and proliferation. However, upon antigen clearance, T cells return to a more quiescent oxidative metabolism to support T cell memory generation. In addition, each helper T cell subset engages distinct metabolic pathways to support their functional needs. In this review, we provide an overview of the metabolic changes that occur during the lifespan of a T cell and discuss several important studies that provide insights into the regulation of the metabolic landscape of T cells and how they impact T cell development and function.
Collapse
Affiliation(s)
| | | | - Nicholas R.J. Gascoigne
- Immunology Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore; (R.B.); (N.G.)
| |
Collapse
|
37
|
Cai Z, Ishibashi T, Kozai M, Mita H, Wang S, Takada K, Inaba M. ROR agonist hampers the proliferation and survival of postactivated CD8 + T cells through the downregulation of cholesterol synthesis-related genes. Immunol Cell Biol 2020; 99:288-298. [PMID: 32940916 DOI: 10.1111/imcb.12406] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/31/2020] [Accepted: 09/15/2020] [Indexed: 11/30/2022]
Abstract
Cholesterol is a major component of the lipid bilayers of cellular membranes. The synthesis of cholesterol is acutely elevated during T-cell activation to support T-cell growth and proliferation. There is a limited understanding of cholesterol metabolism reprogramming during T-cell activation. Retinoic acid receptor-related orphan receptors (RORs) are ligand-activated nuclear receptors that regulate the transcription of target genes. In this study, we demonstrated that the activation of RORs by a synthetic agonist (SR1078) impairs the proliferation and survival of postactivated CD8+ T cells. The inhibitory effects of SR1078 on CD8+ T-cell proliferation and survival were attributed to cholesterol depletion and downregulated expression of cholesterol metabolism-related genes. The overexpression of RORα or RORγt promoted apoptosis in the postactivated CD8+ T cells in vitro. The expression of RORα (but not that of RORγt) was markedly upregulated in the CD8+ T cells upon stimulation with an antigen in vivo. The functional deficiency of RORα enhanced CD8+ T-cell expansion during the response to bacterial infection. These results suggest that RORs are involved in the regulation of CD8+ T-cell-mediated immune response through the regulation of cholesterol metabolism, which can be modulated by a synthetic ROR agonist. The findings of this study can aid in the development of immunotherapeutic methods that target nuclear receptors.
Collapse
Affiliation(s)
- Zimeng Cai
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Taishin Ishibashi
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mina Kozai
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hironobu Mita
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Shangyi Wang
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kensuke Takada
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mutsumi Inaba
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
38
|
Hesterberg RS, Beatty MS, Han Y, Fernandez MR, Akuffo AA, Goodheart WE, Yang C, Chang S, Colin CM, Alontaga AY, McDaniel JM, Mailloux AW, Billington JMR, Yue L, Russell S, Gillies RJ, Yun SY, Ayaz M, Lawrence NJ, Lawrence HR, Yu XZ, Fu J, Darville LN, Koomen JM, Ren X, Messina J, Jiang K, Garrett TJ, Rajadhyaksha AM, Cleveland JL, Epling-Burnette PK. Cereblon harnesses Myc-dependent bioenergetics and activity of CD8+ T lymphocytes. Blood 2020; 136:857-870. [PMID: 32403132 PMCID: PMC7426646 DOI: 10.1182/blood.2019003257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 04/20/2020] [Indexed: 01/08/2023] Open
Abstract
Immunomodulatory drugs, such as thalidomide and related compounds, potentiate T-cell effector functions. Cereblon (CRBN), a substrate receptor of the DDB1-cullin-RING E3 ubiquitin ligase complex, is the only molecular target for this drug class, where drug-induced, ubiquitin-dependent degradation of known "neosubstrates," such as IKAROS, AIOLOS, and CK1α, accounts for their biological activity. Far less clear is whether these CRBN E3 ligase-modulating compounds disrupt the endogenous functions of CRBN. We report that CRBN functions in a feedback loop that harnesses antigen-specific CD8+ T-cell effector responses. Specifically, Crbn deficiency in murine CD8+ T cells augments their central metabolism manifested as elevated bioenergetics, with supraphysiological levels of polyamines, secondary to enhanced glucose and amino acid transport, and with increased expression of metabolic enzymes, including the polyamine biosynthetic enzyme ornithine decarboxylase. Treatment with CRBN-modulating compounds similarly augments central metabolism of human CD8+ T cells. Notably, the metabolic control of CD8+ T cells by modulating compounds or Crbn deficiency is linked to increased and sustained expression of the master metabolic regulator MYC. Finally, Crbn-deficient T cells have augmented antigen-specific cytolytic activity vs melanoma tumor cells, ex vivo and in vivo, and drive accelerated and highly aggressive graft-versus-host disease. Therefore, CRBN functions to harness the activation of CD8+ T cells, and this phenotype can be exploited by treatment with drugs.
Collapse
Affiliation(s)
- Rebecca S Hesterberg
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
- Cancer Biology PhD Program, University of South Florida, Tampa, FL
| | - Matthew S Beatty
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Ying Han
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Mario R Fernandez
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Afua A Akuffo
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
- Cancer Biology PhD Program, University of South Florida, Tampa, FL
| | - William E Goodheart
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Chunying Yang
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Shiun Chang
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
- Cancer Biology PhD Program, University of South Florida, Tampa, FL
| | - Christelle M Colin
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Aileen Y Alontaga
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Jessica M McDaniel
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Adam W Mailloux
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Julia M R Billington
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
- Cancer Biology PhD Program, University of South Florida, Tampa, FL
| | - Lanzhu Yue
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shonagh Russell
- Cancer Biology PhD Program, University of South Florida, Tampa, FL
- Department of Cancer Physiology
| | | | | | | | - Nicholas J Lawrence
- Department of Drug Discovery, Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Jianing Fu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | | | - John M Koomen
- Proteomics and Metabolomics Core
- Department of Molecular Oncology, and
| | - Xiubao Ren
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jane Messina
- Department of Anatomic Pathology and Cutaneous Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Kun Jiang
- Department of Anatomic Pathology and Cutaneous Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Timothy J Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL; and
| | - Anjali M Rajadhyaksha
- Pediatric Neurology, Pediatrics, Weill Family Brain and Mind Research Institute, and
- Graduate Program in Neuroscience, Weill Cornell Medical College, Cornell University, Cornell, NY
| | - John L Cleveland
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL
| | | |
Collapse
|
39
|
Targeting Multiple Myeloma through the Biology of Long-Lived Plasma Cells. Cancers (Basel) 2020; 12:cancers12082117. [PMID: 32751699 PMCID: PMC7466116 DOI: 10.3390/cancers12082117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/17/2020] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of terminally differentiated bone marrow (BM) resident B lymphocytes known as plasma cells (PC). PC that reside in the bone marrow include a distinct population of long-lived plasma cells (LLPC) that have the capacity to live for very long periods of time (decades in the human population). LLPC biology is critical for understanding MM disease induction and progression because MM shares many of the same extrinsic and intrinsic survival programs as LLPC. Extrinsic survival signals required for LLPC survival include soluble factors and cellular partners in the bone marrow microenvironment. Intrinsic programs that enhance cellular fidelity are also required for LLPC survival including increased autophagy, metabolic fitness, the unfolded protein response (UPR), and enhanced responsiveness to endoplasmic reticulum (ER) stress. Targeting LLPC cell survival mechanisms have led to standard of care treatments for MM including proteasome inhibition (Bortezomib), steroids (Dexamethasone), and immunomodulatory drugs (Lenalidomide). MM patients that relapse often do so by circumventing LLPC survival pathways targeted by treatment. Understanding the mechanisms by which LLPC are able to survive can allow us insight into the treatment of MM, which allows for the enhancement of therapeutic strategies in MM both at diagnosis and upon patient relapse.
Collapse
|
40
|
Qiu J, Villa M, Sanin DE, Buck MD, O'Sullivan D, Ching R, Matsushita M, Grzes KM, Winkler F, Chang CH, Curtis JD, Kyle RL, Van Teijlingen Bakker N, Corrado M, Haessler F, Alfei F, Edwards-Hicks J, Maggi LB, Zehn D, Egawa T, Bengsch B, Klein Geltink RI, Jenuwein T, Pearce EJ, Pearce EL. Acetate Promotes T Cell Effector Function during Glucose Restriction. Cell Rep 2020; 27:2063-2074.e5. [PMID: 31091446 DOI: 10.1016/j.celrep.2019.04.022] [Citation(s) in RCA: 240] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/12/2019] [Accepted: 04/02/2019] [Indexed: 12/26/2022] Open
Abstract
Competition for nutrients like glucose can metabolically restrict T cells and contribute to their hyporesponsiveness during cancer. Metabolic adaptation to the surrounding microenvironment is therefore key for maintaining appropriate cell function. For instance, cancer cells use acetate as a substrate alternative to glucose to fuel metabolism and growth. Here, we show that acetate rescues effector function in glucose-restricted CD8+ T cells. Mechanistically, acetate promotes histone acetylation and chromatin accessibility and enhances IFN-γ gene transcription and cytokine production in an acetyl-CoA synthetase (ACSS)-dependent manner. Ex vivo acetate treatment increases IFN-γ production by exhausted T cells, whereas reducing ACSS expression in T cells impairs IFN-γ production by tumor-infiltrating lymphocytes and tumor clearance. Thus, hyporesponsive T cells can be epigenetically remodeled and reactivated by acetate, suggesting that pathways regulating the use of substrates alternative to glucose could be therapeutically targeted to promote T cell function during cancer.
Collapse
Affiliation(s)
- Jing Qiu
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Matteo Villa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - David E Sanin
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Michael D Buck
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - David O'Sullivan
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Reagan Ching
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Mai Matsushita
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Katarzyna M Grzes
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Frances Winkler
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | | | - Jonathan D Curtis
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Ryan L Kyle
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | | | - Mauro Corrado
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Fabian Haessler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Francesca Alfei
- School of Life Science, Technical University of Munich, 80333 Munich, Germany
| | - Joy Edwards-Hicks
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Leonard B Maggi
- ICCE Institute and Department of Medicine, Division of Molecular Oncology, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dietmar Zehn
- School of Life Science, Technical University of Munich, 80333 Munich, Germany
| | - Takeshi Egawa
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bertram Bengsch
- BIOSS Center for Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
| | | | - Thomas Jenuwein
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Edward J Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Erika L Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany.
| |
Collapse
|
41
|
Abstract
Experiments in culture systems where one cell type is provided with abundant nutrients and oxygen have been used to inform much of our understanding of cancer metabolism. However, many differences have been observed between the metabolism of tumors and the metabolism of cancer cells grown in monoculture. These differences reflect, at least in part, the presence of nonmalignant cells in the tumor microenvironment and the interactions between those cells and cancer cells. However, less is known about how the metabolism of various tumor stromal cell types differs from that of cancer cells, and how this difference might inform therapeutic targeting of metabolic pathways. Emerging data have identified both cooperative and competitive relationships between different cell types in a tumor, and this review examines how four abundant stromal cell types in the tumor microenvironment, fibroblasts, T cells, macrophages, and endothelial cells, contribute to the metabolism of tumors.
Collapse
Affiliation(s)
- Allison N. Lau
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;,
| | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;,
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| |
Collapse
|
42
|
Wei Z, Dong C, Guan L, Wang Y, Huang J, Wen X. A metabolic exploration of the protective effect of Ligusticum wallichii on IL-1β-injured mouse chondrocytes. Chin Med 2020; 15:12. [PMID: 32025239 PMCID: PMC6995652 DOI: 10.1186/s13020-020-0295-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
Background Osteoarthritis (OA) is a metabolic disorder and able to be relieved by traditional Chinese medicines. However, the effect of Ligusticum wallichii on OA is unknown. Methods Cytokine IL-1β and L. wallichii extracts were used to stimulate the primary mouse chondrocytes. MTT assay was used to measure the cell viability. The mRNA and protein level of each gene were test by qRT-PCR and western blotting, respectively. The rate of apoptotic cell was measured by flow cytometry. GC/MS-based metabolomics was utilized to characterize the variation of metabolome. Results Here, we found that L. wallichii attenuated the IL-1β-induced apoptosis, inflammatory response, and extracellular matrix (ECM) degradation in mouse chondrocytes. Then we used GC/MS-based metabolomics to characterize the variation of metabolomes. The established metabolic profile of mouse chondrocytes showed that the abundance of most metabolites (n = 40) altered by IL-1β stimulation could be repressed by L. wallichii treatment. Multivariate data analysis identified that cholesterol, linoleic acid, hexadecandioic acid, proline, l-valine, l-leucine, pyruvate, palmitic acid, and proline are the most key biomarkers for understanding the metabolic role of L. wallichii in IL-1β-treated chondrocytes. Further pathway analysis using these metabolites enriched fourteen metabolic pathways, which were dramatically changed in IL-1β-treated chondrocytes and capable of being reprogrammed by L. wallichii incubation. These enriched pathways were involved in carbon metabolisms, fatty acid biosynthesis, and amino acid metabolisms. Conclusions These findings provide potential clues that metabolic strategies are linked to protective mechanisms of L. wallichii treatment in IL-1β-stimulated chondrocytes and emphasize the importance of metabolic strategies against inflammatory responses in OA development.
Collapse
Affiliation(s)
- Zhiqiang Wei
- 1Orthopaedics Department, Dongfang Hospital, Beijing University of Chinese Medicine, Bejing, 100078 China
| | - Chunjiao Dong
- Cardiology & Neurology Department, Beijing TongRen Tang Traditional Chinese Medicine Hospital, Bejing, 100051 China
| | - Liping Guan
- Intensive Care Unit, Huimin Hospital of Beijing, Bejing, 100013 China
| | - Yafei Wang
- 1Orthopaedics Department, Dongfang Hospital, Beijing University of Chinese Medicine, Bejing, 100078 China
| | - Jianghai Huang
- 1Orthopaedics Department, Dongfang Hospital, Beijing University of Chinese Medicine, Bejing, 100078 China
| | - Xinzhu Wen
- 1Orthopaedics Department, Dongfang Hospital, Beijing University of Chinese Medicine, Bejing, 100078 China
| |
Collapse
|
43
|
Cassim S, Pouyssegur J. Tumor Microenvironment: A Metabolic Player that Shapes the Immune Response. Int J Mol Sci 2019; 21:E157. [PMID: 31881671 PMCID: PMC6982275 DOI: 10.3390/ijms21010157] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023] Open
Abstract
Immune cells survey and patrol throughout the body and sometimes take residence in niche environments with distinct cellular subtypes and nutrients that may fluctuate from those in which they matured. Rooted in immune cell physiology are metabolic pathways and metabolites that not only deliver substrates and energy for growth and survival, but also instruct effector functions and cell differentiation. Unlike cancer cells, immune cells are not subject to a "Darwinian evolutionary pressure" that would allow them to adapt to developing tumors but are often irrevocably affected to local nutrient deprivation. Thus, immune cells must metabolically adapt to these changing conditions in order to perform their necessary functions. On the other hand, there is now a growing appreciation that metabolic changes occurring in cancer cells can impact on immune cell functionality and contribute to tumor immune evasion, and as such, there is a considerable and growing interest in developing techniques that target metabolism for immunotherapy. In this review, we discuss the metabolic plasticity displayed by innate and adaptive immune cells and highlight how tumor-derived lactate and tumor acidity restrict immunity. To our knowledge, this review outlines the most recent insights on how tumor microenvironment metabolically instructs immune responsiveness.
Collapse
Affiliation(s)
- Shamir Cassim
- Department of Medical Biology, Centre Scientifique de Monaco, CSM, 98000 Monaco, Monaco;
| | - Jacques Pouyssegur
- Department of Medical Biology, Centre Scientifique de Monaco, CSM, 98000 Monaco, Monaco;
- University Côte d’Azur, IRCAN, CNRS, Centre A. Lacassagne, 06189 Nice, France
| |
Collapse
|
44
|
Wei CW, Lee CY, Lee DJ, Chu CF, Wang JC, Wang TC, Jane WN, Chang ZF, Leu CM, Dzhagalov IL, Hsu CL. Equilibrative Nucleoside Transporter 3 Regulates T Cell Homeostasis by Coordinating Lysosomal Function with Nucleoside Availability. Cell Rep 2019; 23:2330-2341. [PMID: 29791845 DOI: 10.1016/j.celrep.2018.04.077] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/29/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
T cells are a versatile immune cell population responding to challenges by differentiation and proliferation followed by contraction and memory formation. Dynamic metabolic reprogramming is essential for T cells to meet the biosynthetic needs and the reutilization of biomolecules, processes that require active participation of metabolite transporters. Here, we show that equilibrative nucleoside transporter 3 (ENT3) is highly expressed in peripheral T cells and has a key role in maintaining T cell homeostasis by supporting the proliferation and survival of T cells. ENT3 deficiency leads to an enlarged and disturbed lysosomal compartment, resulting in accumulation of surplus mitochondria, elevation of intracellular reactive oxygen species, and DNA damage in T cells. Our results identify ENT3 as a vital metabolite transporter that supports T cell homeostasis and activation by regulating lysosomal integrity and the availability of nucleosides. Moreover, we uncovered that T cell lysosomes are an important source of salvaged metabolites for survival and proliferation.
Collapse
Affiliation(s)
- Chin-Wen Wei
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Chia-Ying Lee
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Ding-Jin Lee
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Chang-Feng Chu
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Ju-Chu Wang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Tien-Chiao Wang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Wann-Neng Jane
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 115, Taiwan
| | - Zee-Fen Chang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan; Institute of Molecular Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chuen-Miin Leu
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Ivan L Dzhagalov
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Chia-Lin Hsu
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan.
| |
Collapse
|
45
|
Sim SW, Weinstein DA, Lee YM, Jun HS. Glycogen storage disease type Ib: role of glucose‐6‐phosphate transporter in cell metabolism and function. FEBS Lett 2019; 594:3-18. [PMID: 31705665 DOI: 10.1002/1873-3468.13666] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/16/2019] [Accepted: 10/25/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Sang Wan Sim
- Department of Biotechnology and Bioinformatics College of Science and Technology Korea University Sejong Korea
| | - David A. Weinstein
- Glycogen Storage Disease Program University of Connecticut School of Medicine Farmington CT USA
| | - Young Mok Lee
- Glycogen Storage Disease Program University of Connecticut School of Medicine Farmington CT USA
| | - Hyun Sik Jun
- Department of Biotechnology and Bioinformatics College of Science and Technology Korea University Sejong Korea
| |
Collapse
|
46
|
Kobliakov VA. The Mechanisms of Regulation of Aerobic Glycolysis (Warburg Effect) by Oncoproteins in Carcinogenesis. BIOCHEMISTRY (MOSCOW) 2019; 84:1117-1128. [DOI: 10.1134/s0006297919100018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
47
|
ADP-dependent glucokinase regulates energy metabolism via ER-localized glucose sensing. Sci Rep 2019; 9:14248. [PMID: 31582762 PMCID: PMC6776650 DOI: 10.1038/s41598-019-50566-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 09/05/2019] [Indexed: 12/21/2022] Open
Abstract
Modulation of energy metabolism to a highly glycolytic phenotype, i.e. Warburg effect, is a common phenotype of cancer and activated immune cells allowing increased biomass-production for proliferation and cell division. Endoplasmic reticulum (ER)-localized ADP-dependent glucokinase (ADPGK) has been shown to play a critical role in T cell receptor activation-induced remodeling of energy metabolism, however the underlying mechanisms remain unclear. Therefore, we established and characterized in vitro and in vivo models for ADPGK-deficiency using Jurkat T cells and zebrafish. Upon activation, ADPGK knockout Jurkat T cells displayed increased cell death and ER stress. The increase in cell death resulted from a metabolic catastrophe and knockout cells displayed severely disturbed energy metabolism hindering induction of Warburg phenotype. ADPGK knockdown in zebrafish embryos led to short, dorsalized body axis induced by elevated apoptosis. ADPGK hypomorphic zebrafish further displayed dysfunctional glucose metabolism. In both model systems loss of ADPGK function led to defective N- and O-glycosylation. Overall, our data illustrate that ADPGK is part of a glucose sensing system in the ER modulating metabolism via regulation of N- and O-glycosylation.
Collapse
|
48
|
Watson AR, Dai H, Zheng Y, Nakano R, Giannou AD, Menk AV, Stolz DB, Delgoffe GM, Thomson AW. mTORC2 Deficiency Alters the Metabolic Profile of Conventional Dendritic Cells. Front Immunol 2019; 10:1451. [PMID: 31338091 PMCID: PMC6626913 DOI: 10.3389/fimmu.2019.01451] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 06/10/2019] [Indexed: 12/16/2022] Open
Abstract
In myeloid dendritic cells (DC), deletion of the mechanistic target of rapamycin complex 2 (TORC2) results in an augmented pro-inflammatory phenotype and T cell stimulatory activity; however, the underlying mechanism has not been resolved. Here, we demonstrate that mouse bone marrow-derived TORC2-deficient myeloid DC (TORC2−/− DC) utilize an altered metabolic program, characterized by enhanced baseline glycolytic function compared to wild-type WT control (Ctrl) DC, increased dependence on glycolytic ATP production, elevated lipid content and higher viability following stimulation with LPS. In addition, TORC2−/− DC display an increased spare respiratory capacity (SRC) compared to WT Ctrl DC; this metabolic phenotype corresponds with increased mitochondrial mass and mean mitochondrial DNA copy number, and failure of TORC2−/− DC mitochondria to depolarize following LPS stimulation. Our data suggest that the enhanced metabolic activity of TORC2−/− DC may be due to compensatory TORC1 pathway activity, namely increased expression of multiple genes upstream of Akt/TORC1 activity, including the integrin alpha IIb, protein tyrosine kinase 2/focal adhesion kinase, IL-7R and Janus kinase 1(JAK1), and the activation of downstream targets of TORC1, including p70S6K, eukaryotic translation initiation factor 4E binding protein 1 (4EBP1) and CD36 (fatty acid translocase). These enhanced TORC1 pathway activities may culminate in increased expression of the nuclear receptor peroxisome proliferator-activated receptor γ (Pparγ) that regulates fatty acid storage, and the transcription factor sterol regulatory element-binding transcription factor 1 (Srebf1). Taken together, our data suggest that TORC2 may function to restrain TORC1-driven metabolic activity and mitochondrial regulation in myeloid DC.
Collapse
Affiliation(s)
- Alicia R Watson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Helong Dai
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Urological Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yawen Zheng
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Urological Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ryosuke Nakano
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Anastasios D Giannou
- Section of Molecular Immunology and Gastroenterology, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ashley V Menk
- Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States
| | - Donna B Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Greg M Delgoffe
- Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Angus W Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
49
|
Mitochondrial Retrograde Signalling and Metabolic Alterations in the Tumour Microenvironment. Cells 2019; 8:cells8030275. [PMID: 30909478 PMCID: PMC6468901 DOI: 10.3390/cells8030275] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/22/2022] Open
Abstract
This review explores the molecular mechanisms that may be responsible for mitochondrial retrograde signalling related metabolic reprogramming in cancer and host cells in the tumour microenvironment and provides a summary of recent updates with regard to the functional modulation of diverse cells in the tumour microenvironment.
Collapse
|
50
|
Gemta LF, Siska PJ, Nelson ME, Gao X, Liu X, Locasale JW, Yagita H, Slingluff CL, Hoehn KL, Rathmell JC, Bullock TNJ. Impaired enolase 1 glycolytic activity restrains effector functions of tumor-infiltrating CD8 + T cells. Sci Immunol 2019; 4:eaap9520. [PMID: 30683669 PMCID: PMC6824424 DOI: 10.1126/sciimmunol.aap9520] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 08/10/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022]
Abstract
In the context of solid tumors, there is a positive correlation between the accumulation of cytotoxic CD8+ tumor-infiltrating lymphocytes (TILs) and favorable clinical outcomes. However, CD8+ TILs often exhibit a state of functional exhaustion, limiting their activity, and the underlying molecular basis of this dysfunction is not fully understood. Here, we show that TILs found in human and murine CD8+ melanomas are metabolically compromised with deficits in both glycolytic and oxidative metabolism. Although several studies have shown that tumors can outcompete T cells for glucose, thus limiting T cell metabolic activity, we report that a down-regulation in the activity of ENOLASE 1, a critical enzyme in the glycolytic pathway, represses glycolytic activity in CD8+ TILs. Provision of pyruvate, a downstream product of ENOLASE 1, bypasses this inactivity and promotes both glycolysis and oxidative phosphorylation, resulting in improved effector function of CD8+ TILs. We found high expression of both enolase 1 mRNA and protein in CD8+ TILs, indicating that the enzymatic activity of ENOLASE 1 is regulated posttranslationally. These studies provide a critical insight into the biochemical basis of CD8+ TIL dysfunction.
Collapse
Affiliation(s)
- Lelisa F Gemta
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Peter J Siska
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Marin E Nelson
- Department of Pharmacology, University of Virginia, VA 22908, USA
| | - Xia Gao
- Department of Pharmacology and Cancer Biology, Duke University, NC 27710, USA
| | - Xiaojing Liu
- Department of Pharmacology and Cancer Biology, Duke University, NC 27710, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University, NC 27710, USA
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Craig L Slingluff
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia 22908, USA
| | - Kyle L Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Timothy N J Bullock
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|