51
|
Singh AK, Qureshah FA, Drow T, Hou B, Rawlings DJ. Activated PI3Kδ specifically perturbs mouse Treg homeostasis and function leading to immune dysregulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.569665. [PMID: 38187650 PMCID: PMC10769388 DOI: 10.1101/2023.12.21.569665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Foxp3+ regulatory T cells (Treg) are required for maintaining immune tolerance and preventing systemic autoimmunity. PI3Kδ is required for normal Treg development and function. However, the impacts of dysregulated PI3Kδ signaling on Treg function remain incompletely understood. Here, we used a conditional mouse model of activated PI3Kδ syndrome (APDS) to investigate the role of altered PI3Kδ signaling specifically within the Treg compartment. Aged mice expressing a PIK3CD gain-of-function mutation (aPIK3CD) specifically within the Treg compartment exhibited weight loss and evidence for chronic inflammation as demonstrated by increased memory/effector CD4+ and CD8+ T cells with enhanced IFN-γ secretion, spontaneous germinal center responses and production of broad-spectrum autoantibodies. Intriguingly, aPIK3CD facilitated Treg precursor development within the thymus and an increase in peripheral Treg numbers. Peripheral Treg, however, exhibited an altered phenotype including increased PD1 expression and reduced competitive fitness. Consistent with these findings, Treg specific-aPIK3CD mice mounted an elevated humoral response following immunization with a T-cell dependent antigen, that correlated with a decrease in follicular Treg. Taken together, these findings demonstrate that an optimal threshold of PI3Kδ activity is critical for Treg homeostasis and function, suggesting that PI3Kδ signaling in Treg might be therapeutically targeted to either augment or inhibit immune responses.
Collapse
Affiliation(s)
- Akhilesh K. Singh
- Center for Immunity and Immunotherapy, Seattle Children’s Research Institute, Seattle, WA
| | - Fahd Al Qureshah
- Center for Immunity and Immunotherapy, Seattle Children’s Research Institute, Seattle, WA
- Department of Immunology, University of Washington, Seattle, WA
- King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Travis Drow
- Center for Immunity and Immunotherapy, Seattle Children’s Research Institute, Seattle, WA
| | - Baidong Hou
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - David J Rawlings
- Center for Immunity and Immunotherapy, Seattle Children’s Research Institute, Seattle, WA
- Department of Immunology, University of Washington, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| |
Collapse
|
52
|
Chen H, Wang X, Wang Y, Chang X. What happens to regulatory T cells in multiple myeloma. Cell Death Discov 2023; 9:468. [PMID: 38129374 PMCID: PMC10739837 DOI: 10.1038/s41420-023-01765-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Abnormal tumor microenvironment and immune escape in multiple myeloma (MM) are associated with regulatory T cells (Tregs), which play an important role in maintaining self-tolerance and regulating the overall immune response to infection or tumor cells. In patients with MM, there are abnormalities in the number, function and distribution of Tregs, and these abnormalities may be related to the disease stage, risk grade and prognosis of patients. During the treatment, Tregs have different responses to various treatment regiments, thus affecting the therapeutic effect of MM. It is also possible to predict the therapeutic response by observing the changes of Tregs. In addition to the above, we reviewed the application of Tregs in the treatment of MM. In conclusion, there is still much room for research on the mechanism and application of Tregs in MM.
Collapse
Affiliation(s)
- Huixian Chen
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xueling Wang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Yan Wang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiaotian Chang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
53
|
Han A, Peng T, Xie Y, Zhang W, Sun W, Xie Y, Ma Y, Wang C, Xie N. Mitochondrial-regulated Tregs: potential therapeutic targets for autoimmune diseases of the central nervous system. Front Immunol 2023; 14:1301074. [PMID: 38149252 PMCID: PMC10749924 DOI: 10.3389/fimmu.2023.1301074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Regulatory T cells (Tregs) can eliminate autoreactive lymphocytes, induce self-tolerance, and suppress the inflammatory response. Mitochondria, as the energy factories of cells, are essential for regulating the survival, differentiation, and function of Tregs. Studies have shown that patients with autoimmune diseases of the central nervous system, such as multiple sclerosis, neuromyelitis optica spectrum disorder, and autoimmune encephalitis, have aberrant Tregs and mitochondrial damage. However, the role of mitochondrial-regulated Tregs in autoimmune diseases of the central nervous system remains inconclusive. Therefore, this study reviews the mitochondrial regulation of Tregs in autoimmune diseases of the central nervous system and investigates the possible mitochondrial therapeutic targets.
Collapse
Affiliation(s)
- Aoya Han
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tingting Peng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yinyin Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wanwan Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenlin Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunqing Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cui Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nanchang Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
54
|
Zhao L, Ma D, Wang L, Su X, Feng L, Zhu L, Chen Y, Hao Y, Wang X, Feng J. Metabolic changes with the occurrence of atherosclerotic plaques and the effects of statins. Front Immunol 2023; 14:1301051. [PMID: 38143759 PMCID: PMC10739339 DOI: 10.3389/fimmu.2023.1301051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/23/2023] [Indexed: 12/26/2023] Open
Abstract
Atherosclerosis is a common cardiovascular disease caused by the abnormal expression of multiple factors and genes influenced by both environmental and genetic factors. The primary manifestation of atherosclerosis is plaque formation, which occurs when inflammatory cells consume excess lipids, affecting their retention and modification within the arterial intima. This triggers endothelial cell (EC) activation, immune cell infiltration, vascular smooth muscle cell (VSMC) proliferation and migration, foam cell formation, lipid streaks, and fibrous plaque development. These processes can lead to vascular wall sclerosis, lumen stenosis, and thrombosis. Immune cells, ECs, and VSMCs in atherosclerotic plaques undergo significant metabolic changes and inflammatory responses. The interaction of cytokines and chemokines secreted by these cells leads to the onset, progression, and regression of atherosclerosis. The regulation of cell- or cytokine-based immune responses is a novel therapeutic approach for atherosclerosis. Statins are currently the primary pharmacological agents utilised for managing unstable plaques owing to their ability to enhance endothelial function, regulate VSMC proliferation and apoptosis by reducing cholesterol levels, and mitigate the expression and activity of inflammatory cytokines. In this review, we provide an overview of the metabolic changes associated with atherosclerosis, describe the effects of inflammatory responses on atherosclerotic plaques, and discuss the mechanisms through which statins contribute to plaque stabilisation. Additionally, we examine the role of statins in combination with other drugs in the management of atherosclerosis.
Collapse
Affiliation(s)
| | - Di Ma
- Bethune First Hospital, Jilin University, Changchun, China
| | - LiJuan Wang
- Bethune First Hospital, Jilin University, Changchun, China
| | | | | | | | | | | | | | | |
Collapse
|
55
|
Cosorich I, Filoni J, Di Dedda C, Ferrari A, Jofra T, Cesarano S, Bonini C, Piemonti L, Monti P. Interleukin-7 improves the fitness of regulatory T cells for adoptive transfer. Immunology 2023; 170:540-552. [PMID: 37671510 DOI: 10.1111/imm.13690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/18/2023] [Indexed: 09/07/2023] Open
Abstract
Adoptive regulatory T-cell (Treg) transfer has emerged as a promising therapeutic strategy for regulating immune responses in organ transplantation, graft versus host disease, and autoimmunity, including Type 1 diabetes. Traditionally, Treg for adoptive therapy have been sorted and expanded in vitro using high doses of IL-2, demonstrating stability and suppressive capabilities. However, limitations in their long-term survival post-infusion into patients have been observed. To address this challenge, we investigated a novel expansion protocol incorporating interleukin-7 (IL-7) alongside the traditional method utilizing IL-2 (referred to as IL-7 method, IL-7M). Our study revealed that naïve Treg express significant levels of CD127 and display robust responsiveness to IL-7, characterized by STAT-5 phosphorylation. Expanding naïve Treg with the IL-7M protocol led to a substantial enrichment of CD45RA+ CD62L+ CD95+ Treg but showing a reduction in the final cell yield and suppressive function. Moreover, Treg expanded with the IL-7M exhibited preserved telomere length and demonstrated enhanced resistance to cytokine withdrawal and fas-mediated apoptosis. When transferred into NSG mice IL-7M-Treg persisted longer and reduced the expansion of T cells, but did not significantly reduce the severity of xenoGvHD. In conclusion, our data demonstrate the feasibility of expanding naïve Treg in the presence of IL-7 to generate a Treg product enriched in poorly differentiated CD45RA+ cells with enhanced survival capabilities.
Collapse
Affiliation(s)
- Ilaria Cosorich
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Jessica Filoni
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Carla Di Dedda
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Arianna Ferrari
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Tatiana Jofra
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Susanna Cesarano
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Monti
- Transplant Immunology Lab, San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| |
Collapse
|
56
|
Chen Z, Guan D, Wang Z, Li X, Dong S, Huang J, Zhou W. Microbiota in cancer: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2023; 4:e417. [PMID: 37937304 PMCID: PMC10626288 DOI: 10.1002/mco2.417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 11/09/2023] Open
Abstract
The diverse bacterial populations within the symbiotic microbiota play a pivotal role in both health and disease. Microbiota modulates critical aspects of tumor biology including cell proliferation, invasion, and metastasis. This regulation occurs through mechanisms like enhancing genomic damage, hindering gene repair, activating aberrant cell signaling pathways, influencing tumor cell metabolism, promoting revascularization, and remodeling the tumor immune microenvironment. These microbiota-mediated effects significantly impact overall survival and the recurrence of tumors after surgery by affecting the efficacy of chemoradiotherapy. Moreover, leveraging the microbiota for the development of biovectors, probiotics, prebiotics, and synbiotics, in addition to utilizing antibiotics, dietary adjustments, defensins, oncolytic virotherapy, and fecal microbiota transplantation, offers promising alternatives for cancer treatment. Nonetheless, due to the extensive and diverse nature of the microbiota, along with tumor heterogeneity, the molecular mechanisms underlying the role of microbiota in cancer remain a subject of intense debate. In this context, we refocus on various cancers, delving into the molecular signaling pathways associated with the microbiota and its derivatives, the reshaping of the tumor microenvironmental matrix, and the impact on tolerance to tumor treatments such as chemotherapy and radiotherapy. This exploration aims to shed light on novel perspectives and potential applications in the field.
Collapse
Affiliation(s)
- Zhou Chen
- The First Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The First Hospital of Lanzhou UniversityLanzhouGansuChina
| | - Defeng Guan
- The First Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The First Hospital of Lanzhou UniversityLanzhouGansuChina
| | - Zhengfeng Wang
- The First Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The First Hospital of Lanzhou UniversityLanzhouGansuChina
| | - Xin Li
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The Department of General SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Shi Dong
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The Department of General SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Junjun Huang
- The First Hospital of Lanzhou UniversityLanzhouGansuChina
| | - Wence Zhou
- The First Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The Department of General SurgeryLanzhou University Second HospitalLanzhouGansuChina
| |
Collapse
|
57
|
Flynn AL, Gans J, Escobedo J, Zhu C, Florescu AM, Shankara S, Madden SL, Kim PS, Pao LI. RGS1 Modulates Autophagic and Metabolic Programs and Is a Critical Mediator of Human Regulatory T Cell Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1656-1668. [PMID: 37850953 DOI: 10.4049/jimmunol.2200402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/27/2023] [Indexed: 10/19/2023]
Abstract
Regulatory T cells (Tregs) are critical mediators of immune tolerance and play a diametric role in cancer and autoimmunity. Tumor-infiltrating Tregs are often associated with poor prognosis in solid tumors because their enrichment in the tumor microenvironment contributes to immunosuppression. Conversely, dysregulation in the Treg compartment can disrupt self-tolerance, leading to autoimmunity. In the present study, we describe what is, to our knowledge, a novel regulator of Tregs, the GTPase activator regulator of G protein 1 (RGS1), demonstrating that RGS1-deficient human Tregs show downregulation of Treg-associated genes and are less immunosuppressive. These RGS1-deficient Tregs exhibit perturbations to the FOXP3-c-MYC transcriptional axis and downstream metabolic and autophagy programs by shifting their energy demands toward glycolysis and rendering them less autophagic. Taken together, RGS1 may serve as an apical node of Treg function by regulating the FOXP3-c-MYC transcriptional axis, thereby providing a therapeutic rationale for targeting RGS1 for treatment of cancer and autoimmune diseases.
Collapse
Affiliation(s)
| | - Joseph Gans
- Department of Translational Sciences, Sanofi, Cambridge, MA
| | | | - Cheng Zhu
- Department of Translational Sciences, Sanofi, Cambridge, MA
| | | | | | | | - Peter S Kim
- Department of Oncology, Sanofi, Cambridge, MA
| | - Lily I Pao
- Department of Oncology, Sanofi, Cambridge, MA
| |
Collapse
|
58
|
Chen H, Zhang Y, Chen X, Xu R, Zhu Y, He D, Cheng Y, Wang Z, Qing X, Cao K. Hypoxia is correlated with the tumor immune microenvironment: Potential application of immunotherapy in bladder cancer. Cancer Med 2023; 12:22333-22353. [PMID: 38063246 PMCID: PMC10757107 DOI: 10.1002/cam4.6617] [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: 04/06/2022] [Revised: 10/16/2022] [Accepted: 11/17/2022] [Indexed: 12/31/2023] Open
Abstract
OBJECTIVE Hypoxia, which can considerably affect the tumor microenvironment, hinders the use of immunotherapy in bladder cancer (BLCA). Therefore, we aimed to identify reliable hypoxia-related biomarkers to guide clinical immunotherapy in BLCA. METHODS Using data downloaded from TCGA-BLCA cohort, we determined BLCA subtypes which divide 408 samples into different subtypes. Tumor immune infiltration levels of two clusters were quantified using ssGSEA, MCPcounter, EPIC, ESTIMATE, and TIMER algorithms. Next, we constructed a hypoxia score based on the expression of hypoxia-related genes. The IMvigor210 cohort and SubMap analysis were used to predict immunotherapeutic responses in patients with different hypoxia scores. Hub genes were screened using cytoscape, immunohistochemistry (IHC), and multispectral immunofluorescence were used to detect the spatial distribution of immune markers. RESULTS Patients with BLCA were categorized into cluster1 (n = 227) and Cluster2 (n = 181). Immune infiltration and expression of immune markers were higher in Cluster1. Immune infiltration was also more obvious in the high-hypoxia score group which related to a better predicted response to immunotherapy. IHC, and multispectral immunofluorescence confirmed the importance of TLR8 in immune infiltration and immune phenotype. CONCLUSIONS BLCA subtype can evaluate the infiltration of immune cells in the tumor microenvironment of different patients. Hypoxia score in this study could effectively predict immunotherapeutic responses in patients with BLCA. TLR8 may be a potential target for clinical immunotherapy.
Collapse
Affiliation(s)
- Haotian Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yao Zhang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xingyu Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Runshi Xu
- Department of Pathology, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Yuxing Zhu
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Dong He
- Department of Respiration, The Second People's Hospital of Hunan Province of Hunan University of Chinese Medicine, Changsha, China
| | - YaXin Cheng
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhanwang Wang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiang Qing
- Department of Otolaryngology-Head and Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
59
|
Pinioti S, Sharma H, Flerin NC, Yu Q, Tzoumpa A, Cafarello ST, De Bousser E, Callewaert N, Oldenhove G, Schlenner S, Thienpont B, Garg AD, Di Matteo M, Mazzone M. A Metabolic Gene Survey Pinpoints Fucosylation as a Key Pathway Underlying the Suppressive Function of Regulatory T Cells in Cancer. Cancer Immunol Res 2023; 11:1611-1629. [PMID: 37933083 PMCID: PMC7615342 DOI: 10.1158/2326-6066.cir-22-0606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 02/22/2023] [Accepted: 10/18/2023] [Indexed: 11/08/2023]
Abstract
Forkhead box P3 (Foxp3)-expressing regulatory T cells (Treg) are the guardians of controlled immune reactions and prevent the development of autoimmune diseases. However, in the tumor context, their increased number suppresses antitumor immune responses, indicating the importance of understanding the mechanisms behind their function and stability. Metabolic reprogramming can affect Foxp3 regulation and, therefore, Treg suppressive function and fitness. Here, we performed a metabolic CRISPR/Cas9 screen and pinpointed novel candidate positive and negative metabolic regulators of Foxp3. Among the positive regulators, we revealed that targeting the GDP-fucose transporter Slc35c1, and more broadly fucosylation (Fuco), in Tregs compromises their proliferation and suppressive function both in vitro and in vivo, leading to alteration of the tumor microenvironment and impaired tumor progression and protumoral immune responses. Pharmacologic inhibition of Fuco dampened tumor immunosuppression mostly by targeting Tregs, thus resulting in reduced tumor growth. In order to substantiate these findings in humans, tumoral Tregs from patients with colorectal cancer were clustered on the basis of the expression of Fuco-related genes. FucoLOW Tregs were found to exhibit a more immunogenic profile compared with FucoHIGH Tregs. Furthermore, an enrichment of a FucoLOW signature, mainly derived from Tregs, correlated with better prognosis and response to immune checkpoint blockade in melanoma patients. In conclusion, Slc35c1-dependent Fuco is able to regulate the suppressive function of Tregs, and measuring its expression in Tregs might pave the way towards a useful biomarker model for patients with cancer. See related Spotlight by Silveria and DuPage, p. 1570.
Collapse
Affiliation(s)
- Sotiria Pinioti
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B3000, Belgium
| | - Himal Sharma
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B3000, Belgium
| | - Nina C Flerin
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B3000, Belgium
| | - Qian Yu
- laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Leuven B3000, Belgium
| | - Amalia Tzoumpa
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B3000, Belgium
| | - Sarah Trusso Cafarello
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B3000, Belgium
| | - Elien De Bousser
- Medical Biotechnology Center, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Nico Callewaert
- Medical Biotechnology Center, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Guillaume Oldenhove
- laboratory of Immunobiology, Université Libre de Bruxelles, Gosselies, Belgium
- U-CRI (UCL Center for Research in Immunobiology), Université Libre de Bruxelles, Gosselies, Belgium
| | - Susan Schlenner
- KU Leuven-University of Leuven, Department of Microbiology, Immunology and Transplantation, Leuven B3000, Belgium
| | - Bernard Thienpont
- laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Leuven B3000, Belgium
| | - Abhishek D Garg
- laboratory for Cell Stress & Immunity (CSI), Department for Cellular and Molecular Medicine, KU Leuven, Leuven B3000, Belgium
| | - Mario Di Matteo
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B3000, Belgium
| | - Massimiliano Mazzone
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven B3000, Belgium
- laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B3000, Belgium
| |
Collapse
|
60
|
Deng J, Pan T, Liu Z, McCarthy C, Vicencio JM, Cao L, Alfano G, Suwaidan AA, Yin M, Beatson R, Ng T. The role of TXNIP in cancer: a fine balance between redox, metabolic, and immunological tumor control. Br J Cancer 2023; 129:1877-1892. [PMID: 37794178 PMCID: PMC10703902 DOI: 10.1038/s41416-023-02442-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/07/2023] [Accepted: 09/14/2023] [Indexed: 10/06/2023] Open
Abstract
Thioredoxin-interacting protein (TXNIP) is commonly considered a master regulator of cellular oxidation, regulating the expression and function of Thioredoxin (Trx). Recent work has identified that TXNIP has a far wider range of additional roles: from regulating glucose and lipid metabolism, to cell cycle arrest and inflammation. Its expression is increased by stressors commonly found in neoplastic cells and the wider tumor microenvironment (TME), and, as such, TXNIP has been extensively studied in cancers. In this review, we evaluate the current literature regarding the regulation and the function of TXNIP, highlighting its emerging role in modulating signaling between different cell types within the TME. We then assess current and future translational opportunities and the associated challenges in this area. An improved understanding of the functions and mechanisms of TXNIP in cancers may enhance its suitability as a therapeutic target.
Collapse
Affiliation(s)
- Jinhai Deng
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
- Clinical Research Center (CRC), Clinical Pathology Center (CPC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China
| | - Teng Pan
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518172, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Caitlin McCarthy
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Jose M Vicencio
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Lulu Cao
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Giovanna Alfano
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Ali Abdulnabi Suwaidan
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Mingzhu Yin
- Clinical Research Center (CRC), Clinical Pathology Center (CPC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China
| | - Richard Beatson
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK.
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College London (UCL), Rayne 9 Building, London, WC1E 6JF, UK.
| | - Tony Ng
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK.
- UCL Cancer Institute, University College London, London, UK.
- Cancer Research UK City of London Centre, London, UK.
| |
Collapse
|
61
|
Perpiñán E, Sanchez-Fueyo A, Safinia N. Immunoregulation: the interplay between metabolism and redox homeostasis. FRONTIERS IN TRANSPLANTATION 2023; 2:1283275. [PMID: 38993920 PMCID: PMC11235320 DOI: 10.3389/frtra.2023.1283275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/13/2023] [Indexed: 07/13/2024]
Abstract
Regulatory T cells are fundamental for the induction and maintenance of immune homeostasis, with their dysfunction resulting in uncontrolled immune responses and tissue destruction predisposing to autoimmunity, transplant rejection and several inflammatory and metabolic disorders. Recent discoveries have demonstrated that metabolic processes and mitochondrial function are critical for the appropriate functioning of these cells in health, with their metabolic adaptation, influenced by microenvironmental factors, seen in several pathological processes. Upon activation regulatory T cells rearrange their oxidation-reduction (redox) system, which in turn supports their metabolic reprogramming, adding a layer of complexity to our understanding of cellular metabolism. Here we review the literature surrounding redox homeostasis and metabolism of regulatory T cells to highlight new mechanistic insights of these interlinked pathways in immune regulation.
Collapse
Affiliation(s)
| | | | - N. Safinia
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Institute of Liver Studies, James Black Centre, King’s College London, London, United Kingdom
| |
Collapse
|
62
|
Su R, Zhao S, Zhang J, Cao M, Peng S. Metabolic influences on T cell in psoriasis: a literature review. Front Immunol 2023; 14:1279846. [PMID: 38035065 PMCID: PMC10684739 DOI: 10.3389/fimmu.2023.1279846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
Psoriasis is a systemic inflammatory disease that frequently coexists with various other conditions, such as essential hypertension, diabetes, metabolic syndrome, and inflammatory bowel disease. The association between these diseases may be attributed to shared inflammatory pathways and abnormal immunomodulatory mechanisms. Furthermore, metabolites also play a regulatory role in the function of different immune cells involved in psoriasis pathogenesis, particularly T lymphocytes. In this review, we have summarized the current research progress on T cell metabolism in psoriasis, encompassing the regulation of metabolites in glucose metabolism, lipid metabolism, amino acid metabolism, and other pathways within T cells affected by psoriasis. We will also explore the interaction and mechanism between psoriatic metabolites and immune cells. Moreover, we further discussed the research progress of metabolomics in psoriasis to gain a deeper understanding of its pathogenesis and identify potential new therapeutic targets through identification of metabolic biomarkers associated with this condition.
Collapse
Affiliation(s)
- Rina Su
- *Correspondence: Shiguang Peng, ; Rina Su,
| | | | | | | | - Shiguang Peng
- Department of Dermatology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
63
|
Kang JH, Zappasodi R. Modulating Treg stability to improve cancer immunotherapy. Trends Cancer 2023; 9:911-927. [PMID: 37598003 DOI: 10.1016/j.trecan.2023.07.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
Immunosuppressive regulatory T cells (Tregs) provide a main mechanism of tumor immune evasion. Targeting Tregs, especially in the tumor microenvironment (TME), continues to be investigated to improve cancer immunotherapy. Recent studies have unveiled intratumoral Treg heterogeneity and plasticity, furthering the complexity of the role of Tregs in tumor immunity and immunotherapy response. The phenotypic and functional diversity of intratumoral Tregs can impact their response to therapy and may offer new targets to modulate specific Treg subsets. In this review we provide a unifying framework of critical factors contributing to Treg heterogeneity and plasticity in the TME, and we discuss how this information can guide the development of more specific Treg-targeting therapies for cancer immunotherapy.
Collapse
Affiliation(s)
- Jee Hye Kang
- Weill Cornell Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY, USA
| | - Roberta Zappasodi
- Weill Cornell Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY, USA.
| |
Collapse
|
64
|
Ma J, Hu W, Liu Y, Duan C, Zhang D, Wang Y, Cheng K, Yang L, Wu S, Jin B, Zhang Y, Zhuang R. CD226 maintains regulatory T cell phenotype stability and metabolism by the mTOR/Myc pathway under inflammatory conditions. Cell Rep 2023; 42:113306. [PMID: 37864795 DOI: 10.1016/j.celrep.2023.113306] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 09/22/2023] [Accepted: 10/04/2023] [Indexed: 10/23/2023] Open
Abstract
Regulatory T (Treg) cells exhibit immunosuppressive phenotypes and particular metabolic patterns with certain degrees of plasticity. Previous studies of the effects of the co-stimulatory molecule CD226 on Treg cells are controversial. Here, we show that CD226 primarily maintains the Treg cell stability and metabolism phenotype under inflammatory conditions. Conditional deletion of CD226 within Foxp3+ cells exacerbates symptoms in murine graft versus host disease models. Treg cell-specific deletion of CD226 increases the Treg cell percentage in immune organs but weakens their immunosuppressive function with a T helper 1-like phenotype conversion under inflammation. CD226-deficient Treg cells exhibit reduced oxidative phosphorylation and increased glycolysis rates, which are regulated by the adenosine 5'-monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR)/myelocytomatosis oncogene (Myc) pathway, and inhibition of Myc signaling restores the impaired functions of CD226-deficient Treg cells in an inflammatory disease model of colitis. This study reveals an Myc-mediated CD226 regulation of Treg cell phenotypic stability and metabolism, providing potential therapeutic strategies for targeted interventions of Treg cell-specific CD226 in inflammatory diseases.
Collapse
Affiliation(s)
- Jingchang Ma
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Wei Hu
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China; Department of Emergency, The Fifth Medical Center of Chinese PLA General Hospital, #100 Western 4th Ring Road, Beijing 100039, China
| | - Yitian Liu
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Chujun Duan
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China; Institute of Medical Research, Northwestern Polytechnical University, #127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Dongliang Zhang
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Yuling Wang
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Kun Cheng
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Lu Yang
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Shuwen Wu
- Institute of Medical Research, Northwestern Polytechnical University, #127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Boquan Jin
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Yuan Zhang
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China; Institute of Medical Research, Northwestern Polytechnical University, #127 West Youyi Road, Xi'an, Shaanxi 710072, China.
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, #169 West Changle Road, Xi'an, Shaanxi 710032, China; Institute of Medical Research, Northwestern Polytechnical University, #127 West Youyi Road, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
65
|
Tippetts TS, Sieber MH, Solmonson A. Beyond energy and growth: the role of metabolism in developmental signaling, cell behavior and diapause. Development 2023; 150:dev201610. [PMID: 37883062 PMCID: PMC10652041 DOI: 10.1242/dev.201610] [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] [Indexed: 10/27/2023]
Abstract
Metabolism is crucial for development through supporting cell growth, energy production, establishing cell identity, developmental signaling and pattern formation. In many model systems, development occurs alongside metabolic transitions as cells differentiate and specialize in metabolism that supports new functions. Some cells exhibit metabolic flexibility to circumvent mutations or aberrant signaling, whereas other cell types require specific nutrients for developmental progress. Metabolic gradients and protein modifications enable pattern formation and cell communication. On an organism level, inadequate nutrients or stress can limit germ cell maturation, implantation and maturity through diapause, which slows metabolic activities until embryonic activation under improved environmental conditions.
Collapse
Affiliation(s)
- Trevor S. Tippetts
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Matthew H. Sieber
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ashley Solmonson
- Laboratory of Developmental Metabolism and Placental Biology, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
66
|
Luo Z, Zhang Y, Saleh QW, Zhang J, Zhu Z, Tepel M. Metabolic regulation of forkhead box P3 alternative splicing isoforms and their impact on health and disease. Front Immunol 2023; 14:1278560. [PMID: 37868998 PMCID: PMC10588449 DOI: 10.3389/fimmu.2023.1278560] [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: 08/16/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Forkhead Box P3 (FOXP3) is crucial for the development and suppressive function of human regulatory T cells (Tregs). There are two predominant FOXP3 splicing isoforms in healthy humans, the full-length isoform and the isoform lacking exon 2, with different functions and regulation mechanisms. FOXP3 splicing isoforms show distinct abilities in the cofactor interaction and the nuclear translocation, resulting in different effects on the differentiation, cytokine secretion, suppressive function, linage stability, and environmental adaptation of Tregs. The balance of FOXP3 splicing isoforms is related to autoimmune diseases, inflammatory diseases, and cancers. In response to environmental challenges, FOXP3 transcription and splicing can be finely regulated by T cell antigen receptor stimulation, glycolysis, fatty acid oxidation, and reactive oxygen species, with various signaling pathways involved. Strategies targeting energy metabolism and FOXP3 splicing isoforms in Tregs may provide potential new approaches for the treatment of autoimmune diseases, inflammatory diseases, and cancers. In this review, we summarize recent discoveries about the FOXP3 splicing isoforms and address the metabolic regulation and specific functions of FOXP3 splicing isoforms in Tregs.
Collapse
Affiliation(s)
- Zhidan Luo
- Department of Geriatrics, Chongqing General Hospital, Chongqing, China
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Yihua Zhang
- Department of Cardiology, Chongqing Fifth People’s Hospital, Chongqing, China
| | - Qais Waleed Saleh
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Jie Zhang
- Department of Geriatrics, Chongqing General Hospital, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Chongqing, China
| | - Martin Tepel
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
67
|
Zhang F, Cheng T, Zhang SX. Mechanistic target of rapamycin (mTOR): a potential new therapeutic target for rheumatoid arthritis. Arthritis Res Ther 2023; 25:187. [PMID: 37784141 PMCID: PMC10544394 DOI: 10.1186/s13075-023-03181-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by systemic synovitis and bone destruction. Proinflammatory cytokines activate pathways of immune-mediated inflammation, which aggravates RA. The mechanistic target of rapamycin (mTOR) signaling pathway associated with RA connects immune and metabolic signals, which regulates immune cell proliferation and differentiation, macrophage polarization and migration, antigen presentation, and synovial cell activation. Therefore, therapy strategies targeting mTOR have become an important direction of current RA treatment research. In the current review, we summarize the biological functions of mTOR, its regulatory effects on inflammation, and the curative effects of mTOR inhibitors in RA, thus providing references for the development of RA therapeutic targets and new drugs.
Collapse
Affiliation(s)
- Fen Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Xinghualing District, Taiyuan, 030001, Shanxi Province, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Ting Cheng
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Xinghualing District, Taiyuan, 030001, Shanxi Province, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Xinghualing District, Taiyuan, 030001, Shanxi Province, China.
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China.
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China.
| |
Collapse
|
68
|
Cheng D, Zhang Z, Mi Z, Tao W, Liu D, Fu J, Fan H. Deciphering the heterogeneity and immunosuppressive function of regulatory T cells in osteosarcoma using single-cell RNA transcriptome. Comput Biol Med 2023; 165:107417. [PMID: 37669584 DOI: 10.1016/j.compbiomed.2023.107417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/03/2023] [Accepted: 08/28/2023] [Indexed: 09/07/2023]
Abstract
Osteosarcoma (OS) is a highly invasive malignant neoplasm with poor prognosis. The tumor microenvironment (TME) plays an essential role in the occurrence and development of OS. Regulatory T cells (Tregs) are known to facilitate immunosuppression, tumor progression, invasion, and metastasis. However, the effect of Tregs in the TME of OS remains unclear. In this study, single-cell RNA sequencing (scRNA-seq) data was used to identify Tregs and various other cell clusters in the TME of OS. Gene set variation analysis (GSVA) was used to investigate the signaling pathways in Tregs from OS and adjacent tissues. The CellChat and iTALK packages were used to analyze cellular communication. In addition, a prognostic model was established based on the Tregs-specific genes using bulk RNA-seq from the TARGET database, and it was verified using a Gene Expression Omnibus dataset. The pRRophetic package was used to predict drug sensitivity. Immunohistochemistry was used to verify the expression of candidate genes in OS. Based on the above methods, we showed that the OS samples were highly infiltrated with Tregs. GSVA revealed that oxidative phosphorylation, angiogenesis and mammalian target of rapamycin complex 1 (mTORC1) were highly activated in Tregs from OS compared with those from adjacent tissues. Using cellular communication analysis, we found that Tregs interacted with osteoblastic, endothelial, and myeloid cells via C-X-C motif chemokine ligand (CXCL) signaling; particularly, they strongly affected the expression of C-X-C motif chemokine receptor 4 (CXCR4) and interacted with other cell clusters through CXCL12/transforming growth factor β1 (TGFB1) to collectively enable tumor growth and progression. Subsequently, two Tregs-specific genes-CD320 and MAF-were screened through univariate, least absolute shrinkage and selection operator regression (LASSO) and multivariate analysis to construct a prognostic model, which showed excellent prognostic accuracy in two independent cohorts. In addition, drug sensitivity analysis demonstrated that OS patients at high Tregs risk were sensitive to sunitinib, sorafenib, and axitinib. We also used immunohistochemistry to validate that CD320 and MAF were significantly upregulated in OS tissues compared with adjacent tissues. Overall, this study reveals the heterogeneity of Tregs in the OS TME, providing new insights into the invasion and treatment of this cancer.
Collapse
Affiliation(s)
- Debin Cheng
- Department of Orthopaedic Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhao Zhang
- Department of Orthopaedic Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhenzhou Mi
- Department of Orthopaedic Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Weidong Tao
- Department of Orthopaedic Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Dong Liu
- Department of Orthopaedic Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun Fu
- Department of Orthopaedic Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hongbin Fan
- Department of Orthopaedic Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
69
|
Lee H, Jeon JH, Kim ES. Mitochondrial dysfunctions in T cells: focus on inflammatory bowel disease. Front Immunol 2023; 14:1219422. [PMID: 37809060 PMCID: PMC10556505 DOI: 10.3389/fimmu.2023.1219422] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
Mitochondria has emerged as a critical ruler of metabolic reprogramming in immune responses and inflammation. In the context of colitogenic T cells and IBD, there has been increasing research interest in the metabolic pathways of glycolysis, pyruvate oxidation, and glutaminolysis. These pathways have been shown to play a crucial role in the metabolic reprogramming of colitogenic T cells, leading to increased inflammatory cytokine production and tissue damage. In addition to metabolic reprogramming, mitochondrial dysfunction has also been implicated in the pathogenesis of IBD. Studies have shown that colitogenic T cells exhibit impaired mitochondrial respiration, elevated levels of mROS, alterations in calcium homeostasis, impaired mitochondrial biogenesis, and aberrant mitochondria-associated membrane formation. Here, we discuss our current knowledge of the metabolic reprogramming and mitochondrial dysfunctions in colitogenic T cells, as well as the potential therapeutic applications for treating IBD with evidence from animal experiments.
Collapse
Affiliation(s)
- Hoyul Lee
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Han Jeon
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Eun Soo Kim
- Division of Gastroenterology, Department of Internal Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
70
|
Halvorson T, Tuomela K, Levings MK. Targeting regulatory T cell metabolism in disease: Novel therapeutic opportunities. Eur J Immunol 2023; 53:e2250002. [PMID: 36891988 DOI: 10.1002/eji.202250002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/28/2023] [Accepted: 03/06/2023] [Indexed: 03/10/2023]
Abstract
Regulatory T cells (Tregs) are essential for immune homeostasis and suppression of pathological autoimmunity but can also play a detrimental role in cancer progression via inhibition of anti-tumor immunity. Thus, there is broad applicability for therapeutic Treg targeting, either to enhance function, for example, through adoptive cell therapy (ACT), or to inhibit function with small molecules or antibody-mediated blockade. For both of these strategies, the metabolic state of Tregs is an important consideration since cellular metabolism is intricately linked to function. Mounting evidence has shown that targeting metabolic pathways can selectively promote or inhibit Treg function. This review aims to synthesize the current understanding of Treg metabolism and discuss emerging metabolic targeting strategies in the contexts of transplantation, autoimmunity, and cancer. We discuss approaches to gene editing and cell culture to manipulate Treg metabolism during ex vivo expansion for ACT, as well as in vivo nutritional and pharmacological interventions to modulate Treg metabolism in disease states. Overall, the intricate connection between metabolism and phenotype presents a powerful opportunity to therapeutically tune Treg function.
Collapse
Affiliation(s)
- Torin Halvorson
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karoliina Tuomela
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
71
|
Kao YS, Mamareli P, Dhillon-LaBrooy A, Stüve P, Godoy GJ, Velasquez LN, Raker VK, Weidenthaler-Barth B, Boukhallouk F, Rampoldi F, Berod L, Sparwasser T. Targeting ACC1 in T cells ameliorates psoriatic skin inflammation. J Mol Med (Berl) 2023; 101:1153-1166. [PMID: 37594540 PMCID: PMC10482807 DOI: 10.1007/s00109-023-02349-w] [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: 01/18/2023] [Revised: 06/14/2023] [Accepted: 07/03/2023] [Indexed: 08/19/2023]
Abstract
Psoriasis is a chronic inflammatory skin disease driven by the IL-23/IL-17 axis. It results from excessive activation of effector T cells, including T helper (Th) and cytotoxic T (Tc) cells, and is associated with dysfunctional regulatory T cells (Tregs). Acetyl-CoA carboxylase 1 (ACC1), a rate-limiting enzyme of fatty acid synthesis (FAS), directs cell fate decisions between Th17 and Tregs and thus could be a promising therapeutic target for psoriasis treatment. Here, we demonstrate that targeting ACC1 in T cells by genetic ablation ameliorates skin inflammation in an experimental model of psoriasis by limiting Th17, Tc17, Th1, and Tc1 cells in skin lesions and increasing the frequency of effector Tregs in skin-draining lymph nodes (LNs). KEY MESSAGES : ACC1 deficiency in T cells ameliorates psoriatic skin inflammation in mice. ACC1 deficiency in T cells reduces IL-17A-producing Th17/Tc17/dysfunctional Treg populations in psoriatic lesions. ACC1 deficiency in T cells restrains IFN-γ-producing Th1/Tc1 populations in psoriatic skin lesions and skin-draining LNs. ACC1 deficiency promotes activated CD44+CD25+ Tregs and effector CD62L-CD44+ Tregs under homeostasis and psoriatic conditions.
Collapse
Affiliation(s)
- Yu-San Kao
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Panagiota Mamareli
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ayesha Dhillon-LaBrooy
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Philipp Stüve
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Leibniz Institute for Immunotherapy, University Regensburg, 93053, Regensburg, Germany
- Chair for Immunology, University Regensburg, 93053, Regensburg, Germany
| | - Gloria Janet Godoy
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lis Noelia Velasquez
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Verena Katharina Raker
- Department of Dermatology, University Hospital, Westfälische Wilhelms-University Münster, Münster, Germany
| | - Beate Weidenthaler-Barth
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Fatima Boukhallouk
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Francesca Rampoldi
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Luciana Berod
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany.
| |
Collapse
|
72
|
Hinkley H, Counts DA, VonCanon E, Lacy M. T Cells in Atherosclerosis: Key Players in the Pathogenesis of Vascular Disease. Cells 2023; 12:2152. [PMID: 37681883 PMCID: PMC10486666 DOI: 10.3390/cells12172152] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipid-rich plaques within arterial walls. T cells play a pivotal role in the pathogenesis of atherosclerosis in which they help orchestrate immune responses and contribute to plaque development and instability. Here, we discuss the recognition of atherosclerosis-related antigens that may trigger T cell activation together with additional signaling from co-stimulatory molecules and lesional cytokines. Although few studies have indicated candidates for the antigen specificity of T cells in atherosclerosis, further research is needed. Furthermore, we describe the pro-atherogenic and atheroprotective roles of diverse subsets of T cells such as CD4+ helper, CD8+ cytotoxic, invariant natural killer, and γδ T cells. To classify and quantify T cell subsets in atherosclerosis, we summarize current methods to analyze cellular heterogeneity including single cell RNA sequencing and T cell receptor (TCR) sequencing. Further insights into T cell biology will help shed light on the immunopathology of atherosclerosis, inform potential therapeutic interventions, and pave the way for precision medicine approaches in combating cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | - Michael Lacy
- Department of Medical Laboratory Sciences, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
73
|
TeSlaa T, Ralser M, Fan J, Rabinowitz JD. The pentose phosphate pathway in health and disease. Nat Metab 2023; 5:1275-1289. [PMID: 37612403 PMCID: PMC11251397 DOI: 10.1038/s42255-023-00863-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 07/12/2023] [Indexed: 08/25/2023]
Abstract
The pentose phosphate pathway (PPP) is a glucose-oxidizing pathway that runs in parallel to upper glycolysis to produce ribose 5-phosphate and nicotinamide adenine dinucleotide phosphate (NADPH). Ribose 5-phosphate is used for nucleotide synthesis, while NADPH is involved in redox homoeostasis as well as in promoting biosynthetic processes, such as the synthesis of tetrahydrofolate, deoxyribonucleotides, proline, fatty acids and cholesterol. Through NADPH, the PPP plays a critical role in suppressing oxidative stress, including in certain cancers, in which PPP inhibition may be therapeutically useful. Conversely, PPP-derived NADPH also supports purposeful cellular generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) for signalling and pathogen killing. Genetic deficiencies in the PPP occur relatively commonly in the committed pathway enzyme glucose-6-phosphate dehydrogenase (G6PD). G6PD deficiency typically manifests as haemolytic anaemia due to red cell oxidative damage but, in severe cases, also results in infections due to lack of leucocyte oxidative burst, highlighting the dual redox roles of the pathway in free radical production and detoxification. This Review discusses the PPP in mammals, covering its roles in biochemistry, physiology and disease.
Collapse
Affiliation(s)
- Tara TeSlaa
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Markus Ralser
- Department of Biochemistry, Charité Universitätsmedizin, Berlin, Germany
- The Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Jing Fan
- Morgride Institute for Research, Madison, WI, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua D Rabinowitz
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Chemistry, Princeton University, Princeton, NJ, USA.
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
74
|
Wu M, Fu X, Xu R, Liu S, Li R, Xu J, Shang W, Chen X, Wang T, Wang F. Glucose metabolism and function of CD4 + Tregs are regulated by the TLR8/mTOR signal in an environment of SKOV3 cell growth. Cancer Med 2023; 12:16310-16322. [PMID: 37317670 PMCID: PMC10469653 DOI: 10.1002/cam4.6247] [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: 12/01/2022] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023] Open
Abstract
PURPOSE To investigate the role of mammalian target of rapamycin (mTOR) signal in Toll-like receptor (TLR) 8-mediated regulation of glucose metabolism and its effect on reversing immunosuppression in CD4+ regulatory T-cells (Tregs) in ovarian cancer (OC). METHODS Fluorescence-activated cell sorting was used to detect the expression levels of mTOR+ and 4E-BP1+ cells in CD4+ Tregs. The prognosis and immune infiltration analysis of mTOR mRNA in OC were performed using the TIMER and Kaplan-Meier plotter database. Furthermore, real-time polymerase chain reaction (RT-PCR) and western blot (WB) were used to detect expression levels of glucose metabolism-related genes and proteins in CD4+ Tregs. Glucose uptake and glycolysis levels were detected by colorimetry, while the effects of CD4+ Tregs on the proliferation of CD4+ T-effector cells (Teffs) were evaluated by carboxyfluorescein diacetate succinimidyl ester (CFSE). RESULTS mTOR expression in CD4+ Tregs was significantly higher in patients with OC compared with controls and in CD4+ Tregs than in CD4+ Teffs in OC. Additionally, the expression level of mTOR mRNA was related to prognosis and immune infiltration levels in patients with OC. Blocking the mTOR signal resulted in downregulation of glucose metabolism in CD4+ Tregs. Simultaneous inhibition of the mTOR signal while activation of the TLR8 signal had a coordinated inhibitory effect on glucose metabolism and the immunosuppressive function of CD4+ Tregs. Furthermore, the mTOR signal played an essential role in TLR8-mediated reversal of immunosuppressive function in CD4+ Tregs. CONCLUSION These findings imply that activation of the TLR8 signal inhibits glucose metabolism in CD4+ Tregs by downregulating mTOR signaling, thereby reversing the immunosuppressive function of these cells in an OC cell growth environment.
Collapse
Affiliation(s)
- Ming Wu
- Department of Laboratory Medicinethe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
- Department of Clinical LaboratoryChildren’s Hospital of Fudan University, National Children’s Medical CenterShanghaiChina
| | - Xin Fu
- Department of Laboratory Medicinethe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Rui Xu
- Department of Laboratory Medicinethe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
- Department of AnesthesiologyNanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Shuna Liu
- Department of Laboratory Medicinethe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Rong Li
- Department of Laboratory Medicinethe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
- Department of GynecologyWomen’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital)NanjingChina
| | - Juan Xu
- Department of Laboratory Medicinethe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Wenwen Shang
- Department of Laboratory Medicinethe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Xian Chen
- Department of Laboratory Medicinethe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Ting Wang
- Department of Laboratory Medicinethe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Fang Wang
- Department of Laboratory Medicinethe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| |
Collapse
|
75
|
Korn T. Foxp3 + regulatory T cells in the central nervous system and other nonlymphoid tissues. Eur J Immunol 2023; 53:e2250227. [PMID: 37143298 DOI: 10.1002/eji.202250227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/23/2023] [Accepted: 05/02/2023] [Indexed: 05/06/2023]
Abstract
Foxp3+ regulatory T (Treg) cells are indispensable for the maintenance of immunologic self-tolerance as well as for the confinement of autoimmune inflammation after the breach of self-tolerance. In order to fulfill these tasks, Treg cells operate in secondary lymphoid tissues and nonlymphoid tissues. The conditions for Treg cell stability and for their modes of action are different according to their compartment of residence. In addition, Treg cells initiate residency programs to inhabit niches in nonlympoid tissues (NLT) in steady state and after re-establishment of previously deflected homeostasis for extended periods of time. These NLT Treg cells are different from lymphoid tissue residing Treg cells and are functionally specialized to subserve not only immune functions but support intrinsic functions of their tissue of residence. This review will highlight current ideas about the functional specialization of NLT Treg cells in particular in the central nervous system (CNS) and discuss challenges that we are facing in an effort to exploit the power of NLT Treg cells for maintenance of tissue homeostasis and perhaps also tissue regeneration.
Collapse
Affiliation(s)
- Thomas Korn
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine, Munich, Germany
- Department of Neurology, Technical University of Munich School of Medicine, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
76
|
Jeong H, Lee B, Han SJ, Sohn DH. Glucose metabolic reprogramming in autoimmune diseases. Anim Cells Syst (Seoul) 2023; 27:149-158. [PMID: 37465289 PMCID: PMC10351453 DOI: 10.1080/19768354.2023.2234986] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/20/2023] Open
Abstract
Autoimmune diseases are conditions in which the immune system mistakenly targets and damages healthy tissue in the body. In recent decades, the incidence of autoimmune diseases has increased, resulting in a significant disease burden. The current autoimmune therapies focus on targeting inflammation or inducing immunosuppression rather than addressing the underlying cause of the diseases. The activity of metabolic pathways is elevated in autoimmune diseases, and metabolic changes are increasingly recognized as important pathogenic processes underlying these. Therefore, metabolically targeted therapies may represent an important strategy for treating autoimmune diseases. This review provides a comprehensive overview of the evidence surrounding glucose metabolic reprogramming and its potential applications in drug discovery and development for autoimmune diseases, such as type 1 diabetes, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, and systemic sclerosis.
Collapse
Affiliation(s)
- Hoim Jeong
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Beomgu Lee
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Seung Jin Han
- Department of Medical Biotechnology, Inje University, Gimhae, Republic of Korea
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| |
Collapse
|
77
|
Wang Y, Huang T, Gu J, Lu L. Targeting the metabolism of tumor-infiltrating regulatory T cells. Trends Immunol 2023:S1471-4906(23)00109-6. [PMID: 37442660 DOI: 10.1016/j.it.2023.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023]
Abstract
Although targeting the tumor metabolism is performed in cooperation with immunotherapy in the era of precision oncology, ignorance of immune cells' metabolism has resulted in unstable antitumor responses. Tumor-infiltrating regulatory T cells (TI-Tregs) are unique, overcoming the hypoxic, acidic, and nutrient-deficient tumor microenvironments (TMEs) and maintaining immunosuppressive functions. However, secondary autoimmunity caused by systemic Treg depletion remains the 'Sword of Damocles' for current Treg-targeted therapies. In this opinion piece, we propose that metabolically reprogrammed TI-Tregs might represent an obstacle to cancer therapies. Indeed, metabolism-based Treg-targeted therapy might provide higher selectivity for clearing TI-Tregs than traditional kinase/checkpoint inhibitors and chemokine/chemokine receptor blockade; it might also restore the efficacy of targeting the tumor metabolism and eliminate certain metabolic barriers to immunotherapy.
Collapse
Affiliation(s)
- Yiming Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Tianning Huang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Jian Gu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Ling Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
78
|
Wu H, Huang H, Zhao Y. Interplay between metabolic reprogramming and post-translational modifications: from glycolysis to lactylation. Front Immunol 2023; 14:1211221. [PMID: 37457701 PMCID: PMC10338923 DOI: 10.3389/fimmu.2023.1211221] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Cellular metabolism plays a critical role in determining the fate and function of cells. Metabolic reprogramming and its byproducts have a complex impact on cellular activities. In quiescent T cells, oxidative phosphorylation (OXPHOS) is the primary pathway for survival. However, upon antigen activation, T cells undergo rapid metabolic reprogramming, characterized by an elevation in both glycolysis and OXPHOS. While both pathways are induced, the balance predominantly shifts towards glycolysis, enabling T cells to rapidly proliferate and enhance their functionality, representing the most distinctive signature during activation. Metabolic processes generate various small molecules resulting from enzyme-catalyzed reactions, which also modulate protein function and exert regulatory control. Notably, recent studies have revealed the direct modification of histones, known as lactylation, by lactate derived from glycolysis. This lactylation process influences gene transcription and adds a novel variable to the regulation of gene expression. Protein lactylation has been identified as an essential mechanism by which lactate exerts its diverse functions, contributing to crucial biological processes such as uterine remodeling, tumor proliferation, neural system regulation, and metabolic regulation. This review focuses on the metabolic reprogramming of T cells, explores the interplay between lactate and the immune system, highlights the impact of lactylation on cellular function, and elucidates the intersection of metabolic reprogramming and epigenetics.
Collapse
Affiliation(s)
- Hengwei Wu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, People's Government of Zhejiang Province, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, People's Government of Zhejiang Province, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Yanmin Zhao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, People's Government of Zhejiang Province, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| |
Collapse
|
79
|
Wang Y, Wang C, Shen L, Xu D. The Role of Regulatory T Cells in Heart Repair After Myocardial Infarction. J Cardiovasc Transl Res 2023:10.1007/s12265-022-10290-5. [PMID: 37347425 DOI: 10.1007/s12265-022-10290-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/04/2022] [Indexed: 06/23/2023]
Abstract
Myocardial infarction (MI) remains one of the leading causes of death worldwide. Inflammation and immune responses after MI are of significance to the adverse cardiac remodeling. Regulatory T cells (Tregs) play an important role in suppressing the immune response and thus benefit the post-MI remodeling. After MI, damaged cardiomyocytes may be replaced by scar tissue, leading to systolic and diastolic dysfunction and subsequently adverse remodeling. In this review, we provide an overview of the function and possible mechanisms of Tregs in post-MI heart repair. Specifically, after the occurrence of MI, Tregs infiltrated to peri-infarcted myocardium through CCR5 pathway, CXCR4-CXCL12 axis, and Hippo pathway. Normal functional Tregs can reduce the size of the MI area, improve heart function, and ameliorate myocardial remodeling by inhibiting proinflammatory cells accumulation, changing the proportion of macrophages phenotypes, improving myocardial fibrosis, protecting myocardial cells, and promoting angiogenesis. Eventually, Functional Tregs recruited into the heart can improve MI outcomes. Therefore, targeted therapies with Tregs might provide a promising approach to the treatment of MI remodeling.
Collapse
Affiliation(s)
- Yishu Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, China
| | - Chunfang Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, China
| | - Li Shen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan, China.
| |
Collapse
|
80
|
Yang L, Chu Z, Liu M, Zou Q, Li J, Liu Q, Wang Y, Wang T, Xiang J, Wang B. Amino acid metabolism in immune cells: essential regulators of the effector functions, and promising opportunities to enhance cancer immunotherapy. J Hematol Oncol 2023; 16:59. [PMID: 37277776 DOI: 10.1186/s13045-023-01453-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/13/2023] [Indexed: 06/07/2023] Open
Abstract
Amino acids are basic nutrients for immune cells during organ development, tissue homeostasis, and the immune response. Regarding metabolic reprogramming in the tumor microenvironment, dysregulation of amino acid consumption in immune cells is an important underlying mechanism leading to impaired anti-tumor immunity. Emerging studies have revealed that altered amino acid metabolism is tightly linked to tumor outgrowth, metastasis, and therapeutic resistance through governing the fate of various immune cells. During these processes, the concentration of free amino acids, their membrane bound transporters, key metabolic enzymes, and sensors such as mTOR and GCN2 play critical roles in controlling immune cell differentiation and function. As such, anti-cancer immune responses could be enhanced by supplement of specific essential amino acids, or targeting the metabolic enzymes or their sensors, thereby developing novel adjuvant immune therapeutic modalities. To further dissect metabolic regulation of anti-tumor immunity, this review summarizes the regulatory mechanisms governing reprogramming of amino acid metabolism and their effects on the phenotypes and functions of tumor-infiltrating immune cells to propose novel approaches that could be exploited to rewire amino acid metabolism and enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Luming Yang
- Chongqing University Medical School, Chongqing, 400044, People's Republic of China
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Zhaole Chu
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Meng Liu
- Chongqing University Medical School, Chongqing, 400044, People's Republic of China
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Qiang Zou
- Chongqing University Medical School, Chongqing, 400044, People's Republic of China
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Jinyang Li
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Qin Liu
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Yazhou Wang
- Chongqing University Medical School, Chongqing, 400044, People's Republic of China.
| | - Tao Wang
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China.
| | - Junyu Xiang
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China.
| | - Bin Wang
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China.
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China.
- Jinfeng Laboratory, Chongqing, 401329, People's Republic of China.
| |
Collapse
|
81
|
Iacobini C, Vitale M, Pugliese G, Menini S. The "sweet" path to cancer: focus on cellular glucose metabolism. Front Oncol 2023; 13:1202093. [PMID: 37305566 PMCID: PMC10248238 DOI: 10.3389/fonc.2023.1202093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/17/2023] [Indexed: 06/13/2023] Open
Abstract
The hypoxia-inducible factor-1α (HIF-1α), a key player in the adaptive regulation of energy metabolism, and the M2 isoform of the glycolytic enzyme pyruvate kinase (PKM2), a critical regulator of glucose consumption, are the main drivers of the metabolic rewiring in cancer cells. The use of glycolysis rather than oxidative phosphorylation, even in the presence of oxygen (i.e., Warburg effect or aerobic glycolysis), is a major metabolic hallmark of cancer. Aerobic glycolysis is also important for the immune system, which is involved in both metabolic disorders development and tumorigenesis. More recently, metabolic changes resembling the Warburg effect have been described in diabetes mellitus (DM). Scientists from different disciplines are looking for ways to interfere with these cellular metabolic rearrangements and reverse the pathological processes underlying their disease of interest. As cancer is overtaking cardiovascular disease as the leading cause of excess death in DM, and biological links between DM and cancer are incompletely understood, cellular glucose metabolism may be a promising field to explore in search of connections between cardiometabolic and cancer diseases. In this mini-review, we present the state-of-the-art on the role of the Warburg effect, HIF-1α, and PKM2 in cancer, inflammation, and DM to encourage multidisciplinary research to advance fundamental understanding in biology and pathways implicated in the link between DM and cancer.
Collapse
|
82
|
Henschel P, Landwehr-Kenzel S, Engels N, Schienke A, Kremer J, Riet T, Redel N, Iordanidis K, Saetzler V, John K, Heider M, Hardtke-Wolenski M, Wedemeyer H, Jaeckel E, Noyan F. Supraphysiological FOXP3 expression in human CAR-Tregs results in improved stability, efficacy, and safety of CAR-Treg products for clinical application. J Autoimmun 2023; 138:103057. [PMID: 37224732 DOI: 10.1016/j.jaut.2023.103057] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/26/2023]
Abstract
The forkhead family transcription factor (FOXP3) is an essential regulator for the development of regulatory T cells (Tregs) and orchestrates both suppressive function and Treg lineage identity. Stable expression of FOXP3 enables Tregs to maintain immune homeostasis and prevent autoimmunity. However, under pro-inflammatory conditions, FOXP3 expression in Tregs can become unstable, leading to loss of suppressive function and conversion into pathogenic T effector cells. Therefore, the success of adoptive cell therapy with chimeric antigen receptor (CAR) Tregs is highly dependent on the stability of FOXP3 expression to ensure the safety of the cell product. To warrant the stable expression of FOXP3 in CAR-Treg products, we have developed an HLA-A2-specific CAR vector that co-expresses FOXP3. The transduction of isolated human Tregs with the FOXP3-CAR led to an increase in the safety and efficacy of the CAR-Treg product. In a hostile microenvironment, under pro-inflammatory and IL-2-deficient conditions, FOXP3-CAR-Tregs showed a stable expression of FOXP3 compared to Control-CAR-Tregs. Furthermore, additional exogenous expression of FOXP3 did not induce phenotypic alterations and dysfunctions such as cell exhaustion, loss of functional Treg characteristics or abnormal cytokine secretion. In a humanized mouse model, FOXP3-CAR-Tregs displayed an excellent ability to prevent allograft rejection. Furthermore, FOXP3-CAR-Tregs revealed coherent Treg niche-filling capabilities. Overexpression of FOXP3 in CAR-Tregs has thereby the potential to increase the efficacy and reliability of cellular products, promoting their clinical use in organ transplantation and autoimmune diseases.
Collapse
Affiliation(s)
- Pierre Henschel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Sybille Landwehr-Kenzel
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Niklas Engels
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, Goettingen, Germany
| | - Andrea Schienke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jakob Kremer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Tobias Riet
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Department I of Internal Medicine, Tumor Genetics, University Hospital of Cologne and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Nella Redel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Konstantinos Iordanidis
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Valerie Saetzler
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Katharina John
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Miriam Heider
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Matthias Hardtke-Wolenski
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Institute of Medical Microbiology, Essen University Hospital, University Duisburg-Essen, Essen, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Department of Liver Transplantation, Multi Organ Transplant Program, University Health Network, Toronto, University of Toronto, Canada
| | - Fatih Noyan
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
83
|
Zhang W, Chen Y, Zhao Z, Zheng H, Wang S, Liao Z, Sheng T, Zhao S, Hou W, Yu X, He F, Yu J, Zhang Y, Gu Z. Adoptive T reg therapy with metabolic intervention via perforated microneedles ameliorates psoriasis syndrome. SCIENCE ADVANCES 2023; 9:eadg6007. [PMID: 37196084 DOI: 10.1126/sciadv.adg6007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/14/2023] [Indexed: 05/19/2023]
Abstract
Regulatory T (Treg) cells underlie multiple autoimmune disorders and potentialize an anti-inflammation treatment with adoptive cell therapy. However, systemic delivery of cellular therapeutics often lacks tissue targeting and accumulation for localized autoimmune diseases. Besides, the instability and plasticity of Treg cells also induce phenotype transition and functional loss, impeding clinical translation. Here, we developed a perforated microneedle (PMN) with favorable mechanical performance and a spacious encapsulation cavity to support cell survival, as well as tunable channels to facilitate cell migration for local Treg therapy of psoriasis. In addition, the enzyme-degradable microneedle matrix could release fatty acid in the hyperinflammatory area of psoriasis, enhancing the Treg suppressive functions via the fatty acid oxidation (FAO)-mediated metabolic intervention. Treg cells administered through PMN substantially ameliorated psoriasis syndrome with the assistance of fatty acid-mediated metabolic intervention in a psoriasis mouse model. This tailorable PMN could offer a transformative platform for local cell therapy to treat a variety of diseases.
Collapse
Affiliation(s)
- Wentao Zhang
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yingxin Chen
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Institute of Advanced Magnetic Materials and International Research Center for EM Metamaterials, College of Materials and Environmental Engineering, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Zhengjie Zhao
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hanqi Zheng
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shenqiang Wang
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ziyan Liao
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tao Sheng
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sheng Zhao
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenhui Hou
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinmin Yu
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fang He
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Jicheng Yu
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yuqi Zhang
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Zhen Gu
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
84
|
Kumar V, Stewart JH. Immunometabolic reprogramming, another cancer hallmark. Front Immunol 2023; 14:1125874. [PMID: 37275901 PMCID: PMC10235624 DOI: 10.3389/fimmu.2023.1125874] [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: 12/16/2022] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Molecular carcinogenesis is a multistep process that involves acquired abnormalities in key biological processes. The complexity of cancer pathogenesis is best illustrated in the six hallmarks of the cancer: (1) the development of self-sufficient growth signals, (2) the emergence of clones that are resistant to apoptosis, (3) resistance to the antigrowth signals, (4) neo-angiogenesis, (5) the invasion of normal tissue or spread to the distant organs, and (6) limitless replicative potential. It also appears that non-resolving inflammation leads to the dysregulation of immune cell metabolism and subsequent cancer progression. The present article delineates immunometabolic reprogramming as a critical hallmark of cancer by linking chronic inflammation and immunosuppression to cancer growth and metastasis. We propose that targeting tumor immunometabolic reprogramming will lead to the design of novel immunotherapeutic approaches to cancer.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), New Orleans, LA, United States
| | - John H. Stewart
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), New Orleans, LA, United States
- Louisiana State University- Louisiana Children’s Medical Center, Stanley S. Scott, School of Medicine, Louisiana State University Health Science Center (LSUHSC), New Orleans, LA, United States
| |
Collapse
|
85
|
Yang Y, Hu P, Chen SR, Wu WW, Chen P, Wang SW, Ma JZ, Hu JY. Predicting the Activity of Oral Lichen Planus with Glycolysis-related Molecules: A Scikit-learn-based Function. Curr Med Sci 2023:10.1007/s11596-023-2716-7. [PMID: 37115394 PMCID: PMC10141813 DOI: 10.1007/s11596-023-2716-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/09/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVE Oral lichen planus (OLP) is one of the most common oral mucosa diseases, and is mainly mediated by T lymphocytes. The metabolic reprogramming of activated T cells has been shown to transform from oxidative phosphorylation to aerobic glycolysis. The present study investigated the serum levels of glycolysis-related molecules (lactate dehydrogenase, LDH; pyruvic acid, PA; lactic acid, LAC) in OLP, and the correlation with OLP activity was assessed using the reticular, atrophic and erosive lesion (RAE) scoring system. METHODS Univariate and multivariate linear regression functions based on scikit-learn were designed to predict the RAE scores in OLP patients, and the performance of these two machine learning functions was compared. RESULTS The results revealed that the serum levels of PA and LAC were upregulated in erosive OLP (EOLP) patients, when compared to healthy volunteers. Furthermore, the LDH and LAC levels were significantly higher in the EOLP group than in the nonerosive OLP (NEOLP) group. All glycolysis-related molecules were positively correlated to the RAE scores. Among these, LAC had a strong correlation. The univariate function that involved the LAC level and the multivariate function that involved all glycolysis-related molecules presented comparable prediction accuracy and stability, but the latter was more time-consuming. CONCLUSION It can be concluded that the serum LAC level can be a user-friendly biomarker to monitor the OLP activity, based on the univariate function developed in the present study. The intervention of the glycolytic pathway may provide a potential therapeutic strategy.
Collapse
Affiliation(s)
- Yan Yang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pei Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Su-Rong Chen
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei-Wei Wu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pan Chen
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shi-Wen Wang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing-Zhi Ma
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing-Yu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
86
|
van Hooren L, Handgraaf SM, Kloosterman DJ, Karimi E, van Mil LWHG, Gassama AA, Solsona BG, de Groot MHP, Brandsma D, Quail DF, Walsh LA, Borst GR, Akkari L. CD103 + regulatory T cells underlie resistance to radio-immunotherapy and impair CD8 + T cell activation in glioblastoma. NATURE CANCER 2023; 4:665-681. [PMID: 37081259 DOI: 10.1038/s43018-023-00547-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 03/20/2023] [Indexed: 04/22/2023]
Abstract
Glioblastomas are aggressive primary brain tumors with an inherent resistance to T cell-centric immunotherapy due to their low mutational burden and immunosuppressive tumor microenvironment. Here we report that fractionated radiotherapy of preclinical glioblastoma models induce a tenfold increase in T cell content. Orthogonally, spatial imaging mass cytometry shows T cell enrichment in human recurrent tumors compared with matched primary glioblastoma. In glioblastoma-bearing mice, α-PD-1 treatment applied at the peak of T cell infiltration post-radiotherapy results in a modest survival benefit compared with concurrent α-PD-1 administration. Following α-PD-1 therapy, CD103+ regulatory T cells (Tregs) with upregulated lipid metabolism accumulate in the tumor microenvironment, and restrain immune checkpoint blockade response by repressing CD8+ T cell activation. Treg targeting elicits tertiary lymphoid structure formation, enhances CD4+ and CD8+ T cell frequency and function and unleashes radio-immunotherapeutic efficacy. These results support the rational design of therapeutic regimens limiting the induction of immunosuppressive feedback pathways in the context of T cell immunotherapy in glioblastoma.
Collapse
Affiliation(s)
- Luuk van Hooren
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Shanna M Handgraaf
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Daan J Kloosterman
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Elham Karimi
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Lotte W H G van Mil
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Awa A Gassama
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Beatriz Gomez Solsona
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marnix H P de Groot
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Dieta Brandsma
- Department of Neuro-Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Gerben R Borst
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, UK.
- Department of Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, UK.
| | - Leila Akkari
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
87
|
Lamarche C, Ward-Hartstonge K, Mi T, Lin DTS, Huang Q, Brown A, Edwards K, Novakovsky GE, Qi CN, Kobor MS, Zebley CC, Weber EW, Mackall CL, Levings MK. Tonic-signaling chimeric antigen receptors drive human regulatory T cell exhaustion. Proc Natl Acad Sci U S A 2023; 120:e2219086120. [PMID: 36972454 PMCID: PMC10083618 DOI: 10.1073/pnas.2219086120] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/24/2023] [Indexed: 03/29/2023] Open
Abstract
Regulatory T cell (Treg) therapy is a promising approach to improve outcomes in transplantation and autoimmunity. In conventional T cell therapy, chronic stimulation can result in poor in vivo function, a phenomenon termed exhaustion. Whether or not Tregs are also susceptible to exhaustion, and if so, if this would limit their therapeutic effect, was unknown. To "benchmark" exhaustion in human Tregs, we used a method known to induce exhaustion in conventional T cells: expression of a tonic-signaling chimeric antigen receptor (TS-CAR). We found that TS-CAR-expressing Tregs rapidly acquired a phenotype that resembled exhaustion and had major changes in their transcriptome, metabolism, and epigenome. Similar to conventional T cells, TS-CAR Tregs upregulated expression of inhibitory receptors and transcription factors such as PD-1, TIM3, TOX and BLIMP1, and displayed a global increase in chromatin accessibility-enriched AP-1 family transcription factor binding sites. However, they also displayed Treg-specific changes such as high expression of 4-1BB, LAP, and GARP. DNA methylation analysis and comparison to a CD8+ T cell-based multipotency index showed that Tregs naturally exist in a relatively differentiated state, with further TS-CAR-induced changes. Functionally, TS-CAR Tregs remained stable and suppressive in vitro but were nonfunctional in vivo, as tested in a model of xenogeneic graft-versus-host disease. These data are the first comprehensive investigation of exhaustion in Tregs and reveal key similarities and differences with exhausted conventional T cells. The finding that human Tregs are susceptible to chronic stimulation-driven dysfunction has important implications for the design of CAR Treg adoptive immunotherapy strategies.
Collapse
Affiliation(s)
- Caroline Lamarche
- Department of Surgery, University of British Columbia, VancouverV6T 1Z4, BC, Canada
- BC Children’s Hospital Research Institute, VancouverV5Z 4H4, BC, Canada
- Department of Medicine, Hôpital Maisonneuve-Rosemont Research Center, Université de Montréal, MontrealH1T 2M4, QC, Canada
| | - Kirsten Ward-Hartstonge
- Department of Surgery, University of British Columbia, VancouverV6T 1Z4, BC, Canada
- BC Children’s Hospital Research Institute, VancouverV5Z 4H4, BC, Canada
- Department of Microbiology and Immunology, University of Otago, Dunedin9016, New Zealand
| | - Tian Mi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - David T. S. Lin
- BC Children’s Hospital Research Institute, VancouverV5Z 4H4, BC, Canada
- Department of Medical Genetics, University of British Columbia, VancouverV6T 1Z4, BC, Canada
| | - Qing Huang
- Department of Surgery, University of British Columbia, VancouverV6T 1Z4, BC, Canada
- BC Children’s Hospital Research Institute, VancouverV5Z 4H4, BC, Canada
| | - Andrew Brown
- BC Children’s Hospital Research Institute, VancouverV5Z 4H4, BC, Canada
- School of Biomedical Engineering, University of British Columbia, VancouverV6T 1Z4, BC, Canada
| | - Karlie Edwards
- BC Children’s Hospital Research Institute, VancouverV5Z 4H4, BC, Canada
- Department of Medical Genetics, University of British Columbia, VancouverV6T 1Z4, BC, Canada
| | - Gherman E. Novakovsky
- BC Children’s Hospital Research Institute, VancouverV5Z 4H4, BC, Canada
- Department of Medical Genetics, University of British Columbia, VancouverV6T 1Z4, BC, Canada
| | - Christopher N. Qi
- Department of Surgery, University of British Columbia, VancouverV6T 1Z4, BC, Canada
- BC Children’s Hospital Research Institute, VancouverV5Z 4H4, BC, Canada
| | - Michael S. Kobor
- BC Children’s Hospital Research Institute, VancouverV5Z 4H4, BC, Canada
- Department of Medical Genetics, University of British Columbia, VancouverV6T 1Z4, BC, Canada
| | - Caitlin C. Zebley
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN38105
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Evan W. Weber
- Division of Oncology, Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA19104
| | - Crystal L. Mackall
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA94305
- Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Megan K Levings
- Department of Surgery, University of British Columbia, VancouverV6T 1Z4, BC, Canada
- BC Children’s Hospital Research Institute, VancouverV5Z 4H4, BC, Canada
- School of Biomedical Engineering, University of British Columbia, VancouverV6T 1Z4, BC, Canada
| |
Collapse
|
88
|
Chaudhary S, Rai R, Pal PB, Tedesco D, Singhi AD, Monga SP, Grakoui A, Iyer SS, Raeman R. Western diet dampens T regulatory cell function to fuel hepatic inflammation in nonalcoholic fatty liver disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.23.533977. [PMID: 36993495 PMCID: PMC10055333 DOI: 10.1101/2023.03.23.533977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Background and aims The immunosuppressive T regulatory cells (Tregs) regulate immune responses and maintain immune homeostasis, yet their functions in nonalcoholic fatty liver disease (NAFLD) pathogenesis remains controversial. Methods Mice were fed a normal diet (ND) or a western diet (WD) for 16 weeks to induce NAFLD. Diphtheria toxin injection to deplete Tregs in Foxp3 DTR mice or Treg induction therapy in WT mice to augment Treg numbers was initiated at twelve and eight weeks, respectively. Liver tissues from mice and NASH human subjects were analyzed by histology, confocal imaging, and qRT-PCR. Results WD triggered accumulation of adaptive immune cells, including Tregs and effector T cells, within the liver parenchyma. This pattern was also observed in NASH patients, where an increase in intrahepatic Tregs was noted. In the absence of adaptive immune cells in Rag1 KO mice, WD promoted accumulation of intrahepatic neutrophils and macrophages and exacerbated hepatic inflammation and fibrosis. Similarly, targeted Treg depletion exacerbated WD-induced hepatic inflammation and fibrosis. In Treg-depleted mice, hepatic injury was associated with increased accumulation of neutrophils, macrophages, and activated T cells within the liver. Conversely, induction of Tregs using recombinant IL2/αIL2 mAb cocktail reduced hepatic steatosis, inflammation, and fibrosis in WD-fed mice. Analysis of intrahepatic Tregs from WD-fed mice revealed a phenotypic signature of impaired Treg function in NAFLD. Ex vivo functional studies showed that glucose and palmitate, but not fructose, impaired the immunosuppressive ability of Treg cells. Conclusions Our findings indicate that the liver microenvironment in NAFLD impairs ability of Tregs to suppress effector immune cell activation, thus perpetuating chronic inflammation and driving NAFLD progression. These data suggest that targeted approaches aimed at restoring Treg function may represent a potential therapeutic strategy for treating NAFLD. Lay summary In this study, we elucidate the mechanisms contributing to the perpetuation of chronic hepatic inflammation in nonalcoholic fatty liver disease (NAFLD). We show that dietary sugar and fatty acids promote chronic hepatic inflammation in NAFLD by impairing immunosuppressive function of regulatory T cells. Finally, our preclinical data suggest that targeted approaches aimed at restoring T regulatory cell function have the potential to treat NAFLD.
Collapse
|
89
|
Gu Q, Tung KS, Lorenz UM. Treg-specific deletion of the phosphatase SHP-1 impairs control of inflammation in vivo. Front Immunol 2023; 14:1139326. [PMID: 37006301 PMCID: PMC10060847 DOI: 10.3389/fimmu.2023.1139326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction To achieve a healthy and functional immune system, a delicate balance exists between the activation of conventional T cells (Tcon cells) and the suppression by regulatory T cells (Treg). The tyrosine phosphatase SHP-1, a negative regulator of TCR signaling, shapes this 'activation-suppression' balance by modulating Tcon cell resistance to Treg-mediated suppression. Treg cells also express SHP-1, but its role in influencing Treg function is still not fully understood. Methods We generated a Treg-specific SHP-1 deletion model, Foxp3Cre+ Shp-1f/f , to address how SHP-1 affects Treg function and thereby contributes to T cell homeostasis using a combination of ex vivo studies and in vivo models of inflammation and autoimmunity. Results We show that SHP-1 modulates Treg suppressive function at different levels. First, at the intracellular signaling level in Treg cells, SHP-1 attenuates TCR-dependent Akt phosphorylation, with loss of SHP-1 driving Treg cells towards a glycolysis pathway. At the functional level, SHP-1 expression limits the in vivo accumulation of CD44hiCD62Llo T cells within the steady state Tcon populations (both CD8+ as well as CD4+ Tcon). Further, SHP-1-deficient Treg cells are less efficient in suppressing inflammation in vivo; mechanistically, this appears to be due to a failure to survive or a defect in migration of SHP-1-deficient Treg cells to peripheral inflammation sites. Conclusion Our data identify SHP-1 as an important intracellular mediator for fine-tuning the balance between Treg-mediated suppression and Tcon activation/resistance.
Collapse
Affiliation(s)
- QinLei Gu
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Kenneth S. Tung
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Ulrike M. Lorenz
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Pathology and Immunology, Washington University in St. Louis, Saint Louis, MO, United States
| |
Collapse
|
90
|
Arner EN, Rathmell JC. Metabolic programming and immune suppression in the tumor microenvironment. Cancer Cell 2023; 41:421-433. [PMID: 36801000 PMCID: PMC10023409 DOI: 10.1016/j.ccell.2023.01.009] [Citation(s) in RCA: 174] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/18/2023]
Abstract
Increased glucose metabolism and uptake are characteristic of many tumors and used clinically to diagnose and monitor cancer progression. In addition to cancer cells, the tumor microenvironment (TME) encompasses a wide range of stromal, innate, and adaptive immune cells. Cooperation and competition between these cell populations supports tumor proliferation, progression, metastasis, and immune evasion. Cellular heterogeneity leads to metabolic heterogeneity because metabolic programs within the tumor are dependent not only on the TME cellular composition but also on cell states, location, and nutrient availability. In addition to driving metabolic plasticity of cancer cells, altered nutrients and signals in the TME can lead to metabolic immune suppression of effector cells and promote regulatory immune cells. Here we discuss how metabolic programming of cells within the TME promotes tumor proliferation, progression, and metastasis. We also discuss how targeting metabolic heterogeneity may offer therapeutic opportunities to overcome immune suppression and augment immunotherapies.
Collapse
Affiliation(s)
- Emily N Arner
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Jeffrey C Rathmell
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA.
| |
Collapse
|
91
|
Tomaszewicz M, Ronowska A, Zieliński M, Jankowska-Kulawy A, Trzonkowski P. T regulatory cells metabolism: The influence on functional properties and treatment potential. Front Immunol 2023; 14:1122063. [PMID: 37033990 PMCID: PMC10081158 DOI: 10.3389/fimmu.2023.1122063] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
CD4+CD25highFoxP3+ regulatory T cells (Tregs) constitute a small but substantial fraction of lymphocytes in the immune system. Tregs control inflammation associated with infections but also when it is improperly directed against its tissues or cells. The ability of Tregs to suppress (inhibit) the immune system is possible due to direct interactions with other cells but also in a paracrine fashion via the secretion of suppressive compounds. Today, attempts are made to use Tregs to treat autoimmune diseases, allergies, and rejection after bone marrow or organ transplantation. There is strong evidence that the metabolic program of Tregs is connected with the phenotype and function of these cells. A modulation towards a particular metabolic stage of Tregs may improve or weaken cells’ stability and function. This may be an essential tool to drive the immune system keeping it activated during infections or suppressed when autoimmunity occurs.
Collapse
Affiliation(s)
- Martyna Tomaszewicz
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Gdanísk, Poland
- Poltreg S.A., Gdanísk, Poland
- *Correspondence: Martyna Tomaszewicz,
| | - Anna Ronowska
- Department of Laboratory Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdanísk, Poland
| | - Maciej Zieliński
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Gdanísk, Poland
- Poltreg S.A., Gdanísk, Poland
| | | | - Piotr Trzonkowski
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Gdanísk, Poland
- Poltreg S.A., Gdanísk, Poland
| |
Collapse
|
92
|
Rao D, Stunnenberg JA, Lacroix R, Dimitriadis P, Kaplon J, Verburg F, van van Royen PT, Hoefsmit EP, Renner K, Blank CU, Peeper DS. Acidity-mediated induction of FoxP3 + regulatory T cells. Eur J Immunol 2023:e2250258. [PMID: 36788428 DOI: 10.1002/eji.202250258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/12/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
Glucose limitation and increased lactic acid levels are consequences of the elevated glycolytic activity of tumor cells, and constitute a metabolic barrier for the function of tumor infiltrating effector immune cells. The immune-suppressive functions of regulatory T cells (Tregs) are unobstructed in lactic-acid rich environments. However, the impact of lactic acid on the induction of Tregs remains unknown. We observed increased TGFβ-mediated induction of Forkhead box P3+ (FoxP3+ ) cells in the presence of extracellular lactic acid, in a glycolysis-independent, acidity-dependent manner. These CD4+ FoxP3+ cells expressed Treg-associated markers, including increased expression of CD39, and were capable of exerting suppressive functions. Corroborating these results in vivo, we observed that neutralizing the tumor pH by systemic administration of sodium bicarbonate (NaBi) decreased Treg abundance. We conclude that acidity augments Treg induction and propose that therapeutic targeting of acidity in the tumor microenvironment (TME) might reduce Treg-mediated immune suppression within tumors.
Collapse
Affiliation(s)
- Disha Rao
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Johanna A Stunnenberg
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ruben Lacroix
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Petros Dimitriadis
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joanna Kaplon
- Department of Clinical Chemistry, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Fabienne Verburg
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Paula T van van Royen
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esmée P Hoefsmit
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kathrin Renner
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, Regensburg, Germany.,Department of Otorhinolaryngology, University Hospital Regensburg, Regensburg, Germany
| | - Christian U Blank
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Daniel S Peeper
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, The Netherlands
| |
Collapse
|
93
|
Menezes dos Reis L, Berçot MR, Castelucci BG, Martins AJE, Castro G, Moraes-Vieira PM. Immunometabolic Signature during Respiratory Viral Infection: A Potential Target for Host-Directed Therapies. Viruses 2023; 15:v15020525. [PMID: 36851739 PMCID: PMC9965666 DOI: 10.3390/v15020525] [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: 01/31/2023] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
RNA viruses are known to induce a wide variety of respiratory tract illnesses, from simple colds to the latest coronavirus pandemic, causing effects on public health and the economy worldwide. Influenza virus (IV), parainfluenza virus (PIV), metapneumovirus (MPV), respiratory syncytial virus (RSV), rhinovirus (RhV), and coronavirus (CoV) are some of the most notable RNA viruses. Despite efforts, due to the high mutation rate, there are still no effective and scalable treatments that accompany the rapid emergence of new diseases associated with respiratory RNA viruses. Host-directed therapies have been applied to combat RNA virus infections by interfering with host cell factors that enhance the ability of immune cells to respond against those pathogens. The reprogramming of immune cell metabolism has recently emerged as a central mechanism in orchestrated immunity against respiratory viruses. Therefore, understanding the metabolic signature of immune cells during virus infection may be a promising tool for developing host-directed therapies. In this review, we revisit recent findings on the immunometabolic modulation in response to infection and discuss how these metabolic pathways may be used as targets for new therapies to combat illnesses caused by respiratory RNA viruses.
Collapse
Affiliation(s)
- Larissa Menezes dos Reis
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas 13083-862, SP, Brazil
| | - Marcelo Rodrigues Berçot
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas 13083-862, SP, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-270, SP, Brazil
| | - Bianca Gazieri Castelucci
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas 13083-862, SP, Brazil
| | - Ana Julia Estumano Martins
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas 13083-862, SP, Brazil
- Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas 13083-970, SP, Brazil
| | - Gisele Castro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas 13083-862, SP, Brazil
| | - Pedro M. Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas 13083-862, SP, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas 13083-872, SP, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas 13083-872, SP, Brazil
- Correspondence:
| |
Collapse
|
94
|
Salmond RJ. Regulation of T Cell Activation and Metabolism by Transforming Growth Factor-Beta. BIOLOGY 2023; 12:297. [PMID: 36829573 PMCID: PMC9953227 DOI: 10.3390/biology12020297] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/15/2023]
Abstract
Transforming growth factor beta (TGFβ) receptor signalling regulates T cell development, differentiation and effector function. Expression of the immune-associated isoform of this cytokine, TGFβ1, is absolutely required for the maintenance of immunological tolerance in both mice and humans, whilst context-dependent TGFβ1 signalling regulates the differentiation of both anti- and pro-inflammatory T cell effector populations. Thus, distinct TGFβ-dependent T cell responses are implicated in the suppression or initiation of inflammatory and autoimmune diseases. In cancer settings, TGFβ signals contribute to the blockade of anti-tumour immune responses and disease progression. Given the key functions of TGFβ in the regulation of immune responses and the potential for therapeutic targeting of TGFβ-dependent pathways, the mechanisms underpinning these pleiotropic effects have been the subject of much investigation. This review focuses on accumulating evidence suggesting that modulation of T cell metabolism represents a major mechanism by which TGFβ influences T cell immunity.
Collapse
Affiliation(s)
- Robert J Salmond
- Leeds Institute of Medical Research at St. James's, School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
95
|
Kim D, Kim G, Yu R, Lee J, Kim S, Qiu K, Montauti E, Fang D, Chandel NS, Choi J, Min B. Lymphocyte activation gene 3 (Lag3) supports Foxp3 + Treg cell function by restraining c-Myc-dependent aerobic glycolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528371. [PMID: 36824824 PMCID: PMC9949104 DOI: 10.1101/2023.02.13.528371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Lymphocyte activation gene 3 (Lag3) has emerged as the next-generation immune checkpoint molecule due to its ability to inhibit effector T cell activity. Foxp3 + regulatory T (Treg) cells, a master regulator of immunity and tolerance, also highly express Lag3. While Lag3 is thought to be necessary for Treg cell-mediated regulation of immunity, the precise roles and underlying mechanisms remain largely elusive. In this study, we report that Lag3 is indispensable for Treg cells to control autoimmune inflammation. Utilizing a newly generated Treg cell specific Lag3 mutant mouse model, we found that these animals are highly susceptible to autoimmune diseases, suggesting defective Treg cell function. Genome wide transcriptome analysis further uncovered that Lag3 mutant Treg cells upregulated genes involved in metabolic processes. Mechanistically, we found that Lag3 limits Treg cell expression of Myc, a key regulator of aerobic glycolysis. We further found that Lag3-dependent Myc expression determines Treg cells’ metabolic programming as well as the in vivo function to suppress autoimmune inflammation. Taken together, our results uncovered a novel function of Lag3 in supporting Treg cell suppressive function by regulating Myc-dependent metabolic programming.
Collapse
|
96
|
Wang J, Gong R, Zhao C, Lei K, Sun X, Ren H. Human FOXP3 and tumour microenvironment. Immunology 2023; 168:248-255. [PMID: 35689826 DOI: 10.1111/imm.13520] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/12/2022] [Indexed: 01/17/2023] Open
Abstract
The tumour microenvironment (TME) is a complex system composed of cancer cells, stromal cells and immune cells. Regulatory T cells (Tregs) in the TME impede immune surveillance of tumours and suppress antitumor immune responses. Transcription factor forkhead box protein 3 (FOXP3) is the main marker of Tregs, which dominates the function of Tregs. FOXP3 was originally thought to be a Tregs-specific expression molecule, and recent studies have found that FOXP3 is expressed in a variety of tumours with inconsistent functional roles. This review summarizes the recent progress of infiltrating Treg-FOXP3 and tumour-FOXP3 in TME, discusses the communication mechanism between FOXP3+ cells and effector T cells in TME, the relationship between FOXP3 and clinical prognosis, and the potential of FOXP3-targeted therapy.
Collapse
Affiliation(s)
- Jia Wang
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Qingdao Medical School, Qingdao University, Qingdao, Shandong, China
| | - Ruining Gong
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chenyang Zhao
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ke Lei
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoyuan Sun
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - He Ren
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Tianjin, China
| |
Collapse
|
97
|
Ma R, Su H, Jiao K, Liu J. Role of Th17 cells, Treg cells, and Th17/Treg imbalance in immune homeostasis disorders in patients with chronic obstructive pulmonary disease. Immun Inflamm Dis 2023; 11:e784. [PMID: 36840492 PMCID: PMC9950879 DOI: 10.1002/iid3.784] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/21/2023] [Accepted: 01/30/2023] [Indexed: 02/26/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide, following strokes and cardiovascular diseases. Chronic lung inflammation is believed to play a role in the development of COPD. In addition, accumulating evidence shows that the immune system plays a crucial role in the pathogenesis of COPD. Significant advancements have been made in research on the pathogenesis of immune diseases and chronic inflammation in recent years, and T helper 17 (Th17) cells and regulatory T (Treg) cells have been found to play a crucial role in the autoimmune response. Th17 cells are a proinflammatory subpopulation that causes autoimmune disease and tissue damage. Treg cells, on the other hand, have a negative effect but can contribute to the occurrence of the same disease when their antagonism fails. This review mainly summarizes the biological characteristics of Th17 cells and Treg cells, their roles in chronic inflammatory diseases of COPD, and the role of the Th17/Treg ratio in the onset, development, and outcome of inflammatory disorders, as well as recent advancements in immunomodulatory treatment targeting Th17/Treg cells in COPD.
Collapse
Affiliation(s)
- Ru Ma
- Department of The First Clinical School of MedicineLanzhou UniversityLanzhouChina
- Department of Gansu Provincial People's HospitalLanzhouChina
| | - Hongling Su
- Department of The First Clinical School of MedicineLanzhou UniversityLanzhouChina
- Department of Gansu Provincial People's HospitalLanzhouChina
| | - Keping Jiao
- Department of The First Clinical School of MedicineLanzhou UniversityLanzhouChina
- Department of Gansu Provincial People's HospitalLanzhouChina
| | - Jian Liu
- Department of The First Clinical School of MedicineLanzhou UniversityLanzhouChina
| |
Collapse
|
98
|
Dai M, Wang L, Yang J, Chen J, Dou X, Chen R, Ge Y, Lin Y. LDHA as a regulator of T cell fate and its mechanisms in disease. Biomed Pharmacother 2023; 158:114164. [PMID: 36916398 DOI: 10.1016/j.biopha.2022.114164] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/13/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023] Open
Abstract
T cells are the main force of anti-infection and antitumor and are also involved in autoimmune diseases. During the development of these diseases, T cells need to rapidly produce large amounts of energy to satisfy their activation, proliferation, and differentiation. In this review, we introduced lactate dehydrogenase A(LDHA), predominantly involved in glycolysis, which provides energy for T cells and plays a dual role in disease by mediating lactate production, non-classical enzyme activity, and oxidative stress. Mechanistically, the signaling molecule can interact with the LDHA promoter or regulate LDHA activity through post-translational modifications. These latest findings suggest that modulation of LDHA may have considerable therapeutic effects in diseases where T-cell activation is an important pathogenesis.
Collapse
Affiliation(s)
- Maosha Dai
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Li Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Juexi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Jiayi Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Xiaoke Dou
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Rui Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Yangyang Ge
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China.
| | - Yun Lin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China.
| |
Collapse
|
99
|
Peng M, Li MO. Metabolism along the life journey of T cells. LIFE METABOLISM 2023; 2:load002. [PMID: 37305096 PMCID: PMC10256237 DOI: 10.1093/lifemeta/load002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/26/2022] [Accepted: 01/16/2023] [Indexed: 06/13/2023]
Abstract
T cells are one of few cell types in adult mammals that can proliferate extensively and differentiate diversely upon stimulation, which serves as an excellent example to dissect the metabolic basis of cell fate decisions. During the last decade, there has been an explosion of research into the metabolic control of T-cell responses. The roles of common metabolic pathways, including glycolysis, lipid metabolism, and mitochondrial oxidative phosphorylation, in T-cell responses have been well characterized, and their mechanisms of action are starting to emerge. In this review, we present several considerations for T-cell metabolism-focused research, while providing an overview of the metabolic control of T-cell fate decisions during their life journey. We try to synthesize principles that explain the causal relationship between cellular metabolism and T-cell fate decision. We also discuss key unresolved questions and challenges in targeting T-cell metabolism to treat disease.
Collapse
Affiliation(s)
- Min Peng
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
- Institute for Immunology, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing 100084, China
| | - Ming O Li
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| |
Collapse
|
100
|
Chelakkot C, Chelakkot VS, Shin Y, Song K. Modulating Glycolysis to Improve Cancer Therapy. Int J Mol Sci 2023; 24:2606. [PMID: 36768924 PMCID: PMC9916680 DOI: 10.3390/ijms24032606] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
Cancer cells undergo metabolic reprogramming and switch to a 'glycolysis-dominant' metabolic profile to promote their survival and meet their requirements for energy and macromolecules. This phenomenon, also known as the 'Warburg effect,' provides a survival advantage to the cancer cells and make the tumor environment more pro-cancerous. Additionally, the increased glycolytic dependence also promotes chemo/radio resistance. A similar switch to a glycolytic metabolic profile is also shown by the immune cells in the tumor microenvironment, inducing a competition between the cancer cells and the tumor-infiltrating cells over nutrients. Several recent studies have shown that targeting the enhanced glycolysis in cancer cells is a promising strategy to make them more susceptible to treatment with other conventional treatment modalities, including chemotherapy, radiotherapy, hormonal therapy, immunotherapy, and photodynamic therapy. Although several targeting strategies have been developed and several of them are in different stages of pre-clinical and clinical evaluation, there is still a lack of effective strategies to specifically target cancer cell glycolysis to improve treatment efficacy. Herein, we have reviewed our current understanding of the role of metabolic reprogramming in cancer cells and how targeting this phenomenon could be a potential strategy to improve the efficacy of conventional cancer therapy.
Collapse
Affiliation(s)
| | - Vipin Shankar Chelakkot
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Youngkee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Science, Department of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyoung Song
- College of Pharmacy, Duksung Women’s University, Seoul 01366, Republic of Korea
| |
Collapse
|