1
|
Drexhage HA, Bergink V, Poletti S, Benedetti F, Osborne LM. Conventional and new immunotherapies for immune system dysregulation in postpartum mood disorders: comparisons to immune system dysregulations in bipolar disorder, major depression, and postpartum autoimmune thyroid disease. Expert Rev Clin Immunol 2025; 21:113-135. [PMID: 39441185 PMCID: PMC11786996 DOI: 10.1080/1744666x.2024.2420053] [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: 06/23/2024] [Revised: 09/17/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Postpartum mood disorders are heterogenous disorders and comprise postpartum psychosis and postpartum depression. Evidence is accumulating that systemic monocyte/macrophage activation, low-grade inflammation and (premature senescence related) T cell defects increase the risk for mood disorders outside pregnancy by affecting the function of microglia and T cells in the emotional brain (the cortico-limbic system) leading to inadequate mood regulation upon stress. AREAS COVERED The evidence in the literature that similar immune dysregulations are present in postpartum mood disorders. RESULTS The physiological postpartum period is characterized by a rapid T cell surge and a mild activation of the monocyte/macrophage system. Postpartum mood disorder patients show a diminished T cell surge (including that of T regulatory cells) and an increase in low grade inflammation, that is, an increased inflammatory state of monocytes/macrophages and higher levels of serum pro-inflammatory cytokines. EXPERT OPINION Anti-inflammatory agents (e.g. COX-2 inhibitors) and T cell boosting agents (e.g. low-dose IL-2 therapy) should be further investigated as treatment. The hypothesis should be investigated that postpartum mood disorders are active episodes (triggered by changes in the postpartum immuno-endocrine milieu) in ongoing, dynamically fluctuating aberrant neuro-immune-endocrine trajectories leading to mood disorders in women (inheritably) vulnerable to these disorders.
Collapse
Affiliation(s)
- Hemmo A Drexhage
- Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Veerle Bergink
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sara Poletti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| | - Lauren M Osborne
- Departments of Obstetrics and Gynecology and of Psychiatry, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
2
|
Leboyer M, Foiselle M, Tchitchek N, Tamouza R, Lorenzon R, Richard JR, Arrouasse R, Le Corvoisier P, Le Dudal K, Vicaut E, Ellul P, Rosenzwajg M, Klatzmann D. Low-dose interleukin-2 in patients with bipolar depression: A phase 2 randomised double-blind placebo-controlled trial. Brain Behav Immun 2025; 123:177-184. [PMID: 39242054 DOI: 10.1016/j.bbi.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Immune abnormalities including an insufficiency of regulatory T cells (Treg) and increased blood-based inflammatory markers have been observed in bipolar disorders (BD), particularly during depression. As Tregs are pivotal to control inflammation, Treg stimulation by low-dose IL-2 (IL-2LD) could have a therapeutic impact on bipolar depression. We performed a randomized, double-blind, placebo-controlled (2 active: 1 placebo) proof-of-concept trial of add-on IL-2LD in patients with bipolar depression. Patients received a placebo or IL-2LD (1MIU) once a day for 5 days, and then once a week for 4 weeks starting on week 2. The primary objective was to demonstrate a biological Treg response to IL-2LD assessed by fold increase in Treg percentage of CD4 + cells from baseline to day 5. Secondary objectives included safety assessment and mood improvement throughout the study period. This trial is registered with ClinicalTrials.gov, number NCT04133233. Fourteen patients with bipolar depression were included, with 4 receiving placebo and 10 IL-2LD. Baseline clinical and biological characteristics were balanced between groups. The primary evaluation criterion was met, with IL-2LD expanding 1.17 [95 % CI 1.01-1.34] vs 1.01 [95 % CI 0.90-1.12] (p = 0.0421) and activating Tregs. Secondary evaluation criteria were also met with significant improvements of depressive symptoms and global functioning from day-15 onwards in the IL-2LD treated patients. The treatment was well-tolerated, with no serious adverse events related to treatment. This proof-of-concept trial shows that stimulating Tregs in patients with bipolar depression is safe and associated with clinical improvements. This supports a pathophysiological role of inflammation in BD and warrants pursuing the evaluation of IL-2LD as an adjunct treatment of major mood disorders.
Collapse
Affiliation(s)
- Marion Leboyer
- Université Paris Est Créteil (UPEC), AP-HP, Department of Psychiatry, Hôpital Henri Mondor, DMU IMPACT, FHU ADAPT, INSERM U955, IMRB, Translational NeuroPsychiatry Laboratory, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France.
| | - Marianne Foiselle
- Université Paris Est Créteil (UPEC), AP-HP, Department of Psychiatry, Hôpital Henri Mondor, DMU IMPACT, FHU ADAPT, INSERM U955, IMRB, Translational NeuroPsychiatry Laboratory, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Nicolas Tchitchek
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3), F-75651 Paris, France
| | - Ryad Tamouza
- Université Paris Est Créteil (UPEC), AP-HP, Department of Psychiatry, Hôpital Henri Mondor, DMU IMPACT, FHU ADAPT, INSERM U955, IMRB, Translational NeuroPsychiatry Laboratory, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Roberta Lorenzon
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3), F-75651 Paris, France; AP-HP, Pitié-Salpêtrière Hospital, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), F-75651 Paris, France
| | - Jean-Romain Richard
- Université Paris Est Créteil (UPEC), AP-HP, Department of Psychiatry, Hôpital Henri Mondor, DMU IMPACT, FHU ADAPT, INSERM U955, IMRB, Translational NeuroPsychiatry Laboratory, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Raphaele Arrouasse
- Inserm, Centre d'Investigation Clinique 1430 et AP-HP, Hôpitaux Universitaires Henri Mondor, Univ Paris Est Creteil, F-94010 Créteil, France
| | - Philippe Le Corvoisier
- Inserm, Centre d'Investigation Clinique 1430 et AP-HP, Hôpitaux Universitaires Henri Mondor, Univ Paris Est Creteil, F-94010 Créteil, France
| | - Katia Le Dudal
- Inserm, Centre d'Investigation Clinique 1430 et AP-HP, Hôpitaux Universitaires Henri Mondor, Univ Paris Est Creteil, F-94010 Créteil, France
| | - Eric Vicaut
- AP-HP, Saint Louis/Lariboisière Hospitals, Unité de recherche clinique and Université Paris 7, F-75010 Paris, France
| | - Pierre Ellul
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3), F-75651 Paris, France
| | - Michelle Rosenzwajg
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3), F-75651 Paris, France; AP-HP, Pitié-Salpêtrière Hospital, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), F-75651 Paris, France
| | - David Klatzmann
- Sorbonne Université, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3), F-75651 Paris, France; AP-HP, Pitié-Salpêtrière Hospital, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), F-75651 Paris, France.
| |
Collapse
|
3
|
Wang M, Guo H, Sun BB, Jie XL, Shi XY, Liu YQ, Shi XL, Ding LQ, Xue PH, Qiu F, Cao W, Wang GZ, Zhou GB. Centipeda minima and 6-O-angeloylplenolin enhance the efficacy of immune checkpoint inhibitors in non-small cell lung cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155825. [PMID: 38968790 DOI: 10.1016/j.phymed.2024.155825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Chemotherapeutic agents including cisplatin, gemcitabine, and pemetrexed, significantly enhance the efficacy of immune checkpoint inhibitors (ICIs) in non-small cell lung cancer (NSCLC) by increasing PD-L1 expression and potentiating T cell cytotoxicity. However, the low response rate and adverse effects limit the application of chemotherapy/ICI combinations in patients. METHODS We screened for medicinal herbs that could perturb PD-L1 expression and enhance T cell cytotoxicity in the presence of anti-PD-L1 antibody, and investigated the underlying mechanisms. RESULTS We found that the aqueous extracts of Centipeda minima (CM) significantly enhanced the cancer cell-killing activity and granzyme B expression level of CD8+ T cells, in the presence of anti-PD-L1 antibody. Both CM and its active component 6-O-angeloylplenolin (6-OAP) upregulated PD-L1 expression by suppressing GSK-3β-β-TRCP-mediated ubiquitination and degradation. CM and 6-OAP significantly enhanced ICI-induced reduction of tumor burden and prolongation of overall survival of mice bearing NSCLC cells, accompanied by upregulation of PD-L1 and increase of CD8+ T cell infiltration. CM also exhibited anti-NSCLC activity in cells and in a patient-derived xenograft mouse model. CONCLUSIONS These data demonstrated that the induced expression of PD-L1 and enhancement of CD8+ T cell cytotoxicity underlay the beneficial effects of 6-OAP-rich CM in NSCLCs, providing a clinically available and safe medicinal herb for combined use with ICIs to treat this deadly disease.
Collapse
Affiliation(s)
- Min Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou 450008, China
| | - Hua Guo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bei-Bei Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiao-Liang Jie
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xue-Yan Shi
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yong-Qiang Liu
- Research Center of Chinese Herbal Resources Science and Engineering, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xu-Liu Shi
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Li-Qin Ding
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Peng-Hui Xue
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wei Cao
- Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Gui-Zhen Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Guang-Biao Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
4
|
Keskin-Erdogan Z, Mandakhbayar N, Jin GS, Li YM, Chau DYS, Day RM, Kim HW, Knowles JC. Lithium-loaded GelMA-Phosphate glass fibre constructs: Implications for astrocyte response. J Biomed Mater Res A 2024; 112:1070-1082. [PMID: 38400701 DOI: 10.1002/jbm.a.37686] [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: 08/02/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024]
Abstract
Combinations of different biomaterials with their own advantages as well as functionalization with other components have long been implemented in tissue engineering to improve the performance of the overall material. Biomaterials, particularly hydrogel platforms, have shown great potential for delivering compounds such as drugs, growth factors, and neurotrophic factors, as well as cells, in neural tissue engineering applications. In central the nervous system, astrocyte reactivity and glial scar formation are significant and complex challenges to tackle for neural and functional recovery. GelMA hydrogel-based tissue constructs have been developed in this study and combined with two different formulations of phosphate glass fibers (PGFs) (with Fe3+ or Ti2+ oxide) to impose physical and mechanical cues for modulating astrocyte cell behavior. This study was also aimed at investigating the effects of lithium-loaded GelMA-PGFs hydrogels in alleviating astrocyte reactivity and glial scar formation offering novel perspectives for neural tissue engineering applications. The rationale behind introducing lithium is driven by its long-proven therapeutic benefits in mental disorders, and neuroprotective and pronounced anti-inflammatory properties. The optimal concentrations of lithium and LPS were determined in vitro on primary rat astrocytes. Furthermore, qPCR was conducted for gene expression analysis of GFAP and IL-6 markers on primary astrocytes cultured 3D into GelMA and GelMA-PGFs hydrogels with and without lithium and in vitro stimulated with LPS for astrocyte reactivity. The results suggest that the combination of bioactive phosphate-based glass fibers and lithium loading into GelMA structures may impact GFAP expression and early IL-6 expression. Furthermore, GelMA-PGFs (Fe) constructs have shown improved performance in modulating glial scarring over GFAP regulation.
Collapse
Affiliation(s)
- Zalike Keskin-Erdogan
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, Royal Free Hospital, London, UK
- Chemical Engineering Department, Imperial College London, London, UK
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
| | - Gang Shi Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
| | - Yu-Meng Li
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
| | - David Y S Chau
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, Royal Free Hospital, London, UK
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
| | - Richard M Day
- Centre for Precision Healthcare, UCL Division of Medicine, University College London, London, UK
| | - Hae-Won Kim
- Chemical Engineering Department, Imperial College London, London, UK
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Jonathan C Knowles
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, Royal Free Hospital, London, UK
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
5
|
Poletti S, Zanardi R, Mandelli A, Aggio V, Finardi A, Lorenzi C, Borsellino G, Carminati M, Manfredi E, Tomasi E, Spadini S, Colombo C, Drexhage HA, Furlan R, Benedetti F. Low-dose interleukin 2 antidepressant potentiation in unipolar and bipolar depression: Safety, efficacy, and immunological biomarkers. Brain Behav Immun 2024; 118:52-68. [PMID: 38367846 DOI: 10.1016/j.bbi.2024.02.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024] Open
Abstract
Immune-inflammatory mechanisms are promising targets for antidepressant pharmacology. Immune cell abnormalities have been reported in mood disorders showing a partial T cell defect. Following this line of reasoning we defined an antidepressant potentiation treatment with add-on low-dose interleukin 2 (IL-2). IL-2 is a T-cell growth factor which has proven anti-inflammatory efficacy in autoimmune conditions, increasing thymic production of naïve CD4 + T cells, and possibly correcting the partial T cell defect observed in mood disorders. We performed a single-center, randomised, double-blind, placebo-controlled phase II trial evaluating the safety, clinical efficacy and biological responses of low-dose IL-2 in depressed patients with major depressive (MDD) or bipolar disorder (BD). 36 consecutively recruited inpatients at the Mood Disorder Unit were randomised in a 2:1 ratio to receive either aldesleukin (12 MDD and 12 BD) or placebo (6 MDD and 6 BD). Active treatment significantly potentiated antidepressant response to ongoing SSRI/SNRI treatment in both diagnostic groups, and expanded the population of T regulatory, T helper 2, and percentage of Naive CD4+/CD8 + immune cells. Changes in cell frequences were rapidly induced in the first five days of treatment, and predicted the later improvement of depression severity. No serious adverse effect was observed. This is the first randomised control trial (RCT) evidence supporting the hypothesis that treatment to strengthen the T cell system could be a successful way to correct the immuno-inflammatory abnormalities associated with mood disorders, and potentiate antidepressant response.
Collapse
Affiliation(s)
- Sara Poletti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy.
| | - Raffaella Zanardi
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Alessandra Mandelli
- Clinical Neuroimmunology, Institute of Experimental Neurology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Veronica Aggio
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology, Institute of Experimental Neurology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | | | - Matteo Carminati
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Elena Manfredi
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Enrico Tomasi
- Hospital Pharmacy, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Sara Spadini
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Cristina Colombo
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Hemmo A Drexhage
- Coordinator EU consortium MoodStratification, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Roberto Furlan
- Vita-Salute San Raffaele University, Milano, Italy; Clinical Neuroimmunology, Institute of Experimental Neurology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| |
Collapse
|
6
|
Zhang B, Feng H, Lin H, Li R. Somatostatin-SSTR3-GSK3 modulates human T-cell responses by inhibiting OXPHOS. Front Immunol 2024; 15:1322670. [PMID: 38426092 PMCID: PMC10902055 DOI: 10.3389/fimmu.2024.1322670] [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: 10/16/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Somatostatin (SST) is a peptide hormone primarily synthesized in the digestive and nervous systems. While its impact on the endocrine system is well-established, accumulating evidence suggests a crucial role for SST and its analogues in modulating immune responses. Despite this, the precise mechanism through which SST regulates T cells has remained largely unknown. Methods To elucidate the impact of SST on human T cells, we conducted a series of experiments involving cell culture assays, molecular analyses, and metabolic profiling. Human T cells were treated with SST, and various parameters including proliferation, cytokine production, and metabolic activities were assessed. Additionally, we employed pharmacological inhibitors and genetic manipulations to dissect the signaling pathways mediating SST's effects on T cells. Results We showed that SST diminishes T-cell proliferation by influencing IL-2 production and T-cell mitochondrial respiration, while having no discernible impact on TCR-induced glycolysis. Our findings also identified that the regulatory influence of SST on T-cell responses and metabolism is contingent on its receptor, SSTR3. Moreover, we demonstrated that SST governs T-cell responses and metabolism by acting through the T-cell metabolic checkpoint GSK3. Discussion Our study provides novel insights into the immunoregulatory function of SST in human T cells, highlighting the complex interplay between hormonal signaling and immune regulation. Understanding the molecular mechanisms underlying SST's effects on T cells may offer therapeutic opportunities for manipulating immune responses in various pathological conditions.
Collapse
Affiliation(s)
- Bo Zhang
- Institute of Neuroscience and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Research, Fujian Medical University, Fuzhou, China
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Huiru Feng
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hui Lin
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Rui Li
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
7
|
Montecchi T, Nannini G, De Tommaso D, Cassioli C, Coppola F, Ringressi MN, Carraro F, Naldini A, Taddei A, Marotta G, Amedei A, Baldari CT, Ulivieri C. Human colorectal cancer: upregulation of the adaptor protein Rai in TILs leads to cell dysfunction by sustaining GSK-3 activation and PD-1 expression. Cancer Immunol Immunother 2024; 73:2. [PMID: 38175205 PMCID: PMC10766791 DOI: 10.1007/s00262-023-03614-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND The immunosuppressive tumor microenvironment (TME) of colorectal cancer (CRC) is a major hurdle for immune checkpoint inhibitor-based therapies. Hence characterization of the signaling pathways driving T cell exhaustion within TME is a critical need for the discovery of novel therapeutic targets and the development of effective therapies. We previously showed that (i) the adaptor protein Rai is a negative regulator of T cell receptor signaling and T helper 1 (Th1)/Th17 cell differentiation; and (ii) Rai deficiency is implicated in the hyperactive phenotype of T cells in autoimmune diseases. METHODS The expression level of Rai was measured by qRT-PCR in paired peripheral blood T cells and T cells infiltrating tumor tissue and the normal adjacent tissue in CRC patients. The impact of hypoxia-inducible factor (HIF)-1α on Rai expression was evaluated in T cells exposed to hypoxia and by performing chromatin immunoprecipitation assays and RNA interference assays. The mechanism by which upregulation of Rai in T cells promotes T cell exhaustion were evaluated by flow cytometric, qRT-PCR and western blot analyses. RESULTS We show that Rai is a novel HIF-1α-responsive gene that is upregulated in tumor infiltrating lymphocytes of CRC patients compared to patient-matched circulating T cells. Rai upregulation in T cells promoted Programmed cell Death protein (PD)-1 expression and impaired antigen-dependent degranulation of CD8+ T cells by inhibiting phospho-inactivation of glycogen synthase kinase (GSK)-3, a central regulator of PD-1 expression and T cell-mediated anti-tumor immunity. CONCLUSIONS Our data identify Rai as a hitherto unknown regulator of the TME-induced exhausted phenotype of human T cells.
Collapse
Affiliation(s)
- Tommaso Montecchi
- Department of Life Sciences, University of Siena, Siena, 53100, Italy
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | | | - Chiara Cassioli
- Department of Life Sciences, University of Siena, Siena, 53100, Italy
| | - Federica Coppola
- Department of Molecular and Developmental Medicine, University of Siena, Siena, 53100, Italy
| | - Maria Novella Ringressi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | - Fabio Carraro
- Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Antonella Naldini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, 53100, Italy
| | - Antonio Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy.
| | - Cosima T Baldari
- Department of Life Sciences, University of Siena, Siena, 53100, Italy.
| | - Cristina Ulivieri
- Department of Life Sciences, University of Siena, Siena, 53100, Italy.
| |
Collapse
|
8
|
Crespo AR, Luna SG, Moës B, Rodriguez A, Rudd CE. The many faceted role of glycogen synthase kinase-3 (GSK-3) in T cells and cancer immunotherapy. JOURNAL OF CANCER BIOLOGY 2024; 5:11-16. [PMID: 38813058 PMCID: PMC11134095 DOI: 10.46439/cancerbiology.5.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Affiliation(s)
- Aurora Rivas Crespo
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain
- Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain
- Division of Immunology-Oncology, Centre de Recherche Hôpital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
| | - Silvia Guil Luna
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain
- Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain
| | - Bastien Moës
- Division of Immunology-Oncology, Centre de Recherche Hôpital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Faculty of Medicine, Université de Montreal, Montreal, Canada
| | - Antonio Rodriguez
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
- Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain
- Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain
| | - Christopher E Rudd
- Division of Immunology-Oncology, Centre de Recherche Hôpital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Faculty of Medicine, Université de Montreal, Montreal, Canada
- Department of Microbiology, Infection and Immunology, Faculty of Medicine, Université de Montreal, Montreal, QC, Canada
| |
Collapse
|
9
|
Guil-Luna S, Rivas-Crespo A, Navarrete-Sirvent C, Mantrana A, Pera A, Mena-Osuna R, Toledano-Fonseca M, García-Ortíz MV, Villar C, Sánchez-Montero MT, Krueger J, Medina-Fernández FJ, De La Haba-Rodríguez J, Gómez-España A, Aranda E, Rudd CE, Rodríguez-Ariza A. Clinical significance of glycogen synthase kinase 3 (GSK-3) expression and tumor budding grade in colorectal cancer: Implications for targeted therapy. Biomed Pharmacother 2023; 167:115592. [PMID: 37778272 DOI: 10.1016/j.biopha.2023.115592] [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: 08/04/2023] [Revised: 09/14/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023] Open
Abstract
INTRODUCTION Glycogen synthase kinase 3 (GSK-3) has been proposed as a novel cancer target due to its regulating role in both tumor and immune cells. However, the connection between GSK-3 and immunoevasive contexture, including tumor budding (TB) has not been previously examined. METHODS we investigated the expression levels of total GSK-3 as well as its isoforms (GSK-3β and GSK-3α) and examined their potential correlation with TB grade and the programmed cell death-ligand 1 (PD-L1) in colorectal cancer (CRC) tumor samples. Additionally, we compared the efficacy of GSK-3-inhibition with PD-1/PD-L1 blockade in humanized patient-derived (PDXs) xenografts models of high-grade TB CRC. RESULTS we show that high-grade (BD3) TB CRC is associated with elevated expression levels of total GSK-3, specifically the GSK-3β isoform, along with increased expression of PD-L1 in tumor cells. Moreover, we define an improved risk stratification of CRC patients based on the presence of GSK-3+/PD-L1+/BD3 tumors, which are associated with a worse prognosis. Significantly, in contrast to the PD-L1/PD-1 blockade approach, the inhibition GSK-3 demonstrated a remarkable enhancement in the antitumor response. This was achieved through the reduction of tumor buds via necrosis and apoptosis pathways, along with a notable increase of activated tumor-infiltrating CD8+ T cells, NK cells, and CD4- CD8- T cells. CONCLUSIONS our study provides compelling evidence for the clinical significance of GSK-3 expression and TB grade in risk stratification of CRC patients. Moreover, our findings strongly support GSK-3 inhibition as an effective therapy specifically targeting high-grade TB in CRC.
Collapse
Affiliation(s)
- Silvia Guil-Luna
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain; Department of Comparative Pathology, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain..
| | - Aurora Rivas-Crespo
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain.
| | - Carmen Navarrete-Sirvent
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain.
| | - Ana Mantrana
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain.
| | - Alejandra Pera
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain.
| | - Rafael Mena-Osuna
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain.
| | - Marta Toledano-Fonseca
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain.
| | - María Victoria García-Ortíz
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain.
| | - Carlos Villar
- Pathological Anatomy Department, Reina Sofía University Hospital, Córdoba, Spain.
| | - Maria Teresa Sánchez-Montero
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain.
| | - Janna Krueger
- Division of Immunology-Oncology Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada.
| | | | - Juan De La Haba-Rodríguez
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain; Department of Medicine, Faculty of Medicine, University of Córdoba, Córdoba, Spain; Medical Oncology Department, Reina Sofía University Hospital, Córdoba, Spain.
| | - Auxiliadora Gómez-España
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain; Department of Medicine, Faculty of Medicine, University of Córdoba, Córdoba, Spain; Medical Oncology Department, Reina Sofía University Hospital, Córdoba, Spain.
| | - Enrique Aranda
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain; Department of Medicine, Faculty of Medicine, University of Córdoba, Córdoba, Spain; Medical Oncology Department, Reina Sofía University Hospital, Córdoba, Spain.
| | - Christopher E Rudd
- General and Digestive Surgery Department, Reina Sofía University Hospital, Córdoba, Spain; Faculty of Medicine, Universite de Montreal, Montreal, Canada.
| | - Antonio Rodríguez-Ariza
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Centre (CIBERONC), Madrid, Spain; Andalusia-ROCHE Network Mixed Alliance in Precision Medical Oncology, Spain; Medical Oncology Department, Reina Sofía University Hospital, Córdoba, Spain.
| |
Collapse
|
10
|
Almeida Rodrigues AK, Silva PG, Nogueira C, Ferreira SS, Cordeiro J, Carneiro B, Tavora F. Expression of tumoral GSK3-β, PD-L1, and CD8 cell density in urothelial carcinomas, association with tumor grade and overall survival. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2023; 12:87-97. [PMID: 38022872 PMCID: PMC10658161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/28/2023] [Indexed: 12/01/2023]
Abstract
Bladder cancer is the most common malignancy in the urinary tract, and is biologically and clinically quite heterogeneous. Around 90% of diagnoses are made in the 6th decade, being more prevalent in males. The programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) axis play a putative role in immune checkpoint and as a means through which cancer evades the immune system. Inhibition of the glicogênio synthase kinase (GSK) 3 leads to the downregulation of PD-1 via upregulation of the transcription factor Tbet. The use of biomarkers PD-L1 and GSK-3β and evaluation of the immune infiltrate have very promising correlations with urothelial carcinoma prognosis and treatment prediction. OBJECTIVE To investigate the protein expression of PD-L1 and GSK-3β and the CD8-positive immune infiltrates in bladder carcinomas. MATERIALS AND METHODS This was a cross-sectional study of 140 samples of urothelial carcinomas from 2015 to 2018. Automated digitally assisted scoring and conventional analyses of the markers of GSK-3β (27C10), CD8 (7103β) and PDL-1 (22c3), were reviewed by two pathologists independently and a histologic score was calculated. The density of CD8 was also measured. RESULTS The immunoexpression of GSK-3β (91%) was presented in most samples, PD-L1 in 62.9% and CD8 cells present in 46.3% of cases. When analyzed in conjunction, the levels of GSK-3β and PD-L1 (P = 0.033), and CD8 and PD-L1 (P<0.002) showed significant correlations. No significant associations were observed between GSK-3β and CD8. The positivity of GSK-3β and PD-L1 was predominant in high-grade tumors. CONCLUSION Despite the tumor microenvironment heterogeneity, the expression of CD8, GSK-3β and PDL1 could be valuable and GSK-3β could be a potential target in advanced bladder cancer, especially in the context of immunotherapy.
Collapse
Affiliation(s)
| | - Paulo Goberlanio Silva
- ICC (Ceara Cancer Institute), Laboratory of Molecular Biology and GeneticsFortaleza, CE, Brazil
| | - Cleto Nogueira
- Argos LaboratoryFortaleza, CE, Brazil
- Department of Pathology and Legal Medicine, Federal University of CearaFortaleza, CE, Brazil
| | - Samuel S Ferreira
- Argos LaboratoryFortaleza, CE, Brazil
- Department of Pathology and Legal Medicine, Federal University of CearaFortaleza, CE, Brazil
| | - Juliana Cordeiro
- Argos LaboratoryFortaleza, CE, Brazil
- Department of Pathology and Legal Medicine, Federal University of CearaFortaleza, CE, Brazil
| | | | - Fabio Tavora
- Argos LaboratoryFortaleza, CE, Brazil
- Department of Pathology and Legal Medicine, Federal University of CearaFortaleza, CE, Brazil
| |
Collapse
|
11
|
Fu Y, Wang J, Liu C, Liao K, Gao X, Tang R, Fan B, Hong Y, Xiao N, Xiao C, Liu WH. Glycogen synthase kinase 3 controls T-cell exhaustion by regulating NFAT activation. Cell Mol Immunol 2023; 20:1127-1139. [PMID: 37553428 PMCID: PMC10541428 DOI: 10.1038/s41423-023-01075-0] [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: 03/10/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023] Open
Abstract
Cellular immunity mediated by CD8+ T cells plays an indispensable role in bacterial and viral clearance and cancers. However, persistent antigen stimulation of CD8+ T cells leads to an exhausted or dysfunctional cellular state characterized by the loss of effector function and high expression of inhibitory receptors during chronic viral infection and in tumors. Numerous studies have shown that glycogen synthase kinase 3 (GSK3) controls the function and development of immune cells, but whether GSK3 affects CD8+ T cells is not clearly elucidated. Here, we demonstrate that mice with deletion of Gsk3α and Gsk3β in activated CD8+ T cells (DKO) exhibited decreased CTL differentiation and effector function during acute and chronic viral infection. In addition, DKO mice failed to control tumor growth due to the upregulated expression of inhibitory receptors and augmented T-cell exhaustion in tumor-infiltrating CD8+ T cells. Strikingly, anti-PD-1 immunotherapy substantially restored tumor rejection in DKO mice. Mechanistically, GSK3 regulates T-cell exhaustion by suppressing TCR-induced nuclear import of NFAT, thereby in turn dampening NFAT-mediated exhaustion-related gene expression, including TOX/TOX2 and PD-1. Thus, we uncovered the molecular mechanisms underlying GSK3 regulation of CTL differentiation and T-cell exhaustion in anti-tumor immune responses.
Collapse
Affiliation(s)
- Yubing Fu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, 361102, Fujian, China.
| | - Jinjia Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, 361102, Fujian, China
| | - Chenfeng Liu
- Department of Cell Biology, School of Life Science, Anhui Medical University, Hefei, 230031, Anhui, China
| | - Kunyu Liao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, 361102, Fujian, China
| | - Xianjun Gao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, 361102, Fujian, China
| | - Ronghan Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, 361102, Fujian, China
| | - Binbin Fan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yazhen Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, 361102, Fujian, China
| | - Nengming Xiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, 361102, Fujian, China
| | - Changchun Xiao
- Sanofi Institute for Biomedical Research, Suzhou, Jiangsu, 215123, China
| | - Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Science, Xiamen University, Xiamen, 361102, Fujian, China.
| |
Collapse
|
12
|
Ghanaatfar F, Ghanaatfar A, Isapour P, Farokhi N, Bozorgniahosseini S, Javadi M, Gholami M, Ulloa L, Coleman-Fuller N, Motaghinejad M. Is lithium neuroprotective? An updated mechanistic illustrated review. Fundam Clin Pharmacol 2023; 37:4-30. [PMID: 35996185 DOI: 10.1111/fcp.12826] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 06/17/2022] [Accepted: 08/19/2022] [Indexed: 01/25/2023]
Abstract
Neurodegeneration is a pathological process characterized by progressive neuronal impairment, dysfunction, and loss due to mitochondrial dysfunction, oxidative stress, inflammation, and apoptosis. Many studies have shown that lithium protects against neurodegeneration. Herein, we summarize recent clinical and laboratory studies on the neuroprotective effects of lithium against neurodegeneration and its potential to modulate mitochondrial dysfunction, oxidative stress, inflammation, and apoptosis. Recent findings indicate that lithium regulates critical intracellular pathways such as phosphatidylinositol-3 (PI3)/protein kinase B (Akt)/glycogen synthase kinase-3 (GSK3β) and PI3/Akt/response element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF). We queried PubMed, Web of Science, Scopus, Elsevier, and other related databases using search terms related to lithium and its neuroprotective effect in various neurodegenerative diseases and events from January 2000 to May 2022. We reviewed the major findings and mechanisms proposed for the effects of lithium. Lithium's neuroprotective potential against neural cell degeneration is mediated by inducing anti-inflammatory factors, antioxidant enzymes, and free radical scavengers to prevent mitochondrial dysfunction. Lithium effects are regulated by two essential pathways: PI3/Akt/GSK3β and PI3/Akt/CREB/BDNF. Lithium acts as a neuroprotective agent against neurodegeneration by preventing inflammation, oxidative stress, apoptosis, and mitochondrial dysfunction using PI3/Akt/GSK3β and PI3/Akt/CREB/BDNF signaling pathways.
Collapse
Affiliation(s)
- Fateme Ghanaatfar
- Student Research Committee, School of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Ghanaatfar
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
| | - Parisa Isapour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Negin Farokhi
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | | | - Mahshid Javadi
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Gholami
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, North Carolina, USA
| | - Natalie Coleman-Fuller
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, USA
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Jakob J, Kröger A, Klawonn F, Bruder D, Jänsch L. Translatome analyses by bio-orthogonal non-canonical amino acid labeling reveal that MR1-activated MAIT cells induce an M1 phenotype and antiviral programming in antigen-presenting monocytes. Front Immunol 2023; 14:1091837. [PMID: 36875139 PMCID: PMC9977998 DOI: 10.3389/fimmu.2023.1091837] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
MAIT cells are multifunctional innate-like effector cells recognizing bacterial-derived vitamin B metabolites presented by the non-polymorphic MHC class I related protein 1 (MR1). However, our understanding of MR1-mediated responses of MAIT cells upon their interaction with other immune cells is still incomplete. Here, we performed the first translatome study of primary human MAIT cells interacting with THP-1 monocytes in a bicellular system. We analyzed the interaction between MAIT and THP-1 cells in the presence of the activating 5-OP-RU or the inhibitory Ac-6-FP MR1-ligand. Using bio-orthogonal non-canonical amino acid tagging (BONCAT) we were able to enrich selectively those proteins that were newly translated during MR1-dependent cellular interaction. Subsequently, newly translated proteins were measured cell-type-specifically by ultrasensitive proteomics to decipher the coinciding immune responses in both cell types. This strategy identified over 2,000 MAIT and 3,000 THP-1 active protein translations following MR1 ligand stimulations. Translation in both cell types was found to be increased by 5-OP-RU, which correlated with their conjugation frequency and CD3 polarization at MAIT cell immunological synapses in the presence of 5-OP-RU. In contrast, Ac-6-FP only regulated a few protein translations, including GSK3B, indicating an anergic phenotype. In addition to known effector responses, 5-OP-RU-induced protein translations uncovered type I and type II Interferon-driven protein expression profiles in both MAIT and THP-1 cells. Interestingly, the translatome of THP-1 cells suggested that activated MAIT cells can impact M1/M2 polarization in these cells. Indeed, gene and surface expression of CXCL10, IL-1β, CD80, and CD206 confirmed an M1-like phenotype of macrophages being induced in the presence of 5-OP-RU-activated MAIT cells. Furthermore, we validated that the Interferon-driven translatome was accompanied by the induction of an antiviral phenotype in THP-1 cells, which were found able to suppress viral replication following conjugation with MR1-activated MAIT cells. In conclusion, BONCAT translatomics extended our knowledge of MAIT cell immune responses at the protein level and discovered that MR1-activated MAIT cells are sufficient to induce M1 polarization and an anti-viral program of macrophages.
Collapse
Affiliation(s)
- Josefine Jakob
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Medical Microbiology and Hospital Hygiene, Infection Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andrea Kröger
- Innate Immunity and Infection, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.,Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology, Health Campus Immunology, Infectiology and Inflammation, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Frank Klawonn
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dunja Bruder
- Institute of Medical Microbiology and Hospital Hygiene, Infection Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lothar Jänsch
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
14
|
Cribioli E, Giordano Attianese GMP, Ginefra P, Signorino-Gelo A, Vuillefroy de Silly R, Vannini N, Hess C, Irving M, Coukos G. Enforcing GLUT3 expression in CD8 + T cells improves fitness and tumor control by promoting glucose uptake and energy storage. Front Immunol 2022; 13:976628. [PMID: 36203587 PMCID: PMC9530831 DOI: 10.3389/fimmu.2022.976628] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
Despite the tremendous success of adoptive T-cell therapies (ACT) in fighting certain hematologic malignancies, not all patients respond, a proportion experience relapse, and effective ACT of most solid tumors remains elusive. In order to improve responses to ACT suppressive barriers in the solid tumor microenvironment (TME) including insufficient nutrient availability must be overcome. Here we explored how enforced expression of the high-affinity glucose transporter GLUT3 impacted tumor-directed T cells. Overexpression of GLUT3 in primary murine CD8+ T cells enhanced glucose uptake and increased glycogen and fatty acid storage, and was associated with increased mitochondrial fitness, reduced ROS levels, higher abundance of the anti-apoptotic protein Mcl-1, and better resistance to stress. Importantly, GLUT3-OT1 T cells conferred superior control of B16-OVA melanoma tumors and, in this same model, significantly improved survival. Moreover, a proportion of treated mice were cured and protected from re-challenge, indicative of long-term T cell persistence and memory formation. Enforcing expression of GLUT3 is thus a promising strategy to improve metabolic fitness and sustaining CD8+ T cell effector function in the context of ACT.
Collapse
Affiliation(s)
- Elisabetta Cribioli
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Lausanne University Hospital Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Greta Maria Paola Giordano Attianese
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Lausanne University Hospital Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Pierpaolo Ginefra
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Lausanne University Hospital Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Amandine Signorino-Gelo
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Lausanne University Hospital Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Romain Vuillefroy de Silly
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Lausanne University Hospital Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Nicola Vannini
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Lausanne University Hospital Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Christoph Hess
- Department of Biomedicine, Immunobiology, University of Basel and University Hospital of Basel, Basel, Switzerland
- Department of Medicine, Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
| | - Melita Irving
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Lausanne University Hospital Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - George Coukos
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, University of Lausanne and Lausanne University Hospital Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
15
|
Shaw G, Cavalcante L, Giles FJ, Taylor A. Elraglusib (9-ING-41), a selective small-molecule inhibitor of glycogen synthase kinase-3 beta, reduces expression of immune checkpoint molecules PD-1, TIGIT and LAG-3 and enhances CD8+ T cell cytolytic killing of melanoma cells. J Hematol Oncol 2022; 15:134. [PMID: 36104795 PMCID: PMC9472445 DOI: 10.1186/s13045-022-01352-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/25/2022] [Indexed: 11/26/2022] Open
Abstract
Background Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase with multiple roles in tumour growth, cell invasion and metastasis. We have previously established GSK-3 as an upstream regulator of PD-1 gene expression in CD8 + T cells and demonstrated that GSK-3 inhibition is as effective as anti-PD-1 mAb blockade in controlling tumour growth. Elraglusib (9-ING-41) is a specific small-molecule inhibitor of GSK-3β with clinical activity in patients with advanced cancers, including a patient with refractory melanoma whose response provided the rationale for the current study. Methods The B16 melanoma mouse model was used to observe the effect of elraglusib on tumour growth either as a single agent or in combination (simultaneously and sequentially) with anti-PD-1 mAb treatment. B16 tumour cells were implanted in either the flank, brain or both locations, and Kaplan–Meier plots were used to depict survival and significance determined using log rank tests. Expression of the immune checkpoint molecules, TIGIT, LAG-3 and PD-1, was evaluated using flow cytometry alongside expression of the chemokine receptor, CXCR3. Further evaluation of PD-1 expression was determined through RT-qPCR and immunohistochemistry. Results We demonstrated that elraglusib has a suppressive effect against melanoma as a single agent and enhanced anti-PD-1 therapy. There was a synergistic effect when elraglusib was used in combination with anti-PD-1 mAb, and an even greater effect when used as sequential therapy. Suppression of tumour growth was associated with a reduced expression of immune checkpoint molecules, PD-1, TIGIT and LAG-3 with upregulation of CXCR3 expression. Conclusions These data highlight the potential of elraglusib as an immune-modulatory agent and demonstrate the benefit of a sequential approach with immune checkpoint inhibition followed by GSK-3β inhibition in melanoma and provide a rationale for clinical investigation of elraglusib combined with immune checkpoint inhibitory molecules, including those targeting PD-1, TIGIT and LAG-3. This has several potential implications for current immunotherapy regimes, including possibly reducing the intensity of anti-PD-1 mAb treatment needed for response in patients receiving elraglusib, especially given the benign adverse event profile of elraglusib observed to date. Based on these data, a clinical study of elraglusib, an anti-PD-1 mAb and chemotherapy is ongoing (NCT NCT05239182). Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01352-x.
Collapse
|
16
|
Moore EK, Strazza M, Mor A. Combination Approaches to Target PD-1 Signaling in Cancer. Front Immunol 2022; 13:927265. [PMID: 35911672 PMCID: PMC9330480 DOI: 10.3389/fimmu.2022.927265] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer remains the second leading cause of death in the US, accounting for 25% of all deaths nationwide. Immunotherapy techniques bolster the immune cells' ability to target malignant cancer cells and have brought immense improvements in the field of cancer treatments. One important inhibitory protein in T cells, programmed cell death protein 1 (PD-1), has become an invaluable target for cancer immunotherapy. While anti-PD-1 antibody therapy is extremely successful in some patients, in others it fails or even causes further complications, including cancer hyper-progression and immune-related adverse events. Along with countless translational studies of the PD-1 signaling pathway, there are currently close to 5,000 clinical trials for antibodies against PD-1 and its ligand, PD-L1, around 80% of which investigate combinations with other therapies. Nevertheless, more work is needed to better understand the PD-1 signaling pathway and to facilitate new and improved evidence-based combination strategies. In this work, we consolidate recent discoveries of PD-1 signaling mediators and their therapeutic potential in combination with anti-PD-1/PD-L1 agents. We focus on the phosphatases SHP2 and PTPN2; the kinases ITK, VRK2, GSK-3, and CDK4/6; and the signaling adaptor protein PAG. We discuss their biology both in cancer cells and T cells, with a focus on their role in relation to PD-1 to determine their potential in therapeutic combinations. The literature discussed here was obtained from a search of the published literature and ClinicalTrials.gov with the following key terms: checkpoint inhibition, cancer immunotherapy, PD-1, PD-L1, SHP2, PTPN2, ITK, VRK2, CDK4/6, GSK-3, and PAG. Together, we find that all of these proteins are logical and promising targets for combination therapy, and that with a deeper mechanistic understanding they have potential to improve the response rate and decrease adverse events when thoughtfully used in combination with checkpoint inhibitors.
Collapse
Affiliation(s)
- Emily K. Moore
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| | - Marianne Strazza
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| | - Adam Mor
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
17
|
Barbian HJ, Seaton MS, Narasipura SD, Wallace J, Rajan R, Sha BE, Al-Harthi L. β-catenin regulates HIV latency and modulates HIV reactivation. PLoS Pathog 2022; 18:e1010354. [PMID: 35255110 PMCID: PMC8939789 DOI: 10.1371/journal.ppat.1010354] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 03/22/2022] [Accepted: 02/09/2022] [Indexed: 11/18/2022] Open
Abstract
Latency is the main obstacle towards an HIV cure, with cure strategies aiming to either elicit or prevent viral reactivation. While these strategies have shown promise, they have only succeeded in modulating latency in a fraction of the latent HIV reservoir, suggesting that the mechanisms controlling HIV latency are not completely understood, and that comprehensive latency modulation will require targeting of multiple latency maintenance pathways. We show here that the transcriptional co-activator and the central mediator of canonical Wnt signaling, β-catenin, inhibits HIV transcription in CD4+ T cells via TCF-4 LTR binding sites. Further, we show that inhibiting the β-catenin pathway reactivates HIV in a primary TCM cell model of HIV latency, primary cells from cART-controlled HIV donors, and in CD4+ latent cell lines. β-catenin inhibition or activation also enhanced or inhibited the activity of several classes of HIV latency reversing agents, respectively, in these models, with significant synergy of β-catenin and each LRA class tested. In sum, we identify β-catenin as a novel regulator of HIV latency in vitro and ex vivo, adding new therapeutic targets that may be combined for comprehensive HIV latency modulation in HIV cure efforts.
Collapse
Affiliation(s)
- Hannah J. Barbian
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Melanie S. Seaton
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Srinivas D. Narasipura
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Jennillee Wallace
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Reshma Rajan
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Beverly E. Sha
- Department of Internal Medicine, Division of Infectious Diseases, Rush University Medical Center, Chicago, Illinios United States of America
| | - Lena Al-Harthi
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
18
|
Lipp JJ, Wang L, Yang H, Yao F, Harrer N, Müller S, Berezowska S, Dorn P, Marti TM, Schmid RA, Hegedüs B, Souabni A, Carotta S, Pearson MA, Sommergruber W, Kocher GJ, Hall SR. Functional and molecular characterization of PD1+ tumor-infiltrating lymphocytes from lung cancer patients. Oncoimmunology 2022; 11:2019466. [PMID: 35154905 PMCID: PMC8837234 DOI: 10.1080/2162402x.2021.2019466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Antibody-mediated cancer immunotherapy targets inhibitory surface molecules, such as PD1, PD-L1, and CTLA-4, aiming to re-invigorate dysfunctional T cells. We purified and characterized tumor-infiltrating lymphocytes (TILs) and their patient-matched non-tumor counterparts from treatment-naïve NSCLC patient biopsies to evaluate the effect of PD1 expression on the functional and molecular profiles of tumor-resident T cells. We show that PD1+ CD8+ TILs have elevated expression of the transcriptional regulator ID3 and that the cytotoxic potential of CD8 T cells can be improved by knocking down ID3, defining it as a potential regulator of T cell effector function. PD1+ CD4+ memory TILs display transcriptional patterns consistent with both helper and regulator function, but can robustly facilitate B cell activation and expansion. Furthermore, we show that expanding ex vivo-prepared TILs in vitro broadly preserves their functionality with respect to tumor cell killing, B cell help, and TCR repertoire. Although purified PD1+ CD8+ TILs generally maintain an exhausted phenotype upon expansion in vitro, transcriptional analysis reveals a downregulation of markers of T-cell dysfunction, including the co-inhibitory molecules PD1 and CTLA-4 and transcription factors ID3, TOX and TOX2, while genes involved in cell cycle and DNA repair are upregulated. We find reduced expression of WNT signaling components to be a hallmark of PD1+ CD8+ exhausted T cells in vivo and in vitro and demonstrate that restoring WNT signaling, by pharmacological blockade of GSK3β, can improve effector function. These data unveil novel targets for tumor immunotherapy and have promising implications for the development of a personalized TIL-based cell therapy for lung cancer.
Collapse
Affiliation(s)
- Jesse J. Lipp
- Boehringer Ingelheim, Rcv GmbH & Co Kg, Vienna, Austria
| | - Limei Wang
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Haitang Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Feng Yao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | | | - Stefan Müller
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Patrick Dorn
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
| | - Thomas M. Marti
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Ralph A. Schmid
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Belazs Hegedüs
- Department of Thoracic Surgery, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | | | | | | | - Wolfgang Sommergruber
- Boehringer Ingelheim, Rcv GmbH & Co Kg, Vienna, Austria
- Department of Biotechnology, University of Applied Sciences, Vienna, Austria
| | - Greg J. Kocher
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
| | - Sean R.R. Hall
- Division of General Thoracic Surgery, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Abstract
The global coronavirus disease-19 (COVID-19) has affected more than 140 million and killed more than 3 million people worldwide as of April 20, 2021. The novel human severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been identified as an etiological agent for COVID-19. Several kinases have been proposed as possible mediators of multiple viral infections, including life-threatening coronaviruses like SARS-CoV-1, Middle East syndrome coronavirus (MERS-CoV), and SARS-CoV-2. Viral infections hijack abundant cell signaling pathways, resulting in drastic phosphorylation rewiring in the host and viral proteins. Some kinases play a significant role throughout the viral infection cycle (entry, replication, assembly, and egress), and several of them are involved in the virus-induced hyperinflammatory response that leads to cytokine storm, acute respiratory distress syndrome (ARDS), organ injury, and death. Here, we highlight kinases that are associated with coronavirus infections and their inhibitors with antiviral and potentially anti-inflammatory, cytokine-suppressive, or antifibrotic activity.
Collapse
Affiliation(s)
- Thanigaimalai Pillaiyar
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry
and Tuebingen Center for Academic Drug Discovery, Eberhard Karls University
Tübingen, Auf der Morgenstelle 8, 72076 Tübingen,
Germany
| | - Stefan Laufer
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry
and Tuebingen Center for Academic Drug Discovery, Eberhard Karls University
Tübingen, Auf der Morgenstelle 8, 72076 Tübingen,
Germany
| |
Collapse
|
20
|
Shiraz AK, Panther EJ, Reilly CM. Altered Germinal-Center Metabolism in B Cells in Autoimmunity. Metabolites 2022; 12:metabo12010040. [PMID: 35050162 PMCID: PMC8780703 DOI: 10.3390/metabo12010040] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022] Open
Abstract
B lymphocytes play an important role in the pathophysiology of many autoimmune disorders by producing autoantibodies, secreting cytokines, and presenting antigens. B cells undergo extreme physiological changes as they develop and differentiate. Aberrant function in tolerogenic checkpoints and the metabolic state of B cells might be the contributing factors to the dysfunctionality of autoimmune B cells. Understanding B-cell metabolism in autoimmunity is important as it can give rise to new treatments. Recent investigations have revealed that alterations in metabolism occur in the activation of B cells. Several reports have suggested that germinal center (GC) B cells of individuals with systemic lupus erythematosus (SLE) have altered metabolic function. GCs are unique microenvironments in which the delicate and complex process of B-cell affinity maturation occurs through somatic hypermutation (SHM) and class switching recombination (CSR) and where Bcl6 tightly regulates B-cell differentiation into memory B-cells or plasma cells. GC B cells rely heavily on glucose, fatty acids, and oxidative phosphorylation (OXPHOS) for their energy requirements. However, the complicated association between GC B cells and their metabolism is still not clearly understood. Here, we review several studies of B-cell metabolism, highlighting the significant transformations that occur in GC progression, and suggest possible approaches that may be investigated to more precisely target aberrant B-cell metabolism in SLE.
Collapse
Affiliation(s)
- Ashton K. Shiraz
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 205 Duck Pond Drive, Blacksburg, VA 24061, USA;
- Correspondence: (A.K.S.); (C.M.R.); Tel.: +1-540-231-9365 (C.M.R.)
| | - Eric J. Panther
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 205 Duck Pond Drive, Blacksburg, VA 24061, USA;
| | - Christopher M. Reilly
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 205 Duck Pond Drive, Blacksburg, VA 24061, USA;
- Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA
- Correspondence: (A.K.S.); (C.M.R.); Tel.: +1-540-231-9365 (C.M.R.)
| |
Collapse
|
21
|
Liu C, Ma L, Wang Y, Zhao J, Chen P, Chen X, Wang Y, Hu Y, Liu Y, Jia X, Yang Z, Yin X, Wu J, Wu S, Zheng H, Ma X, Sun X, He Y, Lin L, Fu Y, Liao K, Zhou X, Jiang S, Fu G, Tang J, Han W, Chen XL, Fan W, Hong Y, Han J, Huang X, Li BA, Xiao N, Xiao C, Fu G, Liu WH. Glycogen synthase kinase 3 drives thymocyte egress by suppressing β-catenin activation of Akt. SCIENCE ADVANCES 2021; 7:eabg6262. [PMID: 34623920 PMCID: PMC8500522 DOI: 10.1126/sciadv.abg6262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/19/2021] [Indexed: 06/13/2023]
Abstract
Molecular pathways controlling emigration of mature thymocytes from thymus to the periphery remain incompletely understood. Here, we show that T cell–specific ablation of glycogen synthase kinase 3 (GSK3) led to severely impaired thymic egress. In the absence of GSK3, β-catenin accumulated in the cytoplasm, where it associated with and activated Akt, leading to phosphorylation and degradation of Foxo1 and downregulation of Klf2 and S1P1 expression, thereby preventing emigration of thymocytes. A cytoplasmic membrane-localized β-catenin excluded from the nucleus promoted Akt activation, suggesting a new function of β-catenin independent of its role as a transcriptional activator. Furthermore, genetic ablation of β-catenin, retroviral expression of a dominant negative Akt mutant, and transgenic expression of a constitutively active Foxo1 restored emigration of GSK3-deficient thymocytes. Our findings establish an essential role for GSK3 in thymocyte egress and reveal a previously unidentified signaling function of β-catenin in the cytoplasm.
Collapse
Affiliation(s)
- Chenfeng Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Lei Ma
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yuxuan Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiayi Zhao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Pengda Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xian Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yingxin Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yanyan Hu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yun Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xian Jia
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhanghua Yang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xingzhi Yin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Suqin Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Haiping Zheng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaohong Ma
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiufeng Sun
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Ying He
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Lianghua Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yubing Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Kunyu Liao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaojuan Zhou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Shan Jiang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Guofeng Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jian Tang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wei Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiao Lei Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wenzhu Fan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yazhen Hong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiangyang Huang
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Bo-An Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Nengming Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Changchun Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
22
|
Park R, Coveler AL, Cavalcante L, Saeed A. GSK-3β in Pancreatic Cancer: Spotlight on 9-ING-41, Its Therapeutic Potential and Immune Modulatory Properties. BIOLOGY 2021; 10:biology10070610. [PMID: 34356465 PMCID: PMC8301062 DOI: 10.3390/biology10070610] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/15/2022]
Abstract
Simple Summary Glycogen synthase kinase-3 beta is a protein kinase implicated in the promotion and development of various cancers, including pancreatic cancer. In cell culture and animal studies, drugs targeting the inhibition of this protein show treatment potential in pancreatic cancer. Studies show targeting this protein for treatment may overcome resistance to conventional chemotherapy in pancreatic tumors. Early-stage clinical trials are currently studying small molecule inhibitors targeting glycogen synthase kinase-3 beta and interim results show favorable results. Recent studies also suggest that targeting this protein will create synergy with immunotherapy, such as checkpoint inhibitors. Future studies should aim to study new combination treatments involving glycogen synthase kinase-3 beta targeting drugs with chemotherapy and immunotherapy in pancreatic cancer. Abstract Glycogen synthase kinase-3 beta is a ubiquitously and constitutively expressed molecule with pleiotropic function. It acts as a protooncogene in the development of several solid tumors including pancreatic cancer through its involvement in various cellular processes including cell proliferation, survival, invasion and metastasis, as well as autophagy. Furthermore, the level of aberrant glycogen synthase kinase-3 beta expression in the nucleus is inversely correlated with tumor differentiation and survival in both in vitro and in vivo models of pancreatic cancer. Small molecule inhibitors of glycogen synthase kinase-3 beta have demonstrated therapeutic potential in pre-clinical models and are currently being evaluated in early phase clinical trials involving pancreatic cancer patients with interim results showing favorable results. Moreover, recent studies support a rationale for the combination of glycogen synthase kinase-3 beta inhibitors with chemotherapy and immunotherapy, warranting the evaluation of novel combination regimens in the future.
Collapse
Affiliation(s)
- Robin Park
- Department of Medicine, MetroWest Medical Center, Tufts University School of Medicine, Framingham, MA 01702, USA;
| | - Andrew L. Coveler
- Department of Medicine, Division of Oncology, University of Washington, Seattle, WA 98109-1024, USA;
| | | | - Anwaar Saeed
- Department of Medicine, Division of Medical Oncology, Kansas University Cancer Center & Research Institute, Kansas, KS 66205, USA
- Correspondence: ; Tel.: +1-913-588-6077
| |
Collapse
|
23
|
Park CH. Making Potent CAR T Cells Using Genetic Engineering and Synergistic Agents. Cancers (Basel) 2021; 13:cancers13133236. [PMID: 34209505 PMCID: PMC8269169 DOI: 10.3390/cancers13133236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 12/16/2022] Open
Abstract
Immunotherapies are emerging as powerful weapons for the treatment of malignancies. Chimeric antigen receptor (CAR)-engineered T cells have shown dramatic clinical results in patients with hematological malignancies. However, it is still challenging for CAR T cell therapy to be successful in several types of blood cancer and most solid tumors. Many attempts have been made to enhance the efficacy of CAR T cell therapy by modifying the CAR construct using combination agents, such as compounds, antibodies, or radiation. At present, technology to improve CAR T cell therapy is rapidly developing. In this review, we particularly emphasize the most recent studies utilizing genetic engineering and synergistic agents to improve CAR T cell therapy.
Collapse
Affiliation(s)
- Chi Hoon Park
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Daejeon 34114, Korea; ; Tel.: +82-42-860-7416; Fax: +82-42-861-4246
- Medicinal & Pharmaceutical Chemistry, Korea University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
24
|
Steele L, Mannion AJ, Shaw G, Maclennan KA, Cook GP, Rudd CE, Taylor A. Non-redundant activity of GSK-3α and GSK-3β in T cell-mediated tumor rejection. iScience 2021; 24:102555. [PMID: 34142056 PMCID: PMC8188550 DOI: 10.1016/j.isci.2021.102555] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/13/2021] [Accepted: 05/14/2021] [Indexed: 12/21/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is a positive regulator of PD-1 expression in CD8+ T cells and GSK-3 inhibition enhances T cell function and is effective in the control of tumor growth. GSK-3 has two co-expressed isoforms, GSK-3α and GSK-3β. Using conditional gene targeting, we demonstrate that both isoforms contribute to T cell function to different degrees. Gsk3b-/- mice suppressed tumor growth to the same degree as Gsk3a/b-/- mice, whereas Gsk3a-/- mice behaved similarly to wild-type, revealing an important role for GSK-3β in regulating T cell-mediated anti-tumor immunity. The individual GSK-3α and β isoforms have differential effects on PD-1, IFNγ, and granzyme B expression and operate in synergy to control PD-1 expression and the infiltration of tumors with CD4 and CD8 T cells. Our data reveal a complex interplay of the GSK-3 isoforms in the control of tumor immunity and highlight non-redundant activity of GSK-3 isoforms in T cells, with implications for immunotherapy.
Collapse
Affiliation(s)
- Lynette Steele
- Leeds Institute of Medical Research, University of Leeds, School of Medicine, Wellcome Trust Brenner Building, St James's University Hospital, Leeds LS9 7TF, UK
| | - Aarren J. Mannion
- Leeds Institute of Medical Research, University of Leeds, School of Medicine, Wellcome Trust Brenner Building, St James's University Hospital, Leeds LS9 7TF, UK
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Gary Shaw
- Leeds Institute of Medical Research, University of Leeds, School of Medicine, Wellcome Trust Brenner Building, St James's University Hospital, Leeds LS9 7TF, UK
| | - Kenneth A. Maclennan
- Leeds Institute of Medical Research, University of Leeds, School of Medicine, Wellcome Trust Brenner Building, St James's University Hospital, Leeds LS9 7TF, UK
| | - Graham P. Cook
- Leeds Institute of Medical Research, University of Leeds, School of Medicine, Wellcome Trust Brenner Building, St James's University Hospital, Leeds LS9 7TF, UK
| | - Christopher E. Rudd
- Division of Immunology-Oncology Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada
- Département de Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University Health Center, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Alison Taylor
- Leeds Institute of Medical Research, University of Leeds, School of Medicine, Wellcome Trust Brenner Building, St James's University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
25
|
Dempke WCM, Fenchel K. Has programmed cell death ligand-1 MET an accomplice in non-small cell lung cancer?-a narrative review. Transl Lung Cancer Res 2021; 10:2667-2682. [PMID: 34295669 PMCID: PMC8264346 DOI: 10.21037/tlcr-21-124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022]
Abstract
Recently approved and highly specific small-molecule inhibitors of c-MET exon 14 skipping mutations (e.g., capmatinib, tepotinib) are a new and important therapeutic option for the treatment of non-small cell lung cancer (NSCLC) patients harbouring c-MET alterations. Several experimental studies have provided compelling evidence that c-MET is involved in the regulation of the immune response by up-regulating inhibitory molecules (e.g., PD-L1) and down-regulating of immune stimulators (e.g., CD137, CD252, CD70, etc.). In addition, c-MET was found to be implicated in the regulation of the inflamed tumour microenvironment (TME) and thereby contributing to an increased immune escape of tumour cells from T cell killing. Moreover, it is a major resistance mechanism following treatment of epidermal growth factor receptor mutations (EGFRmut) with tyrosine kinase receptor inhibitors (TKIs). In line with these findings c-MET alterations have also been shown to be associated with a worse clinical outcome and a poorer prognosis in NSCLC patients. However, the underlying mechanisms for these experimental observations are neither fully evaluated nor conclusive, but clearly multifactorial and most likely tumour-specific. In this regard the clinical efficacy of checkpoint inhibitors (CPIs) and TKIs against EGFRmut in NSCLC patients harbouring c-MET alterations is also not yet established, and further research will certainly provide some guidance as to optimally utilise CPIs and c-MET inhibitors in the future.
Collapse
Affiliation(s)
- Wolfram C M Dempke
- Department of Haematology and Oncology, University of Munich, Munich, Germany
| | | |
Collapse
|
26
|
He R, Du S, Lei T, Xie X, Wang Y. Glycogen synthase kinase 3β in tumorigenesis and oncotherapy (Review). Oncol Rep 2020; 44:2373-2385. [PMID: 33125126 PMCID: PMC7610307 DOI: 10.3892/or.2020.7817] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/17/2020] [Indexed: 02/05/2023] Open
Abstract
Glycogen synthase kinase 3β (GSK 3β), a multifunctional serine and threonine kinase, plays a critical role in a variety of cellular activities, including signaling transduction, protein and glycogen metabolism, cell proliferation, cell differentiation, and apoptosis. Therefore, aberrant regulation of GSK 3β results in a broad range of human diseases, such as tumors, diabetes, inflammation and neurodegenerative diseases. Accumulating evidence has suggested that GSK 3β is correlated with tumorigenesis and progression. However, GSK 3β is controversial due to its bifacial roles of tumor suppression and activation. In addition, overexpression of GSK 3β is involved in tumor growth, whereas it contributes to the cell sensitivity to chemotherapy. However, the underlying regulatory mechanisms of GSK 3β in tumorigenesis remain obscure and require further in‑depth investigation. In this review, we comprehensively summarize the roles of GSK 3β in tumorigenesis and oncotherapy, and focus on its potentials as an available target in oncotherapy.
Collapse
Affiliation(s)
- Rui He
- Department of Union, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Suya Du
- Department of Clinical Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Tiantian Lei
- Department of Pharmacy, Chongqing Health Center for Women and Children, Chongqing 400013, P.R. China
| | - Xiaofang Xie
- Department of Medicine, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Yi Wang
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
- Center of Translational Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
27
|
Jiang C, Liu J, Guo M, Gao X, Wu X, Bai N, Guo W, Li N, Yi F, Cheng R, Xu H, Zhou T, Jiang B, Sun T, Wei S, Cao L. The NAD-dependent deacetylase SIRT2 regulates T cell differentiation involved in tumor immune response. Int J Biol Sci 2020; 16:3075-3084. [PMID: 33061819 PMCID: PMC7545715 DOI: 10.7150/ijbs.49735] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
Sirtuin 2 (SIRT2), an NAD+-dependent deacetylase, regulates multiple biologic and pathologic processes including mitosis, genomic integrity, cell homeostasis and tumorigenesis. However, the role of SIRT2 in the immune response to cancer remains largely elusive. In this study, we found significantly lower expression of SIRT2 in peripheral T lymphocytes from breast cancer patients when compared to normal individuals. Moreover, SIRT2 levels positively correlated with CD8+ effector memory T (TEM) cells in breast cancer patients. In keeping with these findings, altered T cells differentiation manifested as decreased TEM cells and increased naive T cells were observed in Sirt2 deficient mice. The upregulation of CD8+ TEM by SIRT2 might attribute to the activation of aerobic oxidation as well as the inhibition of GSK3β acetylation in CD8+ T cells. Taken together, these results suggest that SIRT2 participate in tumor immune response by regulating T cell differentiation, which may provide novel insight for tumor prevention and immune therapy.
Collapse
Affiliation(s)
- Cui Jiang
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China.,Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and & Institute, 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning, China
| | - Jingwei Liu
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Min Guo
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Xiaoxin Gao
- Central laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and & Institute, 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning, China
| | - Xuan Wu
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Ning Bai
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Wendong Guo
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Na Li
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Fei Yi
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Rong Cheng
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Hongde Xu
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Tingting Zhou
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Bo Jiang
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| | - Tao Sun
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and & Institute, 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning, China
| | - Shi Wei
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35249-7331, USA
| | - Liu Cao
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis, Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110042, Liaoning, China
| |
Collapse
|
28
|
Dimou A, Syrigos KN. The Role of GSK3β in T Lymphocytes in the Tumor Microenvironment. Front Oncol 2020; 10:1221. [PMID: 32850361 PMCID: PMC7396595 DOI: 10.3389/fonc.2020.01221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 06/15/2020] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy options for patients with cancer have emerged following decades of research on immune responses against tumors. Most treatments in this category harness T cells with specificity for tumor associated antigens, neoantigens, and cancer-testis antigens. GSK3β is a serine-threonine kinase with the highest number of substrates and multifaceted roles in cell function including immune cells. Importantly, inhibitors of GSK3β are available for clinical and research use. Here, we review the possible role of GSK3β in the immune tumor microenvironment, with goal to guide future research that tests GSK3β inhibition as an immunotherapy adjunct.
Collapse
Affiliation(s)
- Anastasios Dimou
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | - Konstantinos N Syrigos
- Division of Medical Oncology, Third Department of Medicine, University of Athens, Athens, Greece
| |
Collapse
|
29
|
Rudd CE. GSK-3 Inhibition as a Therapeutic Approach Against SARs CoV2: Dual Benefit of Inhibiting Viral Replication While Potentiating the Immune Response. Front Immunol 2020; 11:1638. [PMID: 32695123 PMCID: PMC7333796 DOI: 10.3389/fimmu.2020.01638] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/18/2020] [Indexed: 01/04/2023] Open
Abstract
The SARS-CoV2 (COVID-19) pandemic and uncertainties in developing a vaccine have created an urgent need for new therapeutic approaches. A key question is whether it is possible to make rational predictions of new therapies based on the presently available scientific and medical information. In this regard, I have noticed an omission in the present analysis in the literature related to the exploitation of glycogen synthase kinase 3 (GSK-3) as a therapeutic approach. This is based on two key observations, that GSK-3 inhibitors can simultaneously block SARs viral replication, while boosting CD8+ adaptive T-cell and innate natural killer (NK) responses. Firstly, it is already clear that GSK-3 phosphorylation of SARs CoV1 N protein on key serine residues is needed for viral replication such that small molecule inhibitors (SMIs) of GSK-3 can inhibit viral replication. In comparing protein sequences, I show here that the key sites in the N protein of SARs CoV1 N for replication are conserved in SARs CoV2. This strongly suggests that GSK-3 SMIs will also inhibit SARs Cov2 replication. Secondly, we and others have previously documented that GSK-3 SMIs markedly enhance CD8+ cytolytic T-cell (CTL) and NK cell anti-viral effector functions leading to a reduction in both acute and chronic viral infections in mice. My hypothesis is that the repurposing of low-cost inhibitors of GSK-3 such as lithium will limit SARS-CoV2 infections by both reducing viral replication and potentiating the immune response against the virus. To date, there has been no mention of this dual connection between GSK-3 and SARs CoV2 in the literature. To my knowledge, no other drugs exist with the potential to simultaneously target both viral replication and immune response against SARs CoV2.
Collapse
Affiliation(s)
- Christopher E Rudd
- Centre de Recherche- Maisonneuve-Rosemont Hospital (CR-HMR), Montreal, QC, Canada.,Département de Medicine, Université de Montréal, Montreal, QC, Canada.,Department of Medicine, Division of Endocrinology & Medical Biochemistry, McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
30
|
Augello G, Emma MR, Cusimano A, Azzolina A, Montalto G, McCubrey JA, Cervello M. The Role of GSK-3 in Cancer Immunotherapy: GSK-3 Inhibitors as a New Frontier in Cancer Treatment. Cells 2020; 9:cells9061427. [PMID: 32526891 PMCID: PMC7348946 DOI: 10.3390/cells9061427] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/31/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
The serine/threonine kinase glycogen synthase kinase-3 (GSK-3) was initially identified because of its key role in the regulation of glycogen synthesis. However, it is now well-established that GSK-3 performs critical functions in many cellular processes, such as apoptosis, tumor growth, cell invasion, and metastasis. Aberrant GSK-3 activity has been associated with many human diseases, including cancer, highlighting its potential therapeutic relevance as a target for anticancer therapy. Recently, newly emerging data have demonstrated the pivotal role of GSK-3 in the anticancer immune response. In the last few years, many GSK-3 inhibitors have been developed, and some are currently being tested in clinical trials. This review will discuss preclinical and initial clinical results with GSK-3β inhibitors, highlighting the potential importance of this target in cancer immunotherapy. As described in this review, GSK-3 inhibitors have been shown to have antitumor activity in a wide range of human cancer cells, and they may also contribute to promoting a more efficacious immune response against tumor target cells, thus showing a double therapeutic advantage.
Collapse
Affiliation(s)
- Giuseppa Augello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), 90144 Palermo, Italy; (G.A.); (M.R.E.); (A.C.); (A.A.); (G.M.)
| | - Maria R. Emma
- Institute for Biomedical Research and Innovation, National Research Council (CNR), 90144 Palermo, Italy; (G.A.); (M.R.E.); (A.C.); (A.A.); (G.M.)
| | - Antonella Cusimano
- Institute for Biomedical Research and Innovation, National Research Council (CNR), 90144 Palermo, Italy; (G.A.); (M.R.E.); (A.C.); (A.A.); (G.M.)
| | - Antonina Azzolina
- Institute for Biomedical Research and Innovation, National Research Council (CNR), 90144 Palermo, Italy; (G.A.); (M.R.E.); (A.C.); (A.A.); (G.M.)
| | - Giuseppe Montalto
- Institute for Biomedical Research and Innovation, National Research Council (CNR), 90144 Palermo, Italy; (G.A.); (M.R.E.); (A.C.); (A.A.); (G.M.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - James A. McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA;
| | - Melchiorre Cervello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), 90144 Palermo, Italy; (G.A.); (M.R.E.); (A.C.); (A.A.); (G.M.)
- Correspondence: ; Tel.: +39-091-6809-534
| |
Collapse
|
31
|
Glycogen Synthase Kinase-3β Facilitates Cytokine Production in 12-O-Tetradecanoylphorbol-13-Acetate/Ionomycin-Activated Human CD4 + T Lymphocytes. Cells 2020; 9:cells9061424. [PMID: 32521784 PMCID: PMC7348852 DOI: 10.3390/cells9061424] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/02/2022] Open
Abstract
Cytokines are the major immune regulators secreted from activated CD4+ T lymphocytes that activate adaptive immunity to eradicate nonself cells, including pathogens, tumors, and allografts. The regulation of glycogen synthase kinase (GSK)-3β, a serine/threonine kinase, controls cytokine production by regulating transcription factors. The artificial in vitro activation of CD4+ T lymphocytes by a combination of 12-O-tetradecanoylphorbol-13-acetate and ionomycin, the so-called T/I model, led to an inducible production of cytokines, such as interferon-γ, tumor necrosis factor-α, and interleukin-2. As demonstrated by the approaches of pharmacological targeting and genetic knockdown of GSK-3β, T/I treatment effectively caused GSK-3β activation followed by GSK-3β-regulated cytokine production. In contrast, pharmacological inhibition of the proline-rich tyrosine kinase 2 and calcineurin signaling pathways blocked cytokine production, probably by deactivating GSK-3β. The blockade of GSK-3β led to the inhibition of the nuclear translocation of T-bet, a vital transcription factor of T lymphocyte cytokines. In a mouse model, treatment with the GSK-3β inhibitor 6-bromoindirubin-3’-oxime significantly inhibited T/I-induced mortality and serum cytokine levels. In summary, targeting GSK-3β effectively inhibits CD4+ T lymphocyte activation and cytokine production.
Collapse
|
32
|
Krueger J, Rudd CE, Taylor A. Glycogen synthase 3 (GSK-3) regulation of PD-1 expression and and its therapeutic implications. Semin Immunol 2020; 42:101295. [PMID: 31604533 DOI: 10.1016/j.smim.2019.101295] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022]
Abstract
The past few years have witnessed exciting progress in the application of immune check-point blockade (ICB) for the treatment of various human cancers. ICB was first used against cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) to demonstrate durable anti-tumor responses followed by ICB against programmed cell death-1 (PD-1) or its ligand, PD-L1. Present approaches involve the use of combinations of blocking antibodies against CTLA-4, PD-1 and other inhibitory receptors (IRs) such as TIM3, TIGIT and LAG3. Despite this success, most patients are not cured by ICB therapy and there are limitations to the use of antibodies including cost, tumor penetration, the accessibility of receptors, and clearance from the cell surface as well as inflammatory and autoimmune complications. Recently, we demonstrated that the down-regulation or inhibition of glycogen synthase kinase 3 (GSK-3) down-regulates PD-1 expression in infectious diseases and cancer (Taylor et al., 2016 Immunity 44, 274-86; 2018 Cancer Research 78, 706-717; Krueger and Rudd 2018 Immunity 46, 529-531). In this Review, we outline the use of small molecule inhibitors (SMIs) that target intracellular pathways for co-receptor blockade in cancer immunotherapy.
Collapse
Affiliation(s)
- Janna Krueger
- Division of Immunology-Oncology, Research Center Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; Département de Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Christopher E Rudd
- Division of Immunology-Oncology, Research Center Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; Département de Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada.
| | - Alison Taylor
- Leeds Institute of Medical Research, University of Leeds, School of Medicine, Wellcome Trust Brenner Building, St James's University Hospital, LEEDS LS9 7TF, United Kingdom.
| |
Collapse
|
33
|
Saravia J, Raynor JL, Chapman NM, Lim SA, Chi H. Signaling networks in immunometabolism. Cell Res 2020; 30:328-342. [PMID: 32203134 PMCID: PMC7118125 DOI: 10.1038/s41422-020-0301-1] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Adaptive immunity is essential for pathogen and tumor eradication, but may also trigger uncontrolled or pathological inflammation. T cell receptor, co-stimulatory and cytokine signals coordinately dictate specific signaling networks that trigger the activation and functional programming of T cells. In addition, cellular metabolism promotes T cell responses and is dynamically regulated through the interplay of serine/threonine kinases, immunological cues and nutrient signaling networks. In this review, we summarize the upstream regulators and signaling effectors of key serine/threonine kinase-mediated signaling networks, including PI3K–AGC kinases, mTOR and LKB1–AMPK pathways that regulate metabolism, especially in T cells. We also provide our perspectives about the pending questions and clinical applicability of immunometabolic signaling. Understanding the regulators and effectors of immunometabolic signaling networks may uncover therapeutic targets to modulate metabolic programming and T cell responses in human disease.
Collapse
Affiliation(s)
- Jordy Saravia
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jana L Raynor
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Nicole M Chapman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Seon Ah Lim
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
34
|
Taylor A, Rudd CE. Glycogen synthase kinase 3 (GSK-3) controls T-cell motility and interactions with antigen presenting cells. BMC Res Notes 2020; 13:163. [PMID: 32188506 PMCID: PMC7079518 DOI: 10.1186/s13104-020-04971-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 02/24/2020] [Indexed: 12/21/2022] Open
Abstract
Objective The threonine/serine kinase glycogen synthase kinase 3 (GSK-3) targets multiple substrates in T-cells, regulating the expression of Tbet and PD-1 on T-cells. However, it has been unclear whether GSK-3 can affect the motility of T-cells and their interactions with antigen presenting cells. Results Here, we show that GSK-3 controls T-cell motility and interactions with other cells. Inhibition of GSK-3, using structurally distinct inhibitors, reduced T-cell motility in terms of distance and displacement. While SB415286 reduced the number of cell-cell contacts, the dwell times of cells that established contacts with other cells did not differ for T-cells treated with SB415286. Further, the increase in cytolytic T-cell (CTL) function in killing tumor targets was not affected by the inhibition of motility. This data shows that the inhibition of GSK-3 has differential effects on T-cell motility and CTL function where the negative effects on cell–cell interactions is overridden by the increased cytolytic potential of CTLs.
Collapse
Affiliation(s)
- Alison Taylor
- Leeds Institute of Medical Research, School of Medicine, University of Leeds, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK. .,Cell Signalling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1Q, UK.
| | - Christopher E Rudd
- Cell Signalling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1Q, UK. .,Division of Immunology-Oncology Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, H1T 2M4, Canada. .,Département de Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
| |
Collapse
|
35
|
Xiao D, Zhou Q, Gao Y, Cao B, Zhang Q, Zeng G, Zong S. PDK1 is important lipid kinase for RANKL-induced osteoclast formation and function via the regulation of the Akt-GSK3β-NFATc1 signaling cascade. J Cell Biochem 2020; 121:4542-4557. [PMID: 32048762 DOI: 10.1002/jcb.29677] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/16/2020] [Indexed: 12/17/2022]
Abstract
Perturbations in the balanced process of osteoblast-mediated bone formation and osteoclast-mediated bone resorption leading to excessive osteoclast formation and/or activity is the cause of many pathological bone conditions such as osteoporosis. The osteoclast is the only cell in the body capable of resorbing and degrading the mineralized bone matrix. Osteoclast formation from monocytic precursors is governed by the actions of two key cytokines macrophage-colony-stimulating factor and receptor activator of nuclear factor-κB ligand (RANKL). Binding of RANKL binding to receptor RANK initiates a series of downstream signaling responses leading to monocytic cell differentiation and fusion, and subsequent mature osteoclast bone resorption and survival. The phosphoinositide-3-kinase (PI3K)-protein kinase B (Akt) signaling cascade is one such pathway activated in response to RANKL. The 3-phosphoinositide-dependent protein kinase 1 (PDK1), is considered the master upstream lipid kinase of the PI3K-Akt cascade. PDK1 functions to phosphorylate and partially activate Akt, triggering the activation of downstream effectors. However, the role of PDK1 in osteoclasts has yet to be clearly defined. In this study, we specifically deleted the PDK1 gene in osteoclasts using the cathepsin-K promoter driven Cre-LoxP system. We found that the specific genetic ablation of PDK1 in osteoclasts leads to an osteoclast-poor osteopetrotic phenotype in mice. In vitro cellular assays further confirmed the impairment of osteoclast formation in response to RANKL by PDK1-deficient bone marrow macrophage (BMM) precursor cells. PDK1-deficient BMMs exhibited reduced ability to reorganize actin cytoskeleton to form a podosomal actin belt as a result of diminished capacity to fuse into giant multinucleated osteoclasts. Notably, biochemical analyses showed that PDK1 deficiency attenuated the phosphorylation of Akt and downstream effector GSK3β, and reduced induction of NFATc1. GSK3β is a reported negative regulator of NFATc1. GSK3β activity is inhibited by Akt-dependent phosphorylation. Thus, our data provide clear genetic and mechanistic insights into the important role for PDK1 in osteoclasts.
Collapse
Affiliation(s)
- Dongliang Xiao
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Quan Zhou
- Collaborative Innovation Center of Guangxi Biological Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Yunbing Gao
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Baichuan Cao
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Qiong Zhang
- College of Public Hygiene of Guangxi Medical University, Guangxi, China
| | - Gaofeng Zeng
- College of Public Hygiene of Guangxi Medical University, Guangxi, China
| | - Shaohui Zong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Research Centre for Regenerative Medicine and Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
36
|
Smith WM, Purvis IJ, Bomstad CN, Labak CM, Velpula KK, Tsung AJ, Regan JN, Venkataraman S, Vibhakar R, Asuthkar S. Therapeutic targeting of immune checkpoints with small molecule inhibitors. Am J Transl Res 2019; 11:529-541. [PMID: 30899360 PMCID: PMC6413273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/18/2018] [Indexed: 06/09/2023]
Abstract
Immune checkpoints are known to contribute to tumor progression by enhancing cancer's ability to evade the immune system and metastasize. Immunotherapies, including monoclonal antibodies, have been developed to target specific immunosuppressive molecules on the membranes of cancer cells and have proven revolutionary in the field of oncology. Recently, small molecule inhibitors (SMIs) have gained increased attention in cancer research with potential applications in immunotherapy. SMIs have desirable benefits over large-molecule inhibitors, such as monoclonal antibodies, including greater cell permeability, organ specificity, longer half-lives, cheaper production costs, and the possibility for oral administration. This paper will review the mechanisms by which noteworthy and novel immune checkpoints contribute to tumor progression, and how they may be targeted by SMIs and epigenetic modifiers to offer possible adjuvants to established therapeutic regimens. SMIs target immune checkpoints in several ways, such as blocking signaling between tumorigenic factors, building immune tolerance, and direct inhibition via epigenetic repression of immune inhibitory molecules. Further investigation into combination therapies utilizing SMIs and conventional cancer therapies will uncover new treatment options that may provide better patient outcomes across a range of cancers.
Collapse
Affiliation(s)
- Wade M Smith
- Department of Cancer Biology and Pharmacology, University of Illinois College of MedicinePeoria, IL
| | - Ian J Purvis
- Department of Cancer Biology and Pharmacology, University of Illinois College of MedicinePeoria, IL
| | - Colin N Bomstad
- Department of Cancer Biology and Pharmacology, University of Illinois College of MedicinePeoria, IL
| | - Collin M Labak
- Department of Cancer Biology and Pharmacology, University of Illinois College of MedicinePeoria, IL
| | - Kiran K Velpula
- Department of Cancer Biology and Pharmacology, University of Illinois College of MedicinePeoria, IL
- Department of Neurosurgery, University of Illinois College of MedicinePeoria, IL
| | - Andrew J Tsung
- Department of Cancer Biology and Pharmacology, University of Illinois College of MedicinePeoria, IL
- Department of Neurosurgery, University of Illinois College of MedicinePeoria, IL
- Department of Illinois Neurological Institute, University of Illinois College of MedicinePeoria, IL
| | - Jenna N Regan
- Department of Health Sciences Education, University of Illinois College of MedicinePeoria, IL
| | | | - Rajeev Vibhakar
- Department of Pediatrics, University of Colorado School of MedicineAurora, CO
| | - Swapna Asuthkar
- Department of Cancer Biology and Pharmacology, University of Illinois College of MedicinePeoria, IL
| |
Collapse
|
37
|
Chuntova P, Downey KM, Hegde B, Almeida ND, Okada H. Genetically Engineered T-Cells for Malignant Glioma: Overcoming the Barriers to Effective Immunotherapy. Front Immunol 2019; 9:3062. [PMID: 30740109 PMCID: PMC6357938 DOI: 10.3389/fimmu.2018.03062] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
Malignant gliomas carry a dismal prognosis. Conventional treatment using chemo- and radiotherapy has limited efficacy with adverse events. Therapy with genetically engineered T-cells, such as chimeric antigen receptor (CAR) T-cells, may represent a promising approach to improve patient outcomes owing to their potential ability to attack highly infiltrative tumors in a tumor-specific manner and possible persistence of the adaptive immune response. However, the unique anatomical features of the brain and susceptibility of this organ to irreversible tissue damage have made immunotherapy especially challenging in the setting of glioma. With safety concerns in mind, multiple teams have initiated clinical trials using CAR T-cells in glioma patients. The valuable lessons learnt from those trials highlight critical areas for further improvement: tackling the issues of the antigen presentation and T-cell homing in the brain, immunosuppression in the glioma microenvironment, antigen heterogeneity and off-tumor toxicity, and the adaptation of existing clinical therapies to reflect the intricacies of immune response in the brain. This review summarizes the up-to-date clinical outcomes of CAR T-cell clinical trials in glioma patients and examines the most pressing hurdles limiting the efficacy of these therapies. Furthermore, this review uses these hurdles as a framework upon which to evaluate cutting-edge pre-clinical strategies aiming to overcome those barriers.
Collapse
Affiliation(s)
- Pavlina Chuntova
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Kira M Downey
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Bindu Hegde
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Neil D Almeida
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States.,George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States.,The Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States.,Cancer Immunotherapy Program, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
38
|
Patterson AR, Bolcas P, Lampe K, Cantrell R, Ruff B, Lewkowich I, Hogan SP, Janssen EM, Bleesing J, Khurana Hershey GK, Hoebe K. Loss of GTPase of immunity-associated protein 5 (Gimap5) promotes pathogenic CD4 + T-cell development and allergic airway disease. J Allergy Clin Immunol 2019; 143:245-257.e6. [PMID: 30616774 PMCID: PMC6327968 DOI: 10.1016/j.jaci.2018.10.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 09/14/2018] [Accepted: 10/07/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND GTPase of immunity-associated protein 5 (GIMAP5) is essential for lymphocyte homeostasis and survival. Recently, human GIMAP5 single nucleotide polymorphisms have been linked to an increased risk for asthma, whereas loss of Gimap5 in mice has been associated with severe CD4+ T cell-driven immune pathology. OBJECTIVE We sought to identify the molecular and cellular mechanisms by which Gimap5 deficiency predisposes to allergic airway disease. METHODS CD4+ T-cell polarization and development of pathogenic CD4+ T cells were assessed in Gimap5-deficient mice and a human patient with a GIMAP5 loss-of-function (LOF) mutation. House dust mite-induced airway inflammation was assessed by using a complete Gimap5 LOF (Gimap5sph/sph) and conditional Gimap5fl/flCd4Cre/ert2 mice. RESULTS GIMAP5 LOF mutations in both mice and human subjects are associated with spontaneous polarization toward pathogenic TH17 and TH2 cells in vivo. Mechanistic studies in vitro reveal that impairment of Gimap5-deficient TH cell differentiation is associated with increased DNA damage, particularly during TH1-polarizing conditions. DNA damage in Gimap5-deficient CD4+ T cells could be controlled by TGF-β, thereby promoting TH17 polarization. When challenged with house dust mite in vivo, Gimap5-deficient mice displayed an exacerbated asthma phenotype (inflammation and airway hyperresponsiveness), with increased development of TH2, TH17, and pathogenic TH17/TH2 cells. CONCLUSION Activation of Gimap5-deficient CD4+ T cells is associated with increased DNA damage and reduced survival that can be overcome by TGF-β. This leads to selective survival of pathogenic TH17 cells but also TH2 cells in human subjects and mice, ultimately promoting allergic airway disease.
Collapse
Affiliation(s)
- Andrew R Patterson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Paige Bolcas
- Division of Asthma Research, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kristin Lampe
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio
| | - Rachel Cantrell
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Brandy Ruff
- Division of Asthma Research, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio
| | - Ian Lewkowich
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Simon P Hogan
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Edith M Janssen
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jack Bleesing
- Division of Bone Marrow Transplantation & Immune Deficiency, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kasper Hoebe
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
39
|
Small Molecule Inhibition of Glycogen Synthase Kinase-3 in Cancer Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1164:225-233. [PMID: 31576552 DOI: 10.1007/978-3-030-22254-3_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Immune checkpoint blockade (ICB) has proved successful in the immunotherapeutic treatment of various human cancers. Despite its success, most patients are still not cured while immunogenic cold cancers are still poorly responsive. There is a need for novel clinical interventions in immunotherapy, either alone or in conjunction with ICB. Here, we outline our recent discovery that the intracellular signaling kinase glycogen synthase kinase-3 (GSK-3) is a central regulator of PD-1 in T-cells. We demonstrate the application of small molecule inhibitor (SMI) approaches to down-regulate PD-1 in tumor immunotherapy. GSK-3 SMIs were found as effective as anti-PD-1 in the elimination of melanoma in mouse models. We propose the development of novel SMIs to target co-receptors for the future of immunotherapy.
Collapse
|
40
|
Colomar-Carando N, Meseguer A, Company-Garrido I, Jutz S, Herrera-Fernández V, Olvera A, Kiefer K, Brander C, Steinberger P, Vicente R. Zip6 Transporter Is an Essential Component of the Lymphocyte Activation Machinery. THE JOURNAL OF IMMUNOLOGY 2018; 202:441-450. [PMID: 30552163 DOI: 10.4049/jimmunol.1800689] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/13/2018] [Indexed: 11/19/2022]
Abstract
Zinc deficiency causes immune dysfunction. In T lymphocytes, hypozincemia promotes thymus atrophy, polarization imbalance, and altered cytokine production. Zinc supplementation is commonly used to boost immune function to prevent infectious diseases in at-risk populations. However, the molecular players involved in zinc homeostasis in lymphocytes are poorly understood. In this paper, we wanted to determine the identity of the transporter responsible for zinc entry into lymphocytes. First, in human Jurkat cells, we characterized the effect of zinc on proliferation and activation and found that zinc supplementation enhances activation when T lymphocytes are stimulated using anti-CD3/anti-CD28 Abs. We show that zinc entry depends on specific pathways to correctly tune the NFAT, NF-κB, and AP-1 activation cascades. Second, we used various human and murine models to characterize the zinc transporter family, Zip, during T cell activation and found that Zip6 was strongly upregulated early during activation. Therefore, we generated a Jurkat Zip6 knockout (KO) line to study how the absence of this transporter affects lymphocyte physiology. We found that although Zip6KO cells showed no altered zinc transport or proliferation under basal conditions, under activation, these KO cells showed deficient zinc transport and a drastically impaired activation program. Our work shows that zinc entry into activated lymphocytes depends on Zip6 and that this transporter is essential for the correct function of the cellular activation machinery.
Collapse
Affiliation(s)
- Natalia Colomar-Carando
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain
| | - Alberto Meseguer
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain
| | - Iván Company-Garrido
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain
| | - Sabrina Jutz
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Víctor Herrera-Fernández
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain
| | - Alex Olvera
- HIVACAT, IrsiCaixa AIDS Research Institute, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain.,University of Vic and Central Catalonia, 08500 Vic, Spain; and
| | - Kerstin Kiefer
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain
| | - Christian Brander
- HIVACAT, IrsiCaixa AIDS Research Institute, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain.,University of Vic and Central Catalonia, 08500 Vic, Spain; and.,Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rubén Vicente
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Pompeu Fabra University, 08003 Barcelona, Spain;
| |
Collapse
|
41
|
Ruan L, Liu H, Shi H. Characterization and function of GSK3β from Litopenaeus vannamei in WSSV infection. FISH & SHELLFISH IMMUNOLOGY 2018; 82:220-228. [PMID: 30125702 DOI: 10.1016/j.fsi.2018.08.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 06/08/2023]
Abstract
GSK3β, a serine/threonine protein kinase, is a crucial regulator in several signaling pathway and plays a vital role in multiple cellular processes including cell proliferation, growth, apoptosis and immune response. In this study, a GSK3β homolog from L. vannamei, designed as LvGSK3β, was characterized. Sequence analysis showed that LvGSK3β possessed a highly similarity with GSK3β from other species, which contained a catalytic domain and serine/threonine phosphorylation sites. To analyze the role of LvGSK3β in the process of white spot syndrome virus (WSSV) infection, real-time quantitative PCR and western blot assays were performed. The results showed that the transcription and expression levels of LvGSK3β were inhibited upon WSSV challenge, accompanied with down-regulated phosphorylation levels. When LvGSK3β was silenced, the transcription of WSSV gene ie1 was inhibited, and the apoptosis of hemocytes induced by WSSV was up-regulated remarkably as well. In addition, inactivation of LvGSK3β could also depress virus infection that further validated the results. Conclusively, LvGSK3β was an important protein for shrimp immunomodulation, and shrimp might promote the apoptosis to restrain WSSV infection by inhibition of LvGSK3β. The study will be helpful for understanding the molecular mechanism of host-virus interaction.
Collapse
Affiliation(s)
- Lingwei Ruan
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of State Oceanic Administration, Third Institute of Oceanography, State Oceanic Administration, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Xiamen 361005, People's Republic of China.
| | - Huachun Liu
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of State Oceanic Administration, Third Institute of Oceanography, State Oceanic Administration, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Xiamen 361005, People's Republic of China; School of Life Science, Xiamen University, Xiamen 361005, People's Republic of China
| | - Hong Shi
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of State Oceanic Administration, Third Institute of Oceanography, State Oceanic Administration, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Xiamen 361005, People's Republic of China
| |
Collapse
|
42
|
Sengupta S, Katz SC, Sengupta S, Sampath P. Glycogen synthase kinase 3 inhibition lowers PD-1 expression, promotes long-term survival and memory generation in antigen-specific CAR-T cells. Cancer Lett 2018; 433:131-139. [PMID: 29959057 DOI: 10.1016/j.canlet.2018.06.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/07/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022]
Abstract
Successful remission in hematological cancers by CAR-T cell immunotherapy has yet to be replicated in solid tumors like GBM. A significant impediment of CAR-T immunotherapy in solid tumors is poor exposure of T cells to tumor antigens resulting in suboptimal CAR-T cell activation, which ultimately fails to induce a robust anti-tumor immune response. Costimulatory moieties in advanced-generation CARs, along with additional IL2 therapy has been shown to be insufficient to overcome this hurdle and have its cytotoxic limitations. GSK3 is constitutively active in naïve T cells and is transiently inactivated during T cell activation resulting in rapid T cell proliferation. Pharmacologic inhibition of GSK3 in GBM-specific CAR-T cells reduced FasL expression, increased T cell proliferation and reduced exhaustion by lowering PD-1 levels resulting in the development of CAR-T effector memory phenotype. Treatment with GSK3-inhibited CAR-T cells resulted in 100% tumor elimination during the tumor-rechallenge experiment in GBM-bearing animals and increased accumulation of memory CAR-T cells in secondary lymphoid organs. These adjuvant-like effects of GSK3 inhibition on activated CAR-T cells may be a valuable adjunct to a successful implementation of CAR-T immunotherapy against GBM and other solid tumors.
Collapse
Affiliation(s)
- Sadhak Sengupta
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, RI, USA; Department of Neurosurgery, Alpert School of Medicine, Brown University, Providence, RI, USA.
| | - Steven C Katz
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA; Department of Surgery, Boston University School of Medicine, Boston, MA, USA
| | | | - Prakash Sampath
- Brain Tumor Laboratory, Roger Williams Medical Center, Providence, RI, USA; Department of Neurosurgery, Alpert School of Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
43
|
Patterson AR, Endale M, Lampe K, Aksoylar HI, Flagg A, Woodgett JR, Hildeman D, Jordan MB, Singh H, Kucuk Z, Bleesing J, Hoebe K. Gimap5-dependent inactivation of GSK3β is required for CD4 + T cell homeostasis and prevention of immune pathology. Nat Commun 2018; 9:430. [PMID: 29382851 PMCID: PMC5789891 DOI: 10.1038/s41467-018-02897-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 01/08/2018] [Indexed: 12/16/2022] Open
Abstract
GTPase of immunity-associated protein 5 (Gimap5) is linked with lymphocyte survival, autoimmunity, and colitis, but its mechanisms of action are unclear. Here, we show that Gimap5 is essential for the inactivation of glycogen synthase kinase-3β (GSK3β) following T cell activation. In the absence of Gimap5, constitutive GSK3β activity constrains c-Myc induction and NFATc1 nuclear import, thereby limiting productive CD4+ T cell proliferation. Additionally, Gimap5 facilitates Ser389 phosphorylation and nuclear translocation of GSK3β, thereby limiting DNA damage in CD4+ T cells. Importantly, pharmacological inhibition and genetic targeting of GSK3β can override Gimap5 deficiency in CD4+ T cells and ameliorates immunopathology in mice. Finally, we show that a human patient with a GIMAP5 loss-of-function mutation has lymphopenia and impaired T cell proliferation in vitro that can be rescued with GSK3 inhibitors. Given that the expression of Gimap5 is lymphocyte-restricted, we propose that its control of GSK3β is an important checkpoint in lymphocyte proliferation. Loss of function GIMAP5 mutation is associated with lymphopenia, but how it mediates T cell homeostasis is unclear. Here the authors study Gimap5−/− mice and a patient with GIMAP5 deficiency to show how this GTPAse negatively regulates GSK3β activity to prevent DNA damage and cell death.
Collapse
Affiliation(s)
- Andrew R Patterson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, 231 Albert Sabin Way # E251n, Cincinnati, OH, 45267, USA
| | - Mehari Endale
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Kristin Lampe
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Halil I Aksoylar
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Aron Flagg
- Pediatric Hematology/Oncology and Blood & Marrow Transplant, Cleveland Clinic Children's, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Jim R Woodgett
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - David Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, 231 Albert Sabin Way # E251n, Cincinnati, OH, 45267, USA
| | - Michael B Jordan
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, 231 Albert Sabin Way # E251n, Cincinnati, OH, 45267, USA
| | - Harinder Singh
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, 231 Albert Sabin Way # E251n, Cincinnati, OH, 45267, USA
| | - Zeynep Kucuk
- Division of Bone Marrow Transplantation & Immune Deficiency, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Jack Bleesing
- Division of Bone Marrow Transplantation & Immune Deficiency, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Kasper Hoebe
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA. .,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, 231 Albert Sabin Way # E251n, Cincinnati, OH, 45267, USA. .,Department of Pediatrics, University of Cincinnati, College of Medicine, 3230 Eden Avenue, Cincinnati, OH, 45267, USA.
| |
Collapse
|
44
|
Zhang JY, Zhao YL, Lv YP, Cheng P, Chen W, Duan M, Teng YS, Wang TT, Peng LS, Mao FY, Liu YG, Fu XL, Yu PW, Luo P, Zhang WJ, Zou QM, Zhuang Y. Modulation of CD8 + memory stem T cell activity and glycogen synthase kinase 3β inhibition enhances anti-tumoral immunity in gastric cancer. Oncoimmunology 2018; 7:e1412900. [PMID: 29632726 PMCID: PMC5889281 DOI: 10.1080/2162402x.2017.1412900] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 11/23/2017] [Accepted: 11/28/2017] [Indexed: 01/09/2023] Open
Abstract
The potential contributions of CD8+ memory stem T cells to anti-tumor immunity and immunotherapy responses in gastric cancer has not been demonstrated. We found that CD8+ memory stem T cell frequencies were increased in the peripheral blood of gastric cancer patients compared to healthy donors and declined in frequency with disease progression. Despite minimal in vitro cytotoxic activity, the adoptive transfer of CD8+ memory stem T cells into Rag1-/- tumor bearing mice enhanced tumor regression compared to CD8+ central or effector memory T cell counterparts. This effect was associated with an increase in splenic, draining lymph node and tumor infiltrating CD8+ T cell numbers and the development of an altered CD8+ T cell phenotype not seen during homeostasis. GSK-3β inhibition is known to promote memory stem T cell accumulation by arresting effector T cell differentiation in vivo. Surprisingly however, GSK-3β inhibition conversely increased the cytotoxic capacity of CD8+ memory stem T cells in vitro, and this was associated with the induction of effector T cell-associated effector proteins including FasL. Finally, FasL neutralization following GSK-3β inhibition directly attenuated the anti-tumoral capacity of CD8+ memory stem T cells both in vitro and in vivo. Altogether, our findings identify the therapeutic potential of modulating CD8+ memory stem T cells for improved anti-tumoral responses against gastric cancer.
Collapse
Affiliation(s)
- Jin-Yu Zhang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yong-Liang Zhao
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yi-Pin Lv
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ping Cheng
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Weisan Chen
- School of Molecular Science, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Mubin Duan
- School of Molecular Science, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Yong-Sheng Teng
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ting-Ting Wang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Liu-Sheng Peng
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Fang-Yuan Mao
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yu-Gang Liu
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiao-Long Fu
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Pei-Wu Yu
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ping Luo
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wei-Jun Zhang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Quan-Ming Zou
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yuan Zhuang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
45
|
Tran CW, Saibil SD, Le Bihan T, Hamilton SR, Lang KS, You H, Lin AE, Garza KM, Elford AR, Tai K, Parsons ME, Wigmore K, Vainberg MG, Penninger JM, Woodgett JR, Mak TW, Ohashi PS. Glycogen Synthase Kinase-3 Modulates Cbl-b and Constrains T Cell Activation. THE JOURNAL OF IMMUNOLOGY 2017; 199:4056-4065. [DOI: 10.4049/jimmunol.1600396] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 10/05/2017] [Indexed: 11/19/2022]
|
46
|
Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, Chhabra S, Huang W, Liu H, Aref AR, Ivanova E, Paweletz CP, Bowden M, Zhou CW, Herter-Sprie GS, Sorrentino JA, Bisi JE, Lizotte PH, Merlino AA, Quinn MM, Bufe LE, Yang A, Zhang Y, Zhang H, Gao P, Chen T, Cavanaugh ME, Rode AJ, Haines E, Roberts PJ, Strum JC, Richards WG, Lorch JH, Parangi S, Gunda V, Boland GM, Bueno R, Palakurthi S, Freeman GJ, Ritz J, Haining WN, Sharpless NE, Arthanari H, Shapiro GI, Barbie DA, Gray NS, Wong KK. CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation. Cancer Discov 2017; 8:216-233. [PMID: 29101163 DOI: 10.1158/2159-8290.cd-17-0915] [Citation(s) in RCA: 525] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/24/2017] [Accepted: 10/31/2017] [Indexed: 12/26/2022]
Abstract
Immune checkpoint blockade, exemplified by antibodies targeting the PD-1 receptor, can induce durable tumor regressions in some patients. To enhance the efficacy of existing immunotherapies, we screened for small molecules capable of increasing the activity of T cells suppressed by PD-1. Here, we show that short-term exposure to small-molecule inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6) significantly enhances T-cell activation, contributing to antitumor effects in vivo, due in part to the derepression of NFAT family proteins and their target genes, critical regulators of T-cell function. Although CDK4/6 inhibitors decrease T-cell proliferation, they increase tumor infiltration and activation of effector T cells. Moreover, CDK4/6 inhibition augments the response to PD-1 blockade in a novel ex vivo organotypic tumor spheroid culture system and in multiple in vivo murine syngeneic models, thereby providing a rationale for combining CDK4/6 inhibitors and immunotherapies.Significance: Our results define previously unrecognized immunomodulatory functions of CDK4/6 and suggest that combining CDK4/6 inhibitors with immune checkpoint blockade may increase treatment efficacy in patients. Furthermore, our study highlights the critical importance of identifying complementary strategies to improve the efficacy of immunotherapy for patients with cancer. Cancer Discov; 8(2); 216-33. ©2017 AACR.See related commentary by Balko and Sosman, p. 143See related article by Jenkins et al., p. 196This article is highlighted in the In This Issue feature, p. 127.
Collapse
Affiliation(s)
- Jiehui Deng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Eric S Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Russell W Jenkins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Shuai Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ruben Dries
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kathleen Yates
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sandeep Chhabra
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Wei Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hongye Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amir R Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elena Ivanova
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Cloud P Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michaela Bowden
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Chensheng W Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Grit S Herter-Sprie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - John E Bisi
- G1 Therapeutics, Research Triangle Park, North Carolina
| | - Patrick H Lizotte
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ashley A Merlino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Max M Quinn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lauren E Bufe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Annan Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yanxi Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hua Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Peng Gao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ting Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Megan E Cavanaugh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amanda J Rode
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eric Haines
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jay C Strum
- G1 Therapeutics, Research Triangle Park, North Carolina
| | - William G Richards
- Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jochen H Lorch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Viswanath Gunda
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Genevieve M Boland
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raphael Bueno
- Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sangeetha Palakurthi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Norman E Sharpless
- The Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Haribabu Arthanari
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts.
| | - Kwok-Kin Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
47
|
Taylor A, Rothstein D, Rudd CE. Small-Molecule Inhibition of PD-1 Transcription Is an Effective Alternative to Antibody Blockade in Cancer Therapy. Cancer Res 2017; 78:706-717. [DOI: 10.1158/0008-5472.can-17-0491] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/22/2017] [Accepted: 10/17/2017] [Indexed: 11/16/2022]
|
48
|
Li X, Jia Z, Wang W, Wang L, Liu Z, Yang B, Jia Y, Song X, Yi Q, Qiu L, Song L. Glycogen synthase kinase-3 (GSK3) regulates TNF production and haemocyte phagocytosis in the immune response of Chinese mitten crab Eriocheir sinensis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 73:144-155. [PMID: 28363635 DOI: 10.1016/j.dci.2017.03.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 06/07/2023]
Abstract
Glycogen synthase kinase-3 (GSK3) is a serine/threonine protein kinase firstly identified as a regulator of glycogen synthesis. Recently, it has been proved to be a key regulator of the immune reaction. In the present study, a GSK3 homolog gene (designated as EsGSK3) was cloned from Chinese mitten crab, Eriocheir sinensis. The open reading frame (ORF) was 1824 bp, which encoded a predicted polypeptide of 607 amino acids. There was a conserved Serine/Threonine Kinase domain and a DNA binding domain found in EsGSK3. Phylogenetic analysis showed that EsGSK3 was firstly clustered with GSK3-β from oriental river prawn Macrobrachium nipponense in the invertebrate branch, while GSK3s from vertebrates formed the other distinct branch. EsGSK3 mRNA transcripts could be detected in all tested tissues of the crab including haepatopancreas, eyestalk, muscle, gonad, haemocytes and haematopoietic tissue with the highest expression level in haepatopancreas. And EsGSK3 protein was mostly detected in the cytoplasm of haemocyte by immunofluorescence analysis. The expression levels of EsGSK3 mRNA increased significantly at 6 h after Aeromonas hydrophila challenge (p < 0.05) in comparison with control group, and then gradually decreased to the initial level at 48 h (p > 0.05). The mRNA expression of lipopolysaccharide-induced tumor necrosis factor (TNF)-α factor (EsLITAF) was also induced by A. hydrophila challenge. However, the mRNA expression of EsLITAF and TNF-α production was significantly suppressed after EsGSK3 was blocked in vivo with specific inhibitor lithium, while the phagocytosis of crab haemocytes was significantly promoted. These results collectively demonstrated that EsGSK3 could regulate the innate immune responses of E. sinensis by promoting TNF-α production and inhibiting haemocyte phagocytosis.
Collapse
Affiliation(s)
- Xiaowei Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266200, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhihao Jia
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weilin Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China.
| | - Zhaoqun Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin Yang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunke Jia
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaorui Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Qilin Yi
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Linsheng Song
- Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266200, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
49
|
Hatem-Vaquero M, Griera M, Giermakowska W, Luengo A, Calleros L, Gonzalez Bosc LV, Rodríguez-Puyol D, Rodríguez-Puyol M, De Frutos S. Integrin linked kinase regulates the transcription of AQP2 by NFATC3. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:922-935. [PMID: 28736155 DOI: 10.1016/j.bbagrm.2017.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/01/2022]
Abstract
Two processes are associated with progressive loss of renal function: 1) decreased aquaporin-2 (AQP2) expression and urinary concentrating capacity (Nephrogenic Diabetes Insipidus, NDI); and 2) changes in extracellular matrix (ECM) composition, e.g. increased collagen I (Col I) deposition, characteristic of tubule-interstitial fibrosis. AQP2 expression is regulated by both the ECM-to-intracellular scaffold protein integrin-linked kinase (ILK) by NFATc/AP1 and other transcription factors. In the present work, we used in vivo and in vitro approaches to examine ILK participation in NFATc3/AP-1-mediated increases in AQP2 gene expression. Both NFATc3 knock-out mice and ILK conditional-knockdown mice (cKD-ILK) display symptoms of NDI (polyuria and reduced AQP2 expression). NFATc3 is upregulated in the renal medulla tubular cells of cKD-ILK mice but with reduced nuclear localization. Inner medullary collecting duct mIMCD3 cells were subjected to ILK depletion and transfected with reporter plasmids. Pharmacological activators or inhibitors determined the effect of ILK activity on NFATc/AP-1-dependent increases in transcription of AQP2. Finally, mIMCD3 cultured on Col I showed reduced activity of the ILK/GSK3β/NFATc/AQP2 axis, suggesting this pathway is a potential target for therapeutic treatment of NDI.
Collapse
Affiliation(s)
- Marco Hatem-Vaquero
- Department of Systems Biology, Physiology Unit, Faculty of Medicine, University of Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain.
| | - Mercedes Griera
- Department of Systems Biology, Physiology Unit, Faculty of Medicine, University of Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain.
| | - Wieslawa Giermakowska
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| | - Alicia Luengo
- Department of Systems Biology, Physiology Unit, Faculty of Medicine, University of Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain.
| | - Laura Calleros
- Department of Systems Biology, Physiology Unit, Faculty of Medicine, University of Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain.
| | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| | - Diego Rodríguez-Puyol
- Instituto Reina Sofia de Investigación Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain; Biomedical Research Foundation and Nephrology Department, Hospital Príncipe de Asturias, Alcalá de Henares, Madrid, Spain.
| | - Manuel Rodríguez-Puyol
- Department of Systems Biology, Physiology Unit, Faculty of Medicine, University of Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain.
| | - Sergio De Frutos
- Department of Systems Biology, Physiology Unit, Faculty of Medicine, University of Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
50
|
Taylor A, Harker JA, Chanthong K, Stevenson PG, Zuniga EI, Rudd CE. Glycogen Synthase Kinase 3 Inactivation Drives T-bet-Mediated Downregulation of Co-receptor PD-1 to Enhance CD8(+) Cytolytic T Cell Responses. Immunity 2016; 44:274-86. [PMID: 26885856 PMCID: PMC4760122 DOI: 10.1016/j.immuni.2016.01.018] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 05/12/2015] [Accepted: 11/11/2015] [Indexed: 01/22/2023]
Abstract
Despite the importance of the co-receptor PD-1 in T cell immunity, the upstream signaling pathway that regulates PD-1 expression has not been defined. Glycogen synthase kinase 3 (GSK-3, isoforms α and β) is a serine-threonine kinase implicated in cellular processes. Here, we identified GSK-3 as a key upstream kinase that regulated PD-1 expression in CD8(+) T cells. GSK-3 siRNA downregulation, or inhibition by small molecules, blocked PD-1 expression, resulting in increased CD8(+) cytotoxic T lymphocyte (CTL) function. Mechanistically, GSK-3 inactivation increased Tbx21 transcription, promoting enhanced T-bet expression and subsequent suppression of Pdcd1 (encodes PD-1) transcription in CD8(+) CTLs. Injection of GSK-3 inhibitors in mice increased in vivo CD8(+) OT-I CTL function and the clearance of murine gamma-herpesvirus 68 and lymphocytic choriomeningitis clone 13 and reversed T cell exhaustion. Our findings identify GSK-3 as a regulator of PD-1 expression and demonstrate the applicability of GSK-3 inhibitors in the modulation of PD-1 in immunotherapy.
Collapse
Affiliation(s)
- Alison Taylor
- Cell Signalling Section, Division of Immunology, Department of Pathology, Tennis Court Road, University of Cambridge, Cambridge CB2 1QP, UK
| | - James A Harker
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Kittiphat Chanthong
- Cell Signalling Section, Division of Immunology, Department of Pathology, Tennis Court Road, University of Cambridge, Cambridge CB2 1QP, UK
| | - Philip G Stevenson
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge CB2 2QQ, UK
| | - Elina I Zuniga
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Christopher E Rudd
- Cell Signalling Section, Division of Immunology, Department of Pathology, Tennis Court Road, University of Cambridge, Cambridge CB2 1QP, UK.
| |
Collapse
|