1
|
Jin Y, Yuan H, Mehta I, Ezenwa O, Morel PA. Alternatively Spliced Variants of Murine CD247 Influence T Cell Development and Activation, Revealing the Importance of the CD3ζ C-Terminal Region. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:541-550. [PMID: 38117282 PMCID: PMC10872740 DOI: 10.4049/jimmunol.2300511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023]
Abstract
CD247, also known as CD3ζ, is a crucial signaling molecule that transduces signals delivered by TCR through its three ITAMs. CD3ζ is required for successful thymocyte development. Three additional alternatively spliced variants of murine CD247 have been described, that is, CD3ι, CD3θ, and CD3η, that differ from CD3ζ in the C terminus such that the third ITAM is lost. Previous studies demonstrated defects in T cell development in mice expressing CD3η, but the TCR signaling pathways affected by CD3η and the impacts of the CD3ι and CD3θ on T cell development were not explored. In this study, we used a retrovirus-mediated gene transfer technique to express these three isoforms individually and examined the roles of them on T cell development and activation. Rag1-/- mice reconstituted with CD3θ-expressing bone marrow failed to develop mature T cells. CD3ι-expressing T cells exhibited similar development and activation as cells expressing CD3ζ. In contrast, thymic development was severely impaired in CD3η-reconstituted mice. Single-positive but not double-positive CD3η-expressing thymocytes had reduced TCR expression, and CD5 expression was decreased at the double-positive stage, suggesting a defect in positive selection. Peripheral CD3η-expressing T cells had expanded CD44hi populations and upregulation of exhaustion markers seen by flow cytometry and RNA sequencing analysis. Analysis of early signaling events demonstrated significantly reduced activation of both the PLCγ1 and Akt/mTOR signaling pathways. There was also a reduction in the frequency of activation of CD3η-expressing T cells. These studies reveal the importance of the CD3ζ C-terminal region in T cell development and activation.
Collapse
Affiliation(s)
- Ye Jin
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Huijuan Yuan
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh PA
| | - Isha Mehta
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Ogechukwu Ezenwa
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Penelope A Morel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
2
|
Li Z, Dai R, Chen M, Huang L, Zhu K, Li M, Zhu W, Li Y, Xie N, Li J, Wang L, Lan F, Cao CM. p55γ degrades RIP3 via MG53 to suppress ischaemia-induced myocardial necroptosis and mediates cardioprotection of preconditioning. Cardiovasc Res 2023; 119:2421-2440. [PMID: 37527538 DOI: 10.1093/cvr/cvad123] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 05/04/2023] [Accepted: 06/13/2023] [Indexed: 08/03/2023] Open
Abstract
AIMS Regulated necrosis (necroptosis) and apoptosis are important biological features of myocardial infarction, ischaemia-reperfusion (I/R) injury, and heart failure. However, the molecular mechanisms underlying myocardial necroptosis remain elusive. Ischaemic preconditioning (IPC) is the most powerful intrinsic cardioprotection against myocardial I/R injury. In this study, we aimed to determine whether IPC suppresses I/R-induced necroptosis and the underlying molecular mechanisms. METHODS AND RESULTS We generated p55γ transgenic and knockout mice and used ligation of left anterior descending coronary artery to produce an in vivo I/R model. The effects of p55γ and its downstream molecules were subsequently identified using mass spectroscopy and co-immunoprecipitation and pulldown assays. We found that p55γ expression was down-regulated in failing human myocardium caused by coronary heart disease as well as in I/R mouse hearts. Cardiac-specific p55γ overexpression ameliorated the I/R-induced necroptosis. In striking contrast, p55γ deficiency (p55γ-/-) and cardiac-specific deletion of p55γ (p55γc-KO) worsened I/R-induced injury. IPC up-regulated p55γ expression in vitro and in vivo. Using reporter and chromatin immunoprecipitation assays, we found that Hif1α transcriptionally regulated p55γ expression and mediated the cardioprotection of IPC. IPC-mediated suppression of necroptosis was attenuated in p55γ-/- and p55γc-KO hearts. Mechanistically, p55γ overexpression decreased the protein levels of RIP3 rather than the mRNA levels, while p55γ deficiency increased the protein abundance of RIP3. IPC attenuated the I/R-induced up-regulation of RIP3, which was abolished in p55γ-deficient mice. Up-regulation of RIP3 attenuated the p55γ- or IPC-induced inhibition of necroptosis in vivo. Importantly, p55γ directly bound and degraded RIP3 in a ubiquitin-dependent manner. We identified MG53 as the E3 ligase that mediated the p55γ-induced degradation of RIP3. In addition, we also found that p55γ activated the RISK pathway during IPC. CONCLUSIONS Our findings reveal that activation of the MG53-RIP3 signal pathway by p55γ protects the heart against I/R-induced necroptosis and underlies IPC-induced cardioprotection.
Collapse
Affiliation(s)
- Zhenyan Li
- Laboratory of Cardiovascular Science, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing 100050, China
- Department of Physiology, Capital Institute of Pediatrics, 2 Yabao Road, Chaoyang District, Beijing 100020, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 9 Dongdansantiao, Dongcheng District, Beijing 100730, China
| | - Rilei Dai
- Laboratory of Cardiovascular Science, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing 100050, China
| | - Min Chen
- Department of Physiology, Capital Institute of Pediatrics, 2 Yabao Road, Chaoyang District, Beijing 100020, China
| | - Lixuan Huang
- Department of Dermatology, The Fourth Hospital of Hebei Medical University, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Kun Zhu
- Laboratory of Cardiovascular Science, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing 100050, China
| | - Mingyang Li
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing 100050, China
| | - Wenting Zhu
- Laboratory of Cardiovascular Science, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing 100050, China
| | - Yang Li
- Laboratory of Cardiovascular Science, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing 100050, China
| | - Ning Xie
- Institute of Molecular Medicine, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| | - Jingchen Li
- Laboratory of Cardiovascular Science, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing 100050, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing 100037, China
| | - Feng Lan
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing 100037, China
| | - Chun-Mei Cao
- Laboratory of Cardiovascular Science, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing 100050, China
- Department of Physiology, Capital Institute of Pediatrics, 2 Yabao Road, Chaoyang District, Beijing 100020, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 9 Dongdansantiao, Dongcheng District, Beijing 100730, China
| |
Collapse
|
3
|
Zhong Y, Wang B, Chen W, Zhang H, Sun J, Dong J. Exploring the Mechanisms of Modified Bu-Shen-Yi-Qi Decoction for COPD-Related Osteoporosis Therapy via Transcriptomics and Network Pharmacology Approach. Drug Des Devel Ther 2023; 17:2727-2745. [PMID: 37701046 PMCID: PMC10493229 DOI: 10.2147/dddt.s413532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/11/2023] [Indexed: 09/14/2023] Open
Abstract
Purpose To investigate the effectiveness of modified Bu-Shen-Yi-Qi decoction (MBSYQ) in the treatment of osteoporosis associated with chronic obstructive pulmonary disease (COPD) and its underlying mechanisms of action. Methods Disease targets, active ingredients and targets were predicted by TTD, CTD, DisGeNET, HERB (BenCaoZuJian as its Chinese name), and multiple-TCM databases; In addition, the screened targets were performed via the online platforms DAVID 6.8 and Metascape for GO and KEGG pathway enrichment analysis; The relationship between the MBSYQ and core targets were verified by molecular docking technique. Then we established a COPD-associated osteoporosis rat model by passive 24-week cigarette exposure. We assessed the efficacy of MBSYQ by lung histopathology assessment and distal femur/the first lumbar vertebra (L1) microstructural assay. In addition, we performed tibial RNA sequencing, which was validated by RT-PCR and Western blot. Results Screening revealed that the 350 active compounds of MBSYQ anchored 228 therapeutic targets for COPD-related osteoporosis; KEGG pathway enrichment analysis showed that the key targets mainly regulated MAPK and PI3K/AKT signaling pathways. In vivo studies showed that MBSYQ treatment alleviated pathological alterations in lung tissue, and reversed the bone loss and microstructure damage in the femur/L1 of model rats. The RNA seq indicated that MBSYQ could upregulate genes associated with anti-oxidative stress and aerobic respiration. The GSEA analysis displayed that MAPK and PI3K/AKT pathways were inhibited by CS exposure and activated by MBSYQ. Conclusion MBSYQ is effective in the prevention and treatment of COPD-related osteoporosis, partially achieved by improving oxygen metabolism and activating MAPK and PI3K/AKT pathways.
Collapse
Affiliation(s)
- Yuanyuan Zhong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
- Institute of Integrated Traditional Chinese and Western Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Bin Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
- Institute of Integrated Traditional Chinese and Western Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Wenjing Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
- Institute of Integrated Traditional Chinese and Western Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Hongying Zhang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
- Institute of Integrated Traditional Chinese and Western Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
- Institute of Integrated Traditional Chinese and Western Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
- Institute of Integrated Traditional Chinese and Western Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
4
|
Tserunyan V, Finley SD. A systems and computational biology perspective on advancing CAR therapy. Semin Cancer Biol 2023; 94:34-49. [PMID: 37263529 PMCID: PMC10529846 DOI: 10.1016/j.semcancer.2023.05.009] [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: 10/11/2022] [Revised: 04/24/2023] [Accepted: 05/28/2023] [Indexed: 06/03/2023]
Abstract
In the recent decades, chimeric antigen receptor (CAR) therapy signaled a new revolutionary approach to cancer treatment. This method seeks to engineer immune cells expressing an artificially designed receptor, which would endue those cells with the ability to recognize and eliminate tumor cells. While some CAR therapies received FDA approval and others are subject to clinical trials, many aspects of their workings remain elusive. Techniques of systems and computational biology have been frequently employed to explain the operating principles of CAR therapy and suggest further design improvements. In this review, we sought to provide a comprehensive account of those efforts. Specifically, we discuss various computational models of CAR therapy ranging in scale from organismal to molecular. Then, we describe the molecular and functional properties of costimulatory domains frequently incorporated in CAR structure. Finally, we describe the signaling cascades by which those costimulatory domains elicit cellular response against the target. We hope that this comprehensive summary of computational and experimental studies will further motivate the use of systems approaches in advancing CAR therapy.
Collapse
Affiliation(s)
- Vardges Tserunyan
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Stacey D Finley
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA; Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Sunaoshi M, Blyth BJ, Shang Y, Tsuruoka C, Morioka T, Shinagawa M, Ogawa M, Shimada Y, Tachibana A, Iizuka D, Kakinuma S. Post-Irradiation Thymic Regeneration in B6C3F1 Mice Is Age Dependent and Modulated by Activation of the PI3K-AKT-mTOR Pathway. BIOLOGY 2022; 11:biology11030449. [PMID: 35336821 PMCID: PMC8945464 DOI: 10.3390/biology11030449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/23/2022]
Abstract
Simple Summary Because children have a long life expectancy relative to adults and their tissues and organs are growing and developing rapidly, the risk of radiation carcinogenesis for children is considered higher than that for adults. However, the underlying mechanism(s) is unclear. To uncover the mechanism, we previously revealed that principal causative genes in mouse thymic lymphomas arising in irradiated infants or adults as Pten or Ikzf1, respectively, suggesting that cells with mutation in these genes might be the origin of lymphomas arising after irradiation depending on age at exposure. Here, we clarified the age-dependent differences in thymus-cell dynamics in mice during the initial post-irradiation period. Our results demonstrate that the dynamics of thymocytes during the post-irradiation period depends on the age at exposure. For irradiated infants in particular, the number of proliferating cells increase dramatically, and this correlate with activation of the PI3K-AKT-mTOR pathway. Thus, we conclude that the PI3K-AKT-mTOR pathway in infants contributed, at least in part, to thymus-cell dynamics through the modification of cell proliferation and survival after irradiation, which may be associated with the risk of Pten mutation-associated thymic lymphoma. Abstract The risk of radiation-induced carcinogenesis depends on age at exposure. We previously reported principal causative genes in lymphomas arising after infant or adult exposure to 4-fractionated irradiation as Pten or Ikzf1, respectively, suggesting that cells with mutation in these genes might be the origin of lymphomas arising after irradiation depending on age at exposure. Here, we clarified the age-dependent differences in thymus-cell dynamics in mice during the initial post-irradiation period. The thymocyte number initially decreased, followed by two regeneration phases. During the first regeneration, the proportion of phosphorylated-AKT-positive (p-AKT+) cells in cell-cycle phases S+G2/M of immature CD4−CD8− and CD4+CD8+ thymocytes and in phases G0/G1 of mature CD4+CD8− and CD4−CD8+ thymocytes was significantly greater in irradiated infants than in irradiated adults. During the second regeneration, the proportion of p-AKT+ thymocytes in phases G0/G1 increased in each of the three populations other than CD4−CD8− thymocytes more so than during the first regeneration. Finally, PI3K-AKT-mTOR signaling in infants contributed, at least in part, to biphasic thymic regeneration through the modification of cell proliferation and survival after irradiation, which may be associated with the risk of Pten mutation-associated thymic lymphoma.
Collapse
Affiliation(s)
- Masaaki Sunaoshi
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Benjamin J. Blyth
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Yi Shang
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Chizuru Tsuruoka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Takamitsu Morioka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Mayumi Shinagawa
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Mari Ogawa
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Yoshiya Shimada
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Akira Tachibana
- Graduate School of Science and Engineering, Ibaraki University, 2-1-1 Bunkyo, Mito 310-8512, Japan;
| | - Daisuke Iizuka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
- Correspondence: ; Tel.: +81-43-206-3160
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| |
Collapse
|
6
|
Metformin as a Potential Treatment Option for Endometriosis. Cancers (Basel) 2022; 14:cancers14030577. [PMID: 35158846 PMCID: PMC8833654 DOI: 10.3390/cancers14030577] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/07/2022] [Accepted: 01/21/2022] [Indexed: 02/07/2023] Open
Abstract
Endometriosis is a common disease in women of reproductive age, and its pathogenesis seems to be largely affected by hormone imbalance, inflammation, oxidative stress, and autophagy dysregulation. These pathophysiological disturbances interact with one another through mechanisms that are still awaiting elucidation. The aim of this article is to present current knowledge regarding the possibilities of using metformin in the pharmacological treatment of endometriosis. Metformin is an insulin sensitizer widely used for the treatment of type 2 diabetes mellitus. The pleiotropic effects of metformin are mainly exerted through the activation of AMP-activated protein kinase, which is the key cellular energy homeostasis regulator that inhibits mTOR, a major autophagy suppressor. Metformin regresses endometriotic implants by increasing the activity of superoxide dismutase. It is also an inhibitor of metalloproteinase-2, decreasing the levels of the vascular endothelial growth factor and matrix metalloproteinase-9 in animal studies. In endometriosis, metformin might modify the stroma-epithelium communication via Wnt2/β-catenin. With its unique therapeutic mechanisms and no serious side effects, metformin seems to be a helpful anti-inflammatory and anti-proliferative agent in the treatment of endometriosis. It could be a missing link for the successful treatment of this chronic disease.
Collapse
|
7
|
Vallée A, Vallée JN, Le Blanche A, Lecarpentier Y. PPARγ Agonists: Emergent Therapy in Endometriosis. Pharmaceuticals (Basel) 2021; 14:ph14060543. [PMID: 34204039 PMCID: PMC8229142 DOI: 10.3390/ph14060543] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/31/2021] [Accepted: 06/04/2021] [Indexed: 01/01/2023] Open
Abstract
Endometriosis is one of the major gynecological diseases of reproductive-age women. This disease is characterized by the presence of glands and stroma outside the uterine cavity. Several studies have shown the major role of inflammation, angiogenesis, adhesion and invasion, and apoptosis in endometriotic lesions. Nevertheless, the mechanisms underlying endometriotic mechanisms still remain unclear and therapies are not currently efficient. The introduction of new agents can be effective by improving the condition of patients. PPARγ ligands can directly modulate these pathways in endometriosis. However, data in humans remain low. Thus, the purpose of this review is to summarize the potential actions of PPARγ agonists in endometriosis by acting on inflammation, angiogenesis, invasion, adhesion, and apoptosis.
Collapse
Affiliation(s)
- Alexandre Vallée
- Department of Clinical Research and Innovation (DRCI), Foch Hospital, 92150 Suresnes, France
- Correspondence:
| | - Jean-Noël Vallée
- Centre Hospitalier Universitaire (CHU) Amiens Picardie, Université Picardie Jules Verne (UPJV), 80000 Amiens, France;
- DACTIM-Mis, Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, 86000 Poitiers, France
| | - Alain Le Blanche
- Laboratoire CeRSM (EA-2931), UPL, Université Paris Nanterre, F92000 Nanterre, France;
- Hôpital René-Dubos de Pontoise and Université de Versailles-Saint-Quentin, Simone Veil UFR des Sciences de la Santé, 78180 Montigny-le-Bretonneux, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien (GHEF), 77100 Meaux, France;
| |
Collapse
|
8
|
Ahmed LA, Mohamed AF, Abd El-Haleim EA, El-Tanbouly DM. Boosting Akt Pathway by Rupatadine Modulates Th17/Tregs Balance for Attenuation of Isoproterenol-Induced Heart Failure in Rats. Front Pharmacol 2021; 12:651150. [PMID: 33995066 PMCID: PMC8121023 DOI: 10.3389/fphar.2021.651150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/11/2021] [Indexed: 01/11/2023] Open
Abstract
Disruption of Th17/Tregs homeostasis plays a crucial role in governing the immune response during myocardial fibrosis and its progression to heart failure. The present study aimed to assess for the first time the possible protection afforded by rupatadine against isoproterenol-induced heart failure in rats. It also explored the role of PI3k/Akt as a possible mechanistic pathway, through which rupatadine could modulate Th17/Tregs balance to display its effect. Isoproterenol (85 and 170 mg/kg/day) was injected subcutaneously for 2 successive days, respectively and rupatadine (4 mg/kg/day) was then given orally for 14 days with or without wortmannin (PI3K/Akt inhibitor). Rupatadine succeeded to completely ameliorate isoproterenol-induced cardiac dysfunction as demonstrated by improvements of electrocardiographic and echocardiographic measurements. Moreover, rupatadine prevented the marked elevation of PAF and oxidative stress in addition to Th17 promoting cytokines (IL-6, IL-23, and TGF-β). Accordingly, rupatadine prevented Th17 stimulation or expansion as indicated by increased Foxp3/RORγt ratio and decreased production of its pro-inflammatory cytokine (IL-17). Rupatadine treatment mitigated isoproterenol-induced activation of STAT-3 signaling and the imbalance in p-Akt/total Akt ratio affording marked decrease in atrogin-1 and apoptotic biomarkers. Finally, this therapy was effective in averting cardiac troponin loss and reverting the histological alterations as assessed by myocardial fibrosis and hypertrophy grading. Contrariwise, co-administration of wortmannin mostly attenuated the protective effects of rupatadine affording more or less similar results to that of isoproterenol-untreated rats. In conclusion, rupatadine could be an effective therapy against the development of isoproterenol-induced heart failure where PI3K/Akt pathway seems to play a crucial role in its protective effect.
Collapse
Affiliation(s)
- Lamiaa A Ahmed
- Department of Pharmacology & Toxicology, Cairo University, Cairo, Egypt
| | - Ahmed F Mohamed
- Department of Pharmacology & Toxicology, Cairo University, Cairo, Egypt
| | | | | |
Collapse
|
9
|
Abstract
Akt kinases translate various external cues into intracellular signals that control cell survival, proliferation, metabolism and differentiation. This review discusses the requirement for Akt and its targets in determining the fate and function of T cells. We discuss the importance of Akt at various stages of T cell development including β-selection during which Akt fulfills the energy requirements of highly proliferative DN3 cells. Akt also plays an integral role in CD8 T cell biology where its regulation of Foxo transcription factors and mTORC1 metabolic activity controls effector versus memory CD8 T cell differentiation. Finally, Akt promotes the differentiation of naïve CD4 T cells into Th1, Th17 and Tfh cells but inhibits the development of Treg cells. We also highlight how modulating Akt in T cells is a promising avenue for enhancing cell-based cancer immunotherapy.
Collapse
|
10
|
Fan J, Das JK, Xiong X, Chen H, Song J. Development of CAR-T Cell Persistence in Adoptive Immunotherapy of Solid Tumors. Front Oncol 2021; 10:574860. [PMID: 33489881 PMCID: PMC7815927 DOI: 10.3389/fonc.2020.574860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 11/19/2020] [Indexed: 11/13/2022] Open
Abstract
Chimeric antigen receptor (CAR) T (CAR-T) cell transfer has made great success in hematological malignancies, but only shown a limited effect on solid tumors. One of the major hurdles is the poor persistence of infused cells derived from ex vivo activation/expansion and repeated antigen encounter after re-infusion. Bcl-xL has been demonstrated to play an important role on normal T cell survival and function as well as genetically engineered cells. In the current study, we developed a retroviral CAR construct containing a second-generation carcinoembryonic antigen (CEA)-targeting CAR with the Bcl-xL gene and tested the anti-CEA CAR-T cell immunotherapy for colorectal cancer. In vitro, the anti-CEA CAR-T cells destroyed CEA-expressing tumor cells and sustained survival. In vivo, adoptive cell transfer of anti-CEA CAR-T cells significantly enhanced the ability of the CAR-T cells to accumulate in tumor tissues, suppress tumor growth and increase the overall survival rate of tumor-bearing mice in a murine model of colorectal cancer. These results demonstrate a novel CAR-T platform that has the ability to increase the persistence of CAR-T cells in solid tumors through exogenous expression of persistent genes. The data provide a potentially novel approach to augment CAR-T immunotherapy for solid tumors.
Collapse
Affiliation(s)
- Jiaqiao Fan
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jugal Kishore Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Xiaofang Xiong
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| |
Collapse
|
11
|
Deleonardis A, Papale M. Methods to Study Posttranslational Modification Patterns in Cytotoxic T-Cells and Cancer. Methods Mol Biol 2021; 2325:137-153. [PMID: 34053056 DOI: 10.1007/978-1-0716-1507-2_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Protein posttranslational modifications (PTMs) regulate intracellular signaling associated with development and progression of many diseases; thus, they are key to understanding pathological mechanisms and set up more tailored therapies. In addition, many posttranslationally modified proteins are released into biological fluids and can be used as new and more specific biomarkers. Based on this evidence, we analyzed the role of some PTMs in cancer and described the correlation between specific PTMs and T-cells activation/inhibition in cancer microenvironment. In the second part of this chapter, we analyzed the most commonly used approaches for qualitative and quantitative determination of PTMs. The comparison of three distinct but often complementary methodologies such as immunoblotting, mass spectrometry, and ELISA assays has allowed to highlight the pros and cons of each approach with a focus on their current application and their future developments to obtain more confident biomarkers and therapeutic targets useful for diagnosis, prognosis, and monitoring of the response to therapy.
Collapse
Affiliation(s)
- Annamaria Deleonardis
- R&D Unit, Fluidia srl, Foggia, Italy
- Section of Nephrology, Dialysis and Transplantation, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Massimo Papale
- Clinical Pathology Unit, Department of Laboratory Diagnostics, Policlinic University Hospital "Riuniti", Foggia, Italy.
| |
Collapse
|
12
|
Ellman DG, Lund MC, Nissen M, Nielsen PS, Sørensen C, Lester EB, Thougaard E, Jørgensen LH, Nedospasov SA, Andersen DC, Stubbe J, Brambilla R, Degn M, Lambertsen KL. Conditional Ablation of Myeloid TNF Improves Functional Outcome and Decreases Lesion Size after Spinal Cord Injury in Mice. Cells 2020; 9:E2407. [PMID: 33153044 PMCID: PMC7692197 DOI: 10.3390/cells9112407] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/27/2020] [Accepted: 11/01/2020] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating condition consisting of an instant primary mechanical injury followed by a secondary injury that progresses for weeks to months. The cytokine tumor necrosis factor (TNF) plays an important role in the pathophysiology of SCI. We investigated the effect of myeloid TNF ablation (peripheral myeloid cells (macrophages and neutrophils) and microglia) versus central myeloid TNF ablation (microglia) in a SCI contusion model. We show that TNF ablation in macrophages and neutrophils leads to reduced lesion volume and improved functional outcome after SCI. In contrast, TNF ablation in microglia only or TNF deficiency in all cells had no effect. TNF levels tended to be decreased 3 h post-SCI in mice with peripheral myeloid TNF ablation and was significantly decreased 3 days after SCI. Leukocyte and microglia populations and all other cytokines (IL-1β, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, and IFNγ) and chemokines (CCL2, CCL5, and CXCL1) investigated, in addition to TNFR1 and TNFR2, were comparable between genotypes. Analysis of post-SCI signaling cascades demonstrated that the MAPK kinase SAPK/JNK decreased and neuronal Bcl-XL levels increased post-SCI in mice with ablation of TNF in peripheral myeloid cells. These findings demonstrate that peripheral myeloid cell-derived TNF is pathogenic in SCI.
Collapse
Affiliation(s)
- Ditte Gry Ellman
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (D.G.E.); (M.C.L.); (M.N.); (P.S.N.); (C.S.); (E.B.L.); (E.T.); (R.B.)
| | - Minna Christiansen Lund
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (D.G.E.); (M.C.L.); (M.N.); (P.S.N.); (C.S.); (E.B.L.); (E.T.); (R.B.)
| | - Maiken Nissen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (D.G.E.); (M.C.L.); (M.N.); (P.S.N.); (C.S.); (E.B.L.); (E.T.); (R.B.)
| | - Pernille Sveistrup Nielsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (D.G.E.); (M.C.L.); (M.N.); (P.S.N.); (C.S.); (E.B.L.); (E.T.); (R.B.)
| | - Charlotte Sørensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (D.G.E.); (M.C.L.); (M.N.); (P.S.N.); (C.S.); (E.B.L.); (E.T.); (R.B.)
| | - Emilie Boye Lester
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (D.G.E.); (M.C.L.); (M.N.); (P.S.N.); (C.S.); (E.B.L.); (E.T.); (R.B.)
| | - Estrid Thougaard
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (D.G.E.); (M.C.L.); (M.N.); (P.S.N.); (C.S.); (E.B.L.); (E.T.); (R.B.)
| | - Louise Helskov Jørgensen
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark; (L.H.J.); (D.C.A.)
| | - Sergei A. Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences and Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Ditte Caroline Andersen
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark; (L.H.J.); (D.C.A.)
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark
- Danish Center for Regenerative Medicine, Odense University Hospital, 5000 Odense, Denmark
| | - Jane Stubbe
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark;
| | - Roberta Brambilla
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (D.G.E.); (M.C.L.); (M.N.); (P.S.N.); (C.S.); (E.B.L.); (E.T.); (R.B.)
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Matilda Degn
- Pediatric Oncology Laboratory, Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (D.G.E.); (M.C.L.); (M.N.); (P.S.N.); (C.S.); (E.B.L.); (E.T.); (R.B.)
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark
- BRIGDE—Brain Research—Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
13
|
El Maï M, Marzullo M, de Castro IP, Ferreira MG. Opposing p53 and mTOR/AKT promote an in vivo switch from apoptosis to senescence upon telomere shortening in zebrafish. eLife 2020; 9:54935. [PMID: 32427102 PMCID: PMC7237213 DOI: 10.7554/elife.54935] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Progressive telomere shortening during lifespan is associated with restriction of cell proliferation, genome instability and aging. Apoptosis and senescence are the two major outcomes upon irreversible cellular damage. Here, we show a transition of these two cell fates during aging of telomerase deficient zebrafish. In young telomerase mutants, proliferative tissues exhibit DNA damage and p53-dependent apoptosis, but no senescence. However, these tissues in older animals display loss of cellularity and senescence becomes predominant. Tissue alterations are accompanied by a pro-proliferative stimulus mediated by AKT signaling. Upon AKT activation, FoxO transcription factors are phosphorylated and translocated out of the nucleus. This results in reduced SOD2 expression causing an increase of ROS and mitochondrial dysfunction. These alterations induce p15/16 growth arrest and senescence. We propose that, upon telomere shortening, early apoptosis leads to cell depletion and insufficient compensatory proliferation. Following tissue damage, the mTOR/AKT is activated causing mitochondrial dysfunction and p15/16-dependent senescence.
Collapse
Affiliation(s)
- Mounir El Maï
- Institute for Research on Cancer and Aging of Nice (IRCAN), Université Côte d'Azur, Nice, France.,Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | - Miguel Godinho Ferreira
- Institute for Research on Cancer and Aging of Nice (IRCAN), Université Côte d'Azur, Nice, France.,Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|
14
|
Vallée A, Lecarpentier Y. Curcumin and Endometriosis. Int J Mol Sci 2020; 21:E2440. [PMID: 32244563 PMCID: PMC7177778 DOI: 10.3390/ijms21072440] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 12/14/2022] Open
Abstract
Endometriosis is one of the main common gynecological disorders, which is characterized by the presence of glands and stroma outside the uterine cavity. Some findings have highlighted the main role of inflammation in endometriosis by acting on proliferation, apoptosis and angiogenesis. Oxidative stress, an imbalance between reactive oxygen species and antioxidants, could have a key role in the initiation and progression of endometriosis by resulting in inflammatory responses in the peritoneal cavity. Nevertheless, the mechanisms underlying this disease are still unclear and therapies are not currently efficient. Curcumin is a major anti-inflammatory agent. Several findings have highlighted the anti-oxidant, anti-inflammatory and anti-angiogenic properties of curcumin. The purpose of this review is to summarize the potential action of curcumin in endometriosis by acting on inflammation, oxidative stress, invasion and adhesion, apoptosis and angiogenesis.
Collapse
Affiliation(s)
- Alexandre Vallée
- Diagnosis and Therapeutic Center, Hypertension and Cardiovascular Prevention Unit, Hôtel-Dieu Hospital, AP-HP, Paris-Descartes University, 75004 Paris, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l’Est Francilien (GHEF), 77100 Meaux, France;
| |
Collapse
|
15
|
Deng J, Zhao S, Zhang X, Jia K, Wang H, Zhou C, He Y. OX40 (CD134) and OX40 ligand, important immune checkpoints in cancer. Onco Targets Ther 2019; 12:7347-7353. [PMID: 31564917 PMCID: PMC6735535 DOI: 10.2147/ott.s214211] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/30/2019] [Indexed: 12/26/2022] Open
Abstract
Immunotherapy has shown promising results in cancer treatment. Research shows that most patients might be resistant to these therapies. So, new immune therapies are needed. OX40 (CD134) and OX40 ligand (OX40L), costimulatory molecules, express on different types of immune cells. The interaction between OX40 and OX40L (OX40/OX40L) induces the expansion and proliferation of T cells and decreases the immunosuppression of regulatory T (Treg) cells to enhance the immune response to the specific antigen. For the important role OX40 takes in the process of immunity, many clinical trials are focusing on OX40 to find out whether it may have active effects in clinical cancer treatment. The results of clinical trials are still not enough. So, we reviewed the OX40 and its ligand (OX40L) function in cancer, clinical trials with OX40/OX40L and the correlation between OX40/OX40L and other immune checkpoints to add more ideas to tumor feasible treatment.
Collapse
Affiliation(s)
- Juan Deng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, People's Republic of China.,Medical School, Tongji University, Shanghai 200092, People's Republic of China
| | - Sha Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, People's Republic of China.,Medical School, Tongji University, Shanghai 200092, People's Republic of China
| | - Xiaoshen Zhang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, People's Republic of China.,Medical School, Tongji University, Shanghai 200092, People's Republic of China
| | - Keyi Jia
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, People's Republic of China.,Medical School, Tongji University, Shanghai 200092, People's Republic of China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, People's Republic of China.,Medical School, Tongji University, Shanghai 200092, People's Republic of China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, People's Republic of China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, People's Republic of China
| |
Collapse
|
16
|
Samimi M, Pourhanifeh MH, Mehdizadehkashi A, Eftekhar T, Asemi Z. The role of inflammation, oxidative stress, angiogenesis, and apoptosis in the pathophysiology of endometriosis: Basic science and new insights based on gene expression. J Cell Physiol 2019; 234:19384-19392. [PMID: 31004368 DOI: 10.1002/jcp.28666] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 01/24/2023]
Abstract
Endometriosis is a frequent and chronic illness in young women which could be defined by the existence of endometrial stroma and glands outside of the normal site of the lining of the uterus. It has painful symptoms. The advanced stage of endometriosis may lead to gynecological malignancies, such as ovarian cancer, and other complications, including infertility. However, its exact physiopathology is not well known. Recent studies have shown the possible roles of inflammation along with oxidative stress. Additionally, angiogenesis and apoptosis dysregulation contribute to endometriosis pathophysiology. Therapeutic strategies and continuing attempts, to conquer endometriosis should be done regarding molecular signaling pathways. Thus, the present review summarizes current studies and focuses on molecular mechanisms.
Collapse
Affiliation(s)
- Mansooreh Samimi
- Endometriosis Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Tahereh Eftekhar
- Reproductive Health Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
17
|
Guo C, Chen S, Liu W, Ma Y, Li J, Fisher PB, Fang X, Wang XY. Immunometabolism: A new target for improving cancer immunotherapy. Adv Cancer Res 2019; 143:195-253. [PMID: 31202359 DOI: 10.1016/bs.acr.2019.03.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fundamental metabolic pathways are essential for mammalian cells to provide energy, precursors for biosynthesis of macromolecules, and reducing power for redox regulation. While dysregulated metabolism (e.g., aerobic glycolysis also known as the Warburg effect) has long been recognized as a hallmark of cancer, recent discoveries of metabolic reprogramming in immune cells during their activation and differentiation have led to an emerging concept of "immunometabolism." Considering the recent success of cancer immunotherapy in the treatment of several cancer types, increasing research efforts are being made to elucidate alterations in metabolic profiles of cancer and immune cells during their interplays in the setting of cancer progression and immunotherapy. In this review, we summarize recent advances in studies of metabolic reprogramming in cancer as well as differentiation and functionality of various immune cells. In particular, we will elaborate how distinct metabolic pathways in the tumor microenvironment cause functional impairment of immune cells and contribute to immune evasion by cancer. Lastly, we highlight the potential of metabolically reprogramming the tumor microenvironment to promote effective and long-lasting antitumor immunity for improved immunotherapeutic outcomes.
Collapse
Affiliation(s)
- Chunqing Guo
- Department of Human & Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Shixian Chen
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenjie Liu
- Department of Human & Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Yibao Ma
- Department of Biochemistry & Molecular Biology, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Juan Li
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Paul B Fisher
- Department of Human & Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Xianjun Fang
- Department of Biochemistry & Molecular Biology, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Xiang-Yang Wang
- Department of Human & Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
18
|
Green barley mitigates cytotoxicity in human lymphocytes undergoing aggressive oxidative stress, via activation of both the Lyn/PI3K/Akt and MAPK/ERK pathways. Sci Rep 2019; 9:6005. [PMID: 30979953 PMCID: PMC6461650 DOI: 10.1038/s41598-019-42228-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 03/19/2019] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress plays a critical role in numerous diseases. Therefore, the pursuit of compounds with antioxidant activity remains critical. Green barley young leaves aqueous extract (GB) was tested for its capacity to ameliorate cellular oxidative stress, and its potential cytoprotective mechanism was partially elucidated. Through Folin-Ciocalteau and 1,1-diphenyl-2-picrylhydrazyl (DPPH) colorimetric assays, GB total phenolic content and free radical scavenging activity were found to be 59.91 ± 2.17 mg/L and 110.75 µg/ml (IC50), respectively. Using a live cell-based propidium iodide dye exclusion assay and flow cytometry, GB was found to display significant cytoprotection activity on three human lymphocytic cell lines exposed to an aggressive H2O2-induced oxidative stress. The molecular mechanism for GB cytoprotection activity was assessed via bead-based xMAP technology on the Luminex platform and western blot analysis. GB treatment resulted in activation of Lyn, Akt, and ERK1/2, suggesting that GB is able to mitigate the H2O2-induced oxidative stress via activation of both the Lyn/PI3K/Akt and ERK/MAPK pathways. Our findings support the notion that GB extract has the potential to be a valuable therapeutic agent and may serve to establish a strategy to discover potential compound(s) or biological extracts/mixtures to be incorporated as a treatment to prevent oxidative stress-related diseases.
Collapse
|
19
|
Zhang B, Dai Q, Jin X, Liang D, Li X, Lu H, Liu Y, Ding J, Gao Q, Wen Y. Phosphoinositide 3-kinase/protein kinase B inhibition restores regulatory T cell's function in pulmonary sarcoidosis. J Cell Physiol 2019; 234:19911-19920. [PMID: 30945303 DOI: 10.1002/jcp.28589] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/28/2022]
Abstract
Sarcoidosis is a systemic granulomatous disease associated with Th1/ regulatory T cells (Treg) paradigm. PI3K/Akt signaling, critical for maintaining Treg's homeostasis, is aberrantly activated in sarcoidosis patients. Here we tested the role of the PI3K inhibitors, LY294002 and BKM120, in immune modulation in experimental pulmonary sarcoidosis, concerning Th1/Th17/Treg immune profile detected by fluorescence-activated cell sorting analysis or quantitative polymerase chain reaction, as well as the effect on Treg's suppressive functions. Our investigation showed abnormal activation of PI3K/Akt signaling both in lung and Treg in pulmonary sarcoidosis, along with decreased frequency and damaged function of Treg. Blockage of PI3K suppressed this signaling in Treg, rebalanced Th1/Treg, inhibited the production of inflammatory cytokines, and enhanced Treg's function. These results demonstrate the key role of the PI3K/Akt signaling in regulating Th1/Th2 rebalances and indicates that PI3K/Akt signaling is critical for the optimal Treg responses in pulmonary sarcoidosis. Thus, PI3K inhibitors have potential for therapeutic translation, and can be candidate for add-on drugs to treat pulmonary sarcoidosis.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Qianqian Dai
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Xuguang Jin
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Dongmei Liang
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Xiaojie Li
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Haiyan Lu
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Yu Liu
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Jingjing Ding
- Department of Respiratory Medicine, Jiangsu Key Laboratory of Molecular Medicine, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qian Gao
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Yanting Wen
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| |
Collapse
|
20
|
Abstract
T-cell lymphomas (TCL) are a rare, heterogeneous group of non-Hodgkin lymphomas associated with very poor prognosis with standard cytotoxic chemotherapy. Epigenetic-based therapy, such as with histone deacetylase inhibitors, was initially discovered to be efficacious in TCL. In recent years, our understanding of the mechanisms driving T-cell lymphomagenesis has validated the use of epigenetic-based drugs and has also led to the development of novel agents with promising efficacy in pre-clinical and early clinical trials. These new treatments play upon the prominent existence of epigenetic and immune dysfunction present in T-cell lymphomas. With these advances, novel therapeutic regimens combining traditional chemotherapy as well as epigenetic and/or immunotherapy serve as promising future treatment options for TCL. In this review, we discuss the traditional methods of treatment for TCL as well as novel agents and combinations that will likely change the treatment paradigms resulting in better clinical outcomes.
Collapse
|
21
|
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]
|
22
|
Yang G, Yang X, Zhang J, Li G, Zheng D, Peng A, Hu J, Xu L, Yang B, Yang H, Zhou W, Tuzun E, Li J. Transcriptional repressor Blimp1 regulates follicular regulatory T-cell homeostasis and function. Immunology 2017; 153:105-117. [PMID: 28833081 DOI: 10.1111/imm.12815] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 07/18/2017] [Indexed: 01/23/2023] Open
Abstract
The B-lymphocyte-induced maturation protein 1 (Blimp1) regulates T-cell homeostasis and function. Loss of Blimp1 could double the proportion of follicular regulatory T (Tfr) cells. However, the effects that Blimp1 may have on the function of Tfr cells remain unknown. Here we document the function for Blimp1 in Tfr cells in vitro and in vivo. Data presented in this study demonstrate that Tfr cells indirectly inhibit the activation and differentiation of B cells by negatively regulating follicular helper T cells, so lowering the secretion of antibody. Lack of Blimp1 makes the immune suppression function of Tfr cells impaired in vitro. In the in vivo study, adoptive transfer of Tfr cells could reduce immune responses in germinal centres and relieve the muscle weakness symptoms of mice with experimental autoimmune myasthenia gravis. Blimp1 deficiency resulted in reduced suppressive ability of Tfr cells. This study identifies that Tfr cells are potent suppressors of immunity and are controlled by Blimp1.
Collapse
Affiliation(s)
- Guang Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaosu Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Junmei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Guancheng Li
- Cancer Research Institute, Central South University, Changsha, China
| | - Dandan Zheng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Anjiao Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jue Hu
- Department of Neurology, Changsha Central Hospital, Changsha, China
| | - Liqun Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Baifeng Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenbin Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Erdem Tuzun
- Department of Neurology, University of Istanbul, Istanbul, Turkey
| | - Jing Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
Shahdordizadeh M, Taghdisi SM, Sankian M, Ramezani M, Abnous K. Design, isolation and evaluation of the binding efficiency of a DNA aptamer against interleukin 2 receptor alpha, in vitro. Int Immunopharmacol 2017; 53:96-104. [PMID: 29055191 DOI: 10.1016/j.intimp.2017.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/07/2017] [Accepted: 10/10/2017] [Indexed: 01/19/2023]
Abstract
High levels of CD25, as part of the IL-2 receptor, are expressed on the surface of the activated T lymphocytes and regulatory T cells, indicating that the soluble CD25 (sCD25) could be a clinically valuable tool for treating several diseases. Moreover, progress has been achieved in targeting the IL-2 receptor to treat autoimmune diseases, organ transplantation and certain hematological malignancies. In the current study, generation of an ssDNA aptamer (Apt51) against CD25 is reported. Apt51 bound to CD25 with high affinity (Kd=13.4nM) and specificity. Furthermore, Apt51 was truncated to two shortened variants that almost retained their high affinity for the CD25 protein. Moreover, Apt51 showed good affinity and selectivity for the recognition of CD25 on the cell surface. Importantly, the study showed that Apt51 interfered with the binding of CD25 to its ligand (IL 2) and consequently decreased the IL-2-induced Akt activation.
Collapse
Affiliation(s)
- Mahin Shahdordizadeh
- Pharmaceutical Research Center, Advanced Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Advanced Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Khalil Abnous
- Pharmaceutical Research Center, Advanced Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Chaudhuri S, Singh MK, Bhattacharya D, Datta A, Hazra I, Mondal S, Faruk Sk Md O, Ronsard L, Ghosh TK, Chaudhuri S. T11TS immunotherapy repairs PI3K-AKT signaling in T-cells: Clues toward enhanced T-cell survival in rat glioma model. J Cell Physiol 2017; 233:759-770. [PMID: 28608562 DOI: 10.1002/jcp.26047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 06/12/2017] [Indexed: 01/20/2023]
Abstract
Malignant glioma is the most fatal of astrocytic lineage tumors despite therapeutic advances. Onset and progression of gliomas is accompanied by severe debilitation of T-cell defense and T-cell survival. One of the chief contributors to T-cell survival downstream of activation is the PI3K-AKT pathway. Our prior studies showed that the novel immunotherapeutic molecule T11-target structure (T11TS) blocks T-cell apoptosis in glioma. We also showed activation of immunological synapse components and calcineurin-NFAT pathway following T11TS immunotherapy of glioma-bearing rats. This lead to investigations whether such T-cell activation upon T11TS therapy translates into activation of downstream PI3K/AKT signals which may be related to observed blockade of T-cell apoptosis. For the purpose, we assessed by flowcytometry and immunoblotting, expressions of PI3K, PDK1, AKT, p-AKT, and PTEN in splenic T-cells of normal, experimentally-induced glioma-bearing rats and glioma-bearing rats receiving first, second and third doses of T11TS. We also determined comparative nuclear translocation of NF-κB across groups. We found significant increases in T-cell expressions of PDK1, PI3K, and p-AKT in T11TS-treated animal groups compared to sharp downregulations in glioma. AKT levels remained unchanged across groups. PTEN levels declined sharply after T11TS immunotherapy. T11TS also caused enhanced NF-κB translocation to the T-cell nucleus compared to glioma group. Results showed heightened activation of the PI3K-AKT pathway in glioma-bearing rats following T11TS immunotherapy. These results illustrate the novel role of T11TS immunotherapy in ameliorating the PI3K pathway in T-cells in glioma-bearing animals to enhance T-cell survival, according greater defense against glioma. The study thus has far-reaching clinical outcomes.
Collapse
Affiliation(s)
- Suhnrita Chaudhuri
- Department of Laboratory Medicine, Cellular and Molecular Immunology Lab, School of Tropical Medicine, Kolkata, West Bengal 700073, India.,Department of Physiology, University of Calcutta, Kolkata, West Bengal 700009, India
| | - Manoj K Singh
- Department of Laboratory Medicine, Cellular and Molecular Immunology Lab, School of Tropical Medicine, Kolkata, West Bengal 700073, India
| | - Debanjan Bhattacharya
- Department of Laboratory Medicine, Cellular and Molecular Immunology Lab, School of Tropical Medicine, Kolkata, West Bengal 700073, India
| | - Ankur Datta
- Department of Laboratory Medicine, Cellular and Molecular Immunology Lab, School of Tropical Medicine, Kolkata, West Bengal 700073, India
| | - Iman Hazra
- Department of Laboratory Medicine, Cellular and Molecular Immunology Lab, School of Tropical Medicine, Kolkata, West Bengal 700073, India
| | - Somnath Mondal
- Department of Laboratory Medicine, Cellular and Molecular Immunology Lab, School of Tropical Medicine, Kolkata, West Bengal 700073, India
| | - Omar Faruk Sk Md
- Department of Laboratory Medicine, Cellular and Molecular Immunology Lab, School of Tropical Medicine, Kolkata, West Bengal 700073, India
| | - Larance Ronsard
- Virology Lab, National Institute of Immunology, New Delhi 110067, India
| | - Tushar K Ghosh
- Department of Physiology, University of Calcutta, Kolkata, West Bengal 700009, India
| | - Swapna Chaudhuri
- Department of Laboratory Medicine, Cellular and Molecular Immunology Lab, School of Tropical Medicine, Kolkata, West Bengal 700073, India
| |
Collapse
|
25
|
Kunkl M, Porciello N, Mastrogiovanni M, Capuano C, Lucantoni F, Moretti C, Persson JL, Galandrini R, Buzzetti R, Tuosto L. ISA-2011B, a Phosphatidylinositol 4-Phosphate 5-Kinase α Inhibitor, Impairs CD28-Dependent Costimulatory and Pro-inflammatory Signals in Human T Lymphocytes. Front Immunol 2017; 8:502. [PMID: 28491063 PMCID: PMC5405084 DOI: 10.3389/fimmu.2017.00502] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/12/2017] [Indexed: 12/16/2022] Open
Abstract
Phosphatidylinositol 4,5-biphosphate (PIP2) is a membrane phospholipid that controls the activity of several proteins regulating cytoskeleton reorganization, cytokine gene expression, T cell survival, proliferation, and differentiation. Phosphatidylinositol 4-phosphate 5-kinases (PIP5Ks) are the main enzymes involved in PIP2 biosynthesis by phosphorylating phosphatidylinositol 4-monophosphate (PI4P) at the D5 position of the inositol ring. In human T lymphocytes, we recently found that CD28 costimulatory molecule is pivotal for PIP2 turnover by recruiting and activating PIP5Kα. We also found that PIP5Kα is the main regulator of both CD28 costimulatory signals integrating those delivered by TCR as well as CD28 autonomous signals regulating the expression of pro-inflammatory genes. Given emerging studies linking alterations of PIP2 metabolism to immune-based diseases, PIP5Kα may represent a promising target to modulate immunity and inflammation. Herewith, we characterized a recently discovered inhibitor of PIP5Kα, ISA-2011B, for its inhibitory effects on T lymphocyte functions. We found that the inhibition of PIP5Kα lipid-kinase activity by ISA-2011B significantly impaired CD28 costimulatory signals necessary for TCR-mediated Ca2+ influx, NF-AT transcriptional activity, and IL-2 gene expression as well as CD28 autonomous signals regulating the activation of NF-κB and the transcription of pro-inflammatory cytokine and chemokine genes. Moreover, our data on the inhibitory effects of ISA-2011B on CD28-mediated upregulation of inflammatory cytokines related to Th17 cell phenotype in type 1 diabetes patients suggest ISA-2011B as a promising anti-inflammatory drug.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Nicla Porciello
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Marta Mastrogiovanni
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Cristina Capuano
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | - Chiara Moretti
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Jenny L Persson
- Division of Experimental Cancer Research, Department of Laboratory Medicine, Clinical Research Center, Lund University, Malmö, Sweden.,Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | | | | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
26
|
Qiu Y, Zhang Y, Hu N, Dong F. A Truncated Granulocyte Colony-stimulating Factor Receptor (G-CSFR) Inhibits Apoptosis Induced by Neutrophil Elastase G185R Mutant: IMPLICATION FOR UNDERSTANDING CSF3R GENE MUTATIONS IN SEVERE CONGENITAL NEUTROPENIA. J Biol Chem 2017; 292:3496-3505. [PMID: 28073911 DOI: 10.1074/jbc.m116.755157] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 01/09/2017] [Indexed: 11/06/2022] Open
Abstract
Mutations in ELANE encoding neutrophil elastase (NE) have been identified in the majority of patients with severe congenital neutropenia (SCN). The NE mutants have been shown to activate unfolded protein response and induce premature apoptosis in myeloid cells. Patients with SCN are predisposed to acute myeloid leukemia (AML), and progression from SCN to AML is accompanied by mutations in CSF3R encoding the granulocyte colony-stimulating factor receptor (G-CSFR) in ∼80% of patients. The mutations result in the expression of C-terminally truncated G-CSFRs that promote strong cell proliferation and survival. It is unknown why the CSF3R mutations, which are rare in de novo AML, are so prevalent in SCN/AML. We show here that a G-CSFR mutant, d715, derived from an SCN patient inhibited G-CSF-induced expression of NE in a dominant negative manner. Furthermore, G-CSFR d715 suppressed unfolded protein response and apoptosis induced by an SCN-derived NE mutant, which was associated with sustained activation of AKT and STAT5, and augmented expression of BCL-XL. Thus, the truncated G-CSFRs associated with SCN/AML may protect myeloid precursor cells from apoptosis induced by the NE mutants. We propose that acquisition of CSF3R mutations may represent a mechanism by which myeloid precursor cells carrying the ELANE mutations evade the proapoptotic activity of the NE mutants in SCN patients.
Collapse
Affiliation(s)
- Yaling Qiu
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Yangyang Zhang
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Nan Hu
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Fan Dong
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606.
| |
Collapse
|
27
|
Chen S, Liu Y, Rong X, Li Y, Zhou J, Lu L. Neuroprotective Role of the PI3 Kinase/Akt Signaling Pathway in Zebrafish. Front Endocrinol (Lausanne) 2017; 8:21. [PMID: 28228749 PMCID: PMC5296330 DOI: 10.3389/fendo.2017.00021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/23/2017] [Indexed: 12/27/2022] Open
Abstract
Neuronal survival and growth in the embryo is controlled partly by trophic factors. For most trophic factors (such as Insulin-like growth factor-1), the ability to regulate cell survival has been attributed to the phosphoinositide 3-kinase (PI3K)/Akt kinase cascade. This study presents data illustrating the role of PI3K/Akt in attainment of normal brain size during zebrafish embryogenesis. Blocking PI3K with inhibitor LY294002 caused a significant reduction in brain size (in addition to global growth retardation) during zebrafish embryogenesis. This PI3 Kinase inhibition-induced brain size decrease was recovered by the overexpression of myristoylated Akt (myr-Akt), a constitutive form of Akt. Further analysis reveals that expressing exogenous myr-Akt significantly augmented brain size. Whole mount in situ hybridization analysis of several marker genes showed that myr-Akt overexpression did not alter brain patterning. Furthermore, the expression of myr-Akt was found to protect neuronal cells from apoptosis induced by heat shock and UV light, suggesting that inhibition of neuronal cell death may be part of the underlying cause of the increased brain size. These data provide a foundation for addressing the role of PI3K/Akt in brain growth during zebrafish embryogenesis.
Collapse
Affiliation(s)
- Shuang Chen
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Yunzhang Liu
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Xiaozhi Rong
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Yun Li
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Jianfeng Zhou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Ling Lu
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
- *Correspondence: Ling Lu,
| |
Collapse
|
28
|
Ding Y, Han R, Jiang W, Xiao J, Liu H, Chen X, Li X, Hao J. Programmed Death Ligand 1 Plays a Neuroprotective Role in Experimental Autoimmune Neuritis by Controlling Peripheral Nervous System Inflammation of Rats. THE JOURNAL OF IMMUNOLOGY 2016; 197:3831-3840. [PMID: 27798164 DOI: 10.4049/jimmunol.1601083] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/20/2016] [Indexed: 01/22/2023]
Abstract
Programmed death 1 (PD-1; CD279), a member of the CD28 family, is an inhibitory receptor on T cells and is responsible for T cell dysfunction in infectious diseases and cancers. The ligand for PD-1, programmed death ligand 1 (PD-L1; also known as B7-H1, CD274), is a member of the B7 family. The engagement of PD-1 with programmed death ligand can downregulate autoreactive T cells that participate in multiple autoimmune diseases. Experimental autoimmune neuritis (EAN) is an animal model of Guillain-Barré syndrome, and the pathogenesis of EAN is mediated principally through T cells and macrophages. In this study, we investigated the effects of PD-L1 in EAN rats. For preventative and therapeutic management, we administered PD-L1, which successfully decreased the severity of EAN; it alleviated the neurologic course of EAN, as well as inhibited the infiltration of inflammatory cells and demyelination of sciatic nerves. Our data revealed that PD-L1 treatment inhibited lymphocyte proliferation and altered T cell differentiation by inducing decreases in IFN-γ+CD4+ Th1 cells and IL-17+CD4+ Th17 cells and increases in IL-4+CD4+ Th2 cells and Foxp3+CD4+ regulatory T cells. The expression levels of p-STAT3 and Foxp3 were significantly different in PD-L1-treated groups compared with the control group. Additionally, PD-L1 regulated the expression of Foxp3 and p-STAT3 in EAN, probably by inhibiting PI3K/AKT/mTOR signaling expression. In summary, PD-L1 is a potentially useful agent for the treatment of EAN because of its anti-inflammatory and neuroprotective effects.
Collapse
Affiliation(s)
- Yanan Ding
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ranran Han
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wei Jiang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jinting Xiao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Haijie Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiuju Chen
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiaowen Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Junwei Hao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
29
|
Kitz A, de Marcken M, Gautron AS, Mitrovic M, Hafler DA, Dominguez-Villar M. AKT isoforms modulate Th1-like Treg generation and function in human autoimmune disease. EMBO Rep 2016; 17:1169-83. [PMID: 27312110 DOI: 10.15252/embr.201541905] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 05/13/2016] [Indexed: 12/22/2022] Open
Abstract
Foxp3(+) regulatory T cells (Tregs) exhibit plasticity, which dictates their function. Secretion of the inflammatory cytokine IFNγ, together with the acquisition of a T helper 1 (Th1)-like effector phenotype as observed in cancer, infection, and autoimmune diseases, is associated with loss of Treg suppressor function through an unknown mechanism. Here, we describe the signaling events driving the generation of human Th1-Tregs. Using a genome-wide gene expression approach and pathway analysis, we identify the PI3K/AKT/Foxo1/3 signaling cascade as the major pathway involved in IFNγ secretion by human Tregs. Furthermore, we describe the opposing roles of AKT isoforms in Th1-Treg generation ex vivo Finally, we employ multiple sclerosis as an in vivo model with increased but functionally defective Th1-Tregs. We show that the PI3K/AKT/Foxo1/3 pathway is activated in ex vivo-isolated Tregs from untreated relapsing-remitting MS patients and that blockade of the pathway inhibits IFNγ secretion and restores the immune suppressive function of Tregs. These data define a fundamental pathway regulating the function of human Tregs and suggest a novel treatment paradigm for autoimmune diseases.
Collapse
Affiliation(s)
- Alexandra Kitz
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Marine de Marcken
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Anne-Sophie Gautron
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Mitja Mitrovic
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - David A Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
30
|
Kim B, Kim J, Kim E, Lee J, Joo D, Huh K, Kim M, Kim Y. Role of Thalidomide on the Expression of OX40, 4-1BB, and GITR in T Cell Subsets. Transplant Proc 2016; 48:1270-4. [DOI: 10.1016/j.transproceed.2015.12.088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 12/30/2015] [Indexed: 11/25/2022]
|
31
|
Hyperactive mTOR pathway promotes lymphoproliferation and abnormal differentiation in autoimmune lymphoproliferative syndrome. Blood 2016; 128:227-38. [PMID: 27099149 DOI: 10.1182/blood-2015-11-685024] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/14/2016] [Indexed: 12/23/2022] Open
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a human disorder characterized by defective Fas signaling, resulting in chronic benign lymphoproliferation and accumulation of TCRαβ(+) CD4(-) CD8(-) double-negative T (DNT) cells. Although their phenotype resembles that of terminally differentiated or exhausted T cells, lack of KLRG1, high eomesodermin, and marginal T-bet expression point instead to a long-lived memory state with potent proliferative capacity. Here we show that despite their terminally differentiated phenotype, human ALPS DNT cells exhibit substantial mitotic activity in vivo. Notably, hyperproliferation of ALPS DNT cells is associated with increased basal and activation-induced phosphorylation of serine-threonine kinases Akt and mechanistic target of rapamycin (mTOR). The mTOR inhibitor rapamycin abrogated survival and proliferation of ALPS DNT cells, but not of CD4(+) or CD8(+) T cells in vitro. In vivo, mTOR inhibition reduced proliferation and abnormal differentiation by DNT cells. Importantly, increased mitotic activity and hyperactive mTOR signaling was also observed in recently defined CD4(+) or CD8(+) precursor DNT cells, and mTOR inhibition specifically reduced these cells in vivo, indicating abnormal programming of Fas-deficient T cells before the DNT stage. Thus, our results identify the mTOR pathway as a major regulator of lymphoproliferation and aberrant differentiation in ALPS.
Collapse
|
32
|
Porciello N, Tuosto L. CD28 costimulatory signals in T lymphocyte activation: Emerging functions beyond a qualitative and quantitative support to TCR signalling. Cytokine Growth Factor Rev 2016; 28:11-9. [PMID: 26970725 DOI: 10.1016/j.cytogfr.2016.02.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/22/2016] [Indexed: 01/22/2023]
Abstract
CD28 is one of the most important co-stimulatory receptors necessary for full T lymphocyte activation. By binding its cognate ligands, B7.1/CD80 or B7.2/CD86, expressed on the surface of professional antigen presenting cells (APC), CD28 initiates several signalling cascades, which qualitatively and quantitatively support T cell receptor (TCR) signalling. More recent data evidenced that human CD28 can also act as a TCR-independent signalling unit, by delivering specific signals, which regulate the expression of pro-inflammatory cytokine/chemokines. Despite the enormous progresses made in identifying the mechanisms and molecules involved in CD28 signalling properties, much remains to be elucidated, especially in the light of the functional differences observed between human and mouse CD28. In this review we provide an overview of the current mechanisms and molecules through which CD28 support TCR signalling and highlight recent findings on the specific signalling motifs that regulate the unique pro-inflammatory activity of human CD28.
Collapse
Affiliation(s)
- Nicla Porciello
- Istituto Pasteur-Fondazione Cenci Bolognetti, Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy
| | - Loretta Tuosto
- Istituto Pasteur-Fondazione Cenci Bolognetti, Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy.
| |
Collapse
|
33
|
Kim SJ, Park C, Lee JN, Lim H, Hong GY, Moon SK, Lim DJ, Choe SK, Park R. Erdosteine protects HEI-OC1 auditory cells from cisplatin toxicity through suppression of inflammatory cytokines and induction of Nrf2 target proteins. Toxicol Appl Pharmacol 2015; 288:192-202. [DOI: 10.1016/j.taap.2015.07.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/30/2015] [Accepted: 07/16/2015] [Indexed: 12/20/2022]
|
34
|
Xue G, Zippelius A, Wicki A, Mandala M, Tang F, Massi D, Hemmings BA. Integrated Akt/PKB Signaling in Immunomodulation and Its Potential Role in Cancer Immunotherapy. J Natl Cancer Inst 2015; 107:djv171. [DOI: 10.1093/jnci/djv171] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 05/22/2015] [Indexed: 12/17/2022] Open
|
35
|
Wang L, Zhang W, Ding Y, Xiu B, Li P, Dong Y, Zhu Q, Liang A. Up-regulation of VEGF and its receptor in refractory leukemia cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:5282-5290. [PMID: 26191229 PMCID: PMC4503100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/20/2015] [Indexed: 06/04/2023]
Abstract
OBJECTIVE To analyze the causative mechanisms in refractory leukemia cells. METHODS Vascular endothelial growth factor (VEGF) blood plasma concentrations in 35 de novo, 6 relapse, 20 remission leukemia patients and 10 healthy kids were determined via ELISA analyses. Transcription levels of the VEGF receptors (VEGFR) Fms-like tyrosine kinase-1 (Flt-1) and kinase-domain insert containing receptor (KDR) were determined in participants' leucocytes with RT-PCR. Apoptosis rates as well as Cyt-C and Caspase-3 expression was determined in Jurkat, Jurkat(Bcl-2), healthy and recurrent leukemia leukocytes with and without VP-16 applications via flow cytometry. Total Akt (t-Akt) expression and its phosphorylation (p-AKT) status in leukocytes of the participants were analyzed with western blots. RESULTS Healthy children and the remission group had the lowest blood plasma VEGF concentrations (91.16±41.34 vs. 135.80±111.28 pg/ml), followed by de novo leukemia patients (362.49±195.68 pg/ml-494.19±186.23 pg/ml) and relapse patients (574.37±278.45 pg/ml) (P<0.01). The same trend was statistically significant visible for Flt-1 and KDR expressions in leukocytes of the participants. Stable Bcl-2 overexpression led to reduced apoptosis rates as well as Cyt-C and Caspase-3 expressions in Jurkat cells after VP-16 application, which was similar in leucocytes of remission patients. In contrast to no phosphorylation in healthy children, Akt was phosphorylated in 10% remission samples, 30% de novo leukemia samples and in 67% of recurrent leukemia leucocytes. CONCLUSION High VEGF plus VEGFR expression and AKT phosphorylation are highest in leukocytes of remission patients, suggesting VEGF signaling as a cause of reduced apoptosis susceptibility upon treatments.
Collapse
Affiliation(s)
- Lei Wang
- Department of Hematology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine639 Zhizaoju Road, Shanghai 200011, China
| | - Wenjun Zhang
- Department of Hematology, Tongji Hospital of Tongji University389 Xincun Road, Shanghai 200065, China
| | - Yi Ding
- Department of Hematology, Tongji Hospital of Tongji University389 Xincun Road, Shanghai 200065, China
| | - Bing Xiu
- Department of Hematology, Tongji Hospital of Tongji University389 Xincun Road, Shanghai 200065, China
| | - Ping Li
- Department of Hematology, Tongji Hospital of Tongji University389 Xincun Road, Shanghai 200065, China
| | - Yan Dong
- Department of Hematology, Tongji Hospital of Tongji University389 Xincun Road, Shanghai 200065, China
| | - Qi Zhu
- Department of Hematology, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine639 Zhizaoju Road, Shanghai 200011, China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital of Tongji University389 Xincun Road, Shanghai 200065, China
| |
Collapse
|
36
|
Yu H, Littlewood T, Bennett M. Akt isoforms in vascular disease. Vascul Pharmacol 2015; 71:57-64. [PMID: 25929188 PMCID: PMC4728195 DOI: 10.1016/j.vph.2015.03.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/31/2015] [Indexed: 11/05/2022]
Abstract
The mammalian serine/threonine Akt kinases comprise three closely related isoforms: Akt1, Akt2 and Akt3. Akt activation has been implicated in both normal and disease processes, including in development and metabolism, as well as cancer and cardiovascular disease. Although Akt signalling has been identified as a promising therapeutic target in cancer, its role in cardiovascular disease is less clear. Importantly, accumulating evidence suggests that the three Akt isoforms exhibit distinct tissue expression profiles, localise to different subcellular compartments, and have unique modes of activation. Consistent with in vitro findings, genetic studies in mice show distinct effects of individual Akt isoforms on the pathophysiology of cardiovascular disease. This review summarises recent studies of individual Akt isoforms in atherosclerosis, vascular remodelling and aneurysm formation, to provide a comprehensive overview of Akt function in vascular disease.
Collapse
Affiliation(s)
- Haixiang Yu
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| | - Trevor Littlewood
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK
| | - Martin Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
37
|
Tegla CA, Cudrici CD, Nguyen V, Danoff J, Kruszewski AM, Boodhoo D, Mekala AP, Vlaicu SI, Chen C, Rus V, Badea TC, Rus H. RGC-32 is a novel regulator of the T-lymphocyte cell cycle. Exp Mol Pathol 2015; 98:328-37. [PMID: 25770350 DOI: 10.1016/j.yexmp.2015.03.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 03/09/2015] [Indexed: 10/23/2022]
Abstract
We have previously shown that RGC-32 is involved in cell cycle regulation in vitro. To define the in vivo role of RGC-32, we generated RGC-32 knockout mice. These mice developed normally and did not spontaneously develop overt tumors. To assess the effect of RGC-32 deficiency on cell cycle activation in T cells, we determined the proliferative rates of CD4(+) and CD8(+) T cells from the spleens of RGC-32(-/-) mice, as compared to wild-type (WT, RGC-32(+/+)) control mice. After stimulation with anti-CD3/anti-CD28, CD4(+) T cells from RGC-32(-/-) mice displayed a significant increase in [(3)H]-thymidine incorporation when compared to WT mice. In addition, both CD4(+) and CD8(+) T cells from RGC-32(-/-) mice displayed a significant increase in the proportion of proliferating Ki67(+) cells, indicating that in T cells, RGC-32 has an inhibitory effect on cell cycle activation induced by T-cell receptor/CD28 engagement. Furthermore, Akt and FOXO1 phosphorylation induced in stimulated CD4(+) T-cells from RGC-32(-/-) mice were significantly higher, indicating that RGC-32 inhibits cell cycle activation by suppressing FOXO1 activation. We also found that IL-2 mRNA and protein expression were significantly increased in RGC-32(-/-) CD4(+) T cells when compared to RGC-32(+/+) CD4(+) T cells. In addition, the effect of RGC-32 on the cell cycle and IL-2 expression was inhibited by pretreatment of the samples with LY294002, indicating a role for phosphatidylinositol 3-kinase (PI3K). Thus, RGC-32 is involved in controlling the cell cycle of T cells in vivo, and this effect is mediated by IL-2 in a PI3K-dependent fashion.
Collapse
Affiliation(s)
- Cosmin A Tegla
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA; Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, USA
| | - Cornelia D Cudrici
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Jacob Danoff
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Adam M Kruszewski
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Armugam P Mekala
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Sonia I Vlaicu
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA; Department of Internal Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ching Chen
- Department of Pathology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Tudor C Badea
- Retinal Circuit Development and Genetics Unit, N-NRL, National Eye Institute, Bethesda, MD, USA
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA; Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, USA; Veterans Administration Multiple Sclerosis Center of Excellence, Baltimore, MD, USA.
| |
Collapse
|
38
|
Oghumu S, Stock JC, Varikuti S, Dong R, Terrazas C, Edwards JA, Rappleye CA, Holovatyk A, Sharpe A, Satoskar AR. Transgenic expression of CXCR3 on T cells enhances susceptibility to cutaneous Leishmania major infection by inhibiting monocyte maturation and promoting a Th2 response. Infect Immun 2015; 83:67-76. [PMID: 25312956 PMCID: PMC4288897 DOI: 10.1128/iai.02540-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/03/2014] [Indexed: 12/25/2022] Open
Abstract
Cutaneous leishmaniasis, caused mainly by Leishmania major, an obligate intracellular parasite, is a disfiguring disease characterized by large skin lesions and is transmitted by a sand fly vector. We previously showed that the chemokine receptor CXCR3 plays a critical role in mediating resistance to cutaneous leishmaniasis caused by Leishmania major. Furthermore, T cells from L. major-susceptible BALB/c but not L. major-resistant C57BL/6 mice fail to efficiently upregulate CXCR3 upon activation. We therefore examined whether transgenic expression of CXCR3 on T cells would enhance resistance to L. major infection in susceptible BALB/c mice. We generated BALB/c and C57BL/6 transgenic mice, which constitutively overexpressed CXCR3 under a CD2 promoter, and then examined the outcomes with L. major infection. Contrary to our hypothesis, transgenic expression of CXCR3 (CXCR3(Tg)) on T cells of BALB/c mice resulted in increased lesion sizes and parasite burdens compared to wild-type (WT) littermates after L. major infection. Restimulated lymph node cells from L. major-infected BALB/c-CXCR3(Tg) mice produced more interleukin-4 (IL-4) and IL-10 and less gamma interferon (IFN-γ). Cells in draining lymph nodes from BALB/c-CXCR3(Tg) mice showed enhanced Th2 and reduced Th1 cell accumulation associated with increased neutrophils and inflammatory monocytes. However, monocytes displayed an immature phenotype which correlated with increased parasite burdens. Interestingly, transgenic expression of CXCR3 on T cells did not impact the outcome of L. major infection in C57BL/6 mice, which mounted a predominantly Th1 response and spontaneously resolved their infection similar to WT littermates. Our findings demonstrate that transgenic expression of CXCR3 on T cells increases susceptibility of BALB/c mice to L. major.
Collapse
Affiliation(s)
- Steve Oghumu
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, USA Department of Oral Biology, Ohio State University College of Dentistry, Columbus, Ohio, USA
| | - James C Stock
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, USA
| | - Sanjay Varikuti
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, USA
| | - Ran Dong
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Cesar Terrazas
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, USA
| | - Jessica A Edwards
- Department of Microbiology, Department of Microbial Infection and Immunity, Ohio State University, Columbus, Ohio, USA
| | - Chad A Rappleye
- Department of Microbiology, Department of Microbial Infection and Immunity, Ohio State University, Columbus, Ohio, USA
| | - Ariel Holovatyk
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, USA
| | - Arlene Sharpe
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Abhay R Satoskar
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, USA Department of Microbiology, Department of Microbial Infection and Immunity, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
39
|
Israely E, Ginsberg M, Nolan D, Ding BS, James D, Elemento O, Rafii S, Rabbany SY. Akt suppression of TGFβ signaling contributes to the maintenance of vascular identity in embryonic stem cell-derived endothelial cells. Stem Cells 2014; 32:177-90. [PMID: 23963623 DOI: 10.1002/stem.1521] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 07/23/2013] [Accepted: 07/24/2013] [Indexed: 01/04/2023]
Abstract
The ability to generate and maintain stable in vitro cultures of mouse endothelial cells (ECs) has great potential for genetic dissection of the numerous pathologies involving vascular dysfunction as well as therapeutic applications. However, previous efforts at achieving sustained cultures of primary stable murine vascular cells have fallen short, and the cellular requirements for EC maintenance in vitro remain undefined. In this study, we have generated vascular ECs from mouse embryonic stem (ES) cells and show that active Akt is essential to their survival and propagation as homogeneous monolayers in vitro. These cells harbor the phenotypical, biochemical, and functional characteristics of ECs and expand throughout long-term cultures, while maintaining their angiogenic capacity. Moreover, Akt-transduced embryonic ECs form functional perfused vessels in vivo that anastomose with host blood vessels. We provide evidence for a novel function of Akt in stabilizing EC identity, whereby the activated form of the protein protects mouse ES cell-derived ECs from TGFβ-mediated transdifferentiation by downregulating SMAD3. These findings identify a role for Akt in regulating the developmental potential of ES cell-derived ECs and demonstrate that active Akt maintains endothelial identity in embryonic ECs by interfering with active TGFβ-mediated processes that would ordinarily usher these cells to alternate fates.
Collapse
Affiliation(s)
- Edo Israely
- Department of Genetic Medicine, Ansary Stem Cell Institute, Howard Hughes Medical Institute, Weill Cornell Medical College, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Kshirsagar S, Binder E, Riedl M, Wechselberger G, Steichen E, Edelbauer M. Enhanced activity of Akt in Teff cells from children with lupus nephritis is associated with reduced induction of tumor necrosis factor receptor-associated factor 6 and increased OX40 expression. ACTA ACUST UNITED AC 2014; 65:2996-3006. [PMID: 23896866 DOI: 10.1002/art.38089] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 07/09/2013] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The breakdown of peripheral tolerance mechanisms is central to the pathogenesis of systemic lupus erythematosus (SLE). Although true Treg cells in patients with SLE exhibit intact suppressive activity, Teff cells are resistant to suppression. The underlying mechanisms are incompletely understood. This study was undertaken to examine the Akt signaling pathway and molecules that may alter its activity in T cells in lupus patients. METHODS The Akt pathway and its regulators were analyzed in Teff and Treg cells from children with lupus nephritis and controls using flow cytometry and real-time quantitative polymerase chain reaction. T cell proliferation was assessed by analysis of 5,6-carboxyfluorescein succinimidyl ester dilution. RESULTS CD4+CD45RA-FoxP3(low) and FoxP3- Teff cells from children with lupus nephritis expressed high levels of activated Akt, resulting in the down-regulation of the proapoptotic protein Bim and an enhanced proliferative response. The induction of tumor necrosis factor receptor-associated factor 6 (TRAF6) was impaired, and TRAF6 levels inversely correlated with Akt activity. Although the expression of OX40 was enhanced on Teff cells from children with lupus nephritis compared to controls, OX40 stimulation failed to significantly increase TRAF6 expression in cells from patients, in contrast to those from healthy controls, but resulted in further increased Akt activation that was reversed by blockade of OX40 signaling. Moreover, inhibition of Akt signaling markedly decreased the proliferation of Teff cells from lupus patients. CONCLUSION Our findings indicate that hyperactivation of the Akt pathway in Teff cells from children with lupus nephritis is associated with reduced induction of TRAF6 and up-regulation of OX40, which may cause Teff cell resistance to Treg cell-mediated suppression.
Collapse
|
41
|
Tomar N, De RK. A model of an integrated immune system pathway in Homo sapiens and its interaction with superantigen producing expression regulatory pathway in Staphylococcus aureus: comparing behavior of pathogen perturbed and unperturbed pathway. PLoS One 2013; 8:e80918. [PMID: 24324645 PMCID: PMC3855681 DOI: 10.1371/journal.pone.0080918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 10/17/2013] [Indexed: 11/19/2022] Open
Abstract
Response of an immune system to a pathogen attack depends on the balance between the host immune defense and the virulence of the pathogen. Investigation of molecular interactions between the proteins of a host and a pathogen helps in identifying the pathogenic proteins. It is necessary to understand the dynamics of a normally behaved host system to evaluate the capacity of its immune system upon pathogen attack. In this study, we have compared the behavior of an unperturbed and pathogen perturbed host system. Moreover, we have developed a formalism under Flux Balance Analysis (FBA) for the optimization of conflicting objective functions. We have constructed an integrated pathway system, which includes Staphylococcal Superantigen (SAg) expression regulatory pathway and TCR signaling pathway of Homo sapiens. We have implemented the method on this pathway system and observed the behavior of host signaling molecules upon pathogen attack. The entire study has been divided into six different cases, based on the perturbed/unperturbed conditions. In other words, we have investigated unperturbed and pathogen perturbed human TCR signaling pathway, with different combinations of optimization of concentrations of regulatory and signaling molecules. One of these cases has aimed at finding out whether minimization of the toxin production in a pathogen leads to the change in the concentration levels of the proteins coded by TCR signaling pathway genes in the infected host. Based on the computed results, we have hypothesized that the balance between TCR signaling inhibitory and stimulatory molecules can keep TCR signaling system into resting/stimulating state, depending upon the perturbation. The proposed integrated host-pathogen interaction pathway model has accurately reflected the experimental evidences, which we have used for validation purpose. The significance of this kind of investigation lies in revealing the susceptible interaction points that can take back the Staphylococcal Enterotoxin (SE)-challenged system within the range of normal behavior.
Collapse
Affiliation(s)
- Namrata Tomar
- Machine Intelligence Unit, Indian Statistical Institute, Kolkata, India
| | - Rajat K. De
- Machine Intelligence Unit, Indian Statistical Institute, Kolkata, India
| |
Collapse
|
42
|
Akinleye A, Avvaru P, Furqan M, Song Y, Liu D. Phosphatidylinositol 3-kinase (PI3K) inhibitors as cancer therapeutics. J Hematol Oncol 2013; 6:88. [PMID: 24261963 PMCID: PMC3843585 DOI: 10.1186/1756-8722-6-88] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/12/2013] [Indexed: 02/08/2023] Open
Abstract
Phosphatidylinositol 3-kinases (PI3Ks) are lipid kinases that regulate diverse cellular processes including proliferation, adhesion, survival, and motility. Dysregulated PI3K pathway signaling occurs in one-third of human tumors. Aberrantly activated PI3K signaling also confers sensitivity and resistance to conventional therapies. PI3K has been recognized as an attractive molecular target for novel anti-cancer molecules. In the last few years, several classes of potent and selective small molecule PI3K inhibitors have been developed, and at least fifteen compounds have progressed into clinical trials as new anticancer drugs. Among these, idelalisib has advanced to phase III trials in patients with advanced indolent non-Hodgkin's lymphoma and mantle cell lymphoma. In this review, we summarized the major molecules of PI3K signaling pathway, and discussed the preclinical models and clinical trials of potent small-molecule PI3K inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Delong Liu
- Division of Hematology/Oncology, Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595, USA.
| |
Collapse
|
43
|
Rego A, Trindade D, Chaves SR, Manon S, Costa V, Sousa MJ, Côrte-Real M. The yeast model system as a tool towards the understanding of apoptosis regulation by sphingolipids. FEMS Yeast Res 2013; 14:160-78. [DOI: 10.1111/1567-1364.12096] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/02/2013] [Accepted: 09/06/2013] [Indexed: 11/30/2022] Open
Affiliation(s)
- António Rego
- Departamento de Biologia; Centro de Biologia Molecular e Ambiental; Universidade do Minho; Braga Portugal
- Instituto de Biologia Molecular e Celular; Universidade do Porto; Porto Portugal
| | - Dário Trindade
- Departamento de Biologia; Centro de Biologia Molecular e Ambiental; Universidade do Minho; Braga Portugal
- CNRS; UMR5095; Université de Bordeaux 2; Bordeaux France
| | - Susana R. Chaves
- Departamento de Biologia; Centro de Biologia Molecular e Ambiental; Universidade do Minho; Braga Portugal
| | - Stéphen Manon
- CNRS; UMR5095; Université de Bordeaux 2; Bordeaux France
| | - Vítor Costa
- Instituto de Biologia Molecular e Celular; Universidade do Porto; Porto Portugal
- Departamento de Biologia Molecular; Instituto de Ciências Biomédicas Abel Salazar; Universidade do Porto; Porto Portugal
| | - Maria João Sousa
- Departamento de Biologia; Centro de Biologia Molecular e Ambiental; Universidade do Minho; Braga Portugal
| | - Manuela Côrte-Real
- Departamento de Biologia; Centro de Biologia Molecular e Ambiental; Universidade do Minho; Braga Portugal
| |
Collapse
|
44
|
Muscolini M, Camperio C, Capuano C, Caristi S, Piccolella E, Galandrini R, Tuosto L. Phosphatidylinositol 4-Phosphate 5-Kinase α Activation Critically Contributes to CD28-Dependent Signaling Responses. THE JOURNAL OF IMMUNOLOGY 2013; 190:5279-86. [DOI: 10.4049/jimmunol.1203157] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
45
|
Pierau M, Na SY, Simma N, Lowinus T, Marx A, Schraven B, Bommhardt UH. Constitutive Akt1 signals attenuate B-cell receptor signaling and proliferation, but enhance B-cell migration and effector function. Eur J Immunol 2013; 42:3381-93. [PMID: 22930469 DOI: 10.1002/eji.201242397] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 07/27/2012] [Accepted: 08/20/2012] [Indexed: 11/07/2022]
Abstract
Ligation of the BCR induces a complex signaling network that involves activation of Akt, a family of serine/threonine protein kinases associated with B-cell development, proliferation, and tumor formation. Here, we analyzed the effect of enhanced Akt1 signals on B-cell maturation and function. Unexpectedly, we found that peripheral B cells overexpressing Akt1 were less responsive to BCR stimuli. This correlated with a decrease in Ca(2+) -mobilization and proliferation, in an impaired activation of Erk1/2 and mammalian target of rapamycin (mTOR) kinases and poor mobilization of NFATc1 and NF-κB/p65 factors. In contrast, activation of STAT5 and migration of B cells toward the chemokine SDF1α was found to be enhanced. Akt1 Tg mice showed an altered maturation of peritoneal and splenic B1 B cells and an enhanced production of IgG1 and IgG3 upon immunization with the T-cell independent Ag TNP-Ficoll. Furthermore, mice homo-zygous for Tg Akt1 showed a severe block in the maturation of B-cell precursors in BM and a strong enrichment of plasma cells in spleen. Altogether, our data reveal that enhanced Akt1 signals modify BCR signaling strength and, thereby, B-cell development and effector function.
Collapse
Affiliation(s)
- Mandy Pierau
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Wensveen FM, van Gisbergen KPJM, Eldering E. The fourth dimension in immunological space: how the struggle for nutrients selects high-affinity lymphocytes. Immunol Rev 2013; 249:84-103. [PMID: 22889217 DOI: 10.1111/j.1600-065x.2012.01156.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lymphocyte activation via the antigen receptor is associated with radical shifts in metabolism and changes in requirements for nutrients and cytokines. Concomitantly, drastic changes occur in the expression of pro-and anti-apoptotic proteins that alter the sensitivity of lymphocytes to limiting concentrations of key survival factors. Antigen affinity is a primary determinant for the capacity of activated lymphocytes to access these vital resources. The shift in metabolic needs and the variable access to key survival factors is used by the immune system to eliminate activated low-affinity cells and to generate an optimal high-affinity response. In this review, we focus on the control of apoptosis regulators in activated lymphocytes by nutrients, cytokines, and costimulation. We propose that the struggle among individual clones that leads to the formation of high-affinity effector cell populations is in effect an 'invisible' fourth signal required for effective immune responses.
Collapse
Affiliation(s)
- Felix M Wensveen
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
47
|
Lind EF, Elford AR, Ohashi PS. Micro-RNA 155 is required for optimal CD8+ T cell responses to acute viral and intracellular bacterial challenges. THE JOURNAL OF IMMUNOLOGY 2012; 190:1210-6. [PMID: 23275599 DOI: 10.4049/jimmunol.1202700] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recent studies have begun to define the role of micro-RNAs in regulating the immune response. Micro-RNA155 (mir-155) has been shown to play a role in germinal center formation, T cell inflammation, and regulatory T cell development. In this study, we evaluated the role of mir-155 in cytotoxic T cell function. We report in this study that mice lacking mir-155 have impaired CD8(+) T cell responses to infections with lymphocytic choriomeningitis virus and the intracellular bacteria Listeria monocytogenes. We show by a series of adoptive transfer studies that the impaired CD8(+) T cell response to L. monocytogenes is T cell intrinsic. In addition, we observed that CD8(+) T cells lacking mir-155 have impaired activation of the prosurvival Akt pathway after TCR cross-linking. These data suggest that mir-155 may be a good target for therapies aimed at modulating immune responses.
Collapse
Affiliation(s)
- Evan F Lind
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, Toronto, Ontario M5G 2C1, Canada
| | | | | |
Collapse
|
48
|
Bone RN, Icyuz M, Zhang Y, Zhang Y, Cui W, Wang H, Peng JB, Matthews QL, Siegal GP, Wu H. Gene transfer of active Akt1 by an infectivity-enhanced adenovirus impacts β-cell survival and proliferation differentially in vitro and in vivo. Islets 2012; 4. [PMID: 23183538 PMCID: PMC3605165 DOI: 10.4161/isl.22721] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Type 1 Diabetes is characterized by an absolute insulin deficiency due to the autoimmune destruction of insulin producing β-cells in the pancreatic islets. Akt1/Protein Kinase B is the direct downstream target of PI3 Kinase activation, and has shown potent anti-apoptotic and proliferation-inducing activities. This study was designed to explore whether gene transfer of constitutively active Akt1 (CA-Akt1) would promote β-cell survival and proliferation, thus be protective against experimental diabetes. In the study, a fiber-modified infectivity-enhanced adenoviral vector, Ad5RGDpK7, was used to deliver rat insulin promoter (RIP)-driven CA-Akt1 into β-cells. Our data showed this vector efficiently delivered CA-Akt1 into freshly isolated pancreatic islets, and promoted islet cell survival and β-cell proliferation in vitro. The therapeutic effect of the vector in vivo was assessed using streptozotocin (STZ)-induced diabetes mice. Two means of vector administration were explored: intravenous and intra-bile ductal injections. While direct vector administration into pancreas via bile-ductal injection resulted in local adverse effect, intravenous injection of the vectors offered therapeutic benefits. Further analysis suggests systemic vector administration caused endogenous Akt expression and activation in islets, which may be responsible, at least in part, for the protective effect of the infectivity-enhanced CA-Akt1 gene delivery vector. Taken together, our data suggest CA-Akt1 is effective in promoting β-cell survival and proliferation in vitro, but direct in vivo use is compromised by the efficacy of transgene delivery into β-cells. Nonetheless, the vector evoked the expression and activation of endogenous Akt in the islets, thus offering beneficial bystander effect against STZ-induced diabetes.
Collapse
Affiliation(s)
- Robert N. Bone
- Department of Pathology; University of Alabama at Birmingham; Birmingham, AL USA
| | - Mert Icyuz
- Department of Medicine; University of Alabama at Birmingham; Birmingham, AL USA
| | - Yanqing Zhang
- Department of Medicine; Section of Endocrinology; Tulane University; New Orleans, LA USA
| | - Yuan Zhang
- Department of Medicine; University of Alabama at Birmingham; Birmingham, AL USA
| | - Wanxing Cui
- Department of Surgery; University of Alabama at Birmingham; Birmingham, AL USA
| | - Hongjun Wang
- Department of Surgery; Medical University of South Carolina; Charleston, SC USA
| | - Ji-Bin Peng
- Department of Medicine; University of Alabama at Birmingham; Birmingham, AL USA
| | - Qiana L. Matthews
- Department of Medicine; University of Alabama at Birmingham; Birmingham, AL USA
| | - Gene P. Siegal
- Department of Pathology; University of Alabama at Birmingham; Birmingham, AL USA
| | - Hongju Wu
- Department of Medicine; Section of Endocrinology; Tulane University; New Orleans, LA USA
- Correspondence to: Hongju Wu,
| |
Collapse
|
49
|
Pagán AJ, Pepper M, Chu HH, Green JM, Jenkins MK. CD28 promotes CD4+ T cell clonal expansion during infection independently of its YMNM and PYAP motifs. THE JOURNAL OF IMMUNOLOGY 2012; 189:2909-17. [PMID: 22896637 DOI: 10.4049/jimmunol.1103231] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
CD28 is required for maximal proliferation of CD4+ T cells stimulated through their TCRs. Two sites within the cytoplasmic tail of CD28, a YMNM sequence that recruits PI3K and activates NF-κB and a PYAP sequence that recruits Lck, are candidates as transducers of the signals responsible for these biological effects. We tested this proposition by tracking polyclonal peptide:MHCII-specific CD4+ T cells in vivo in mice with mutations in these sites. Mice lacking CD28 or its cytoplasmic tail had the same number of naive T cells specific for a peptide:MHCII ligand as wild-type mice. However, the mutant cells produced one tenth as many effector and memory cells as wild-type T cells after infection with bacteria expressing the antigenic peptide. Remarkably, T cells with a mutated PI3K binding site, a mutated PYAP site, or both mutations proliferated to the same extent as wild-type T cells. The only observed defect was that T cells with a mutated PYAP or Y170F site proliferated even more weakly in response to peptide without adjuvant than wild-type T cells. These results show that CD28 enhances T cell proliferation during bacterial infection by signals emanating from undiscovered sites in the cytoplasmic tail.
Collapse
Affiliation(s)
- Antonio J Pagán
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
50
|
Pfeifhofer-Obermair C, Thuille N, Baier G. Involvement of distinct PKC gene products in T cell functions. Front Immunol 2012; 3:220. [PMID: 22888329 PMCID: PMC3412260 DOI: 10.3389/fimmu.2012.00220] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 07/08/2012] [Indexed: 01/07/2023] Open
Abstract
It is well established that members of the protein kinase C (PKC) family seem to have important roles in T cells. Focusing on the physiological and non-redundant PKC functions established in primary mouse T cells via germline gene-targeting approaches, our current knowledge defines two particularly critical PKC gene products, PKCθ and PKCα, as the "flavor of PKC" in T cells that appear to have a positive role in signaling pathways that are necessary for full antigen receptor-mediated T cell activation ex vivo and T cell-mediated immunity in vivo. Consistently, in spite of the current dogma that PKCθ inhibition might be sufficient to achieve complete immunosuppressive effects, more recent results have indicated that the pharmacological inhibition of PKCθ, and additionally, at least PKCα, appears to be needed to provide a successful approach for the prevention of allograft rejection and treatment of autoimmune diseases.
Collapse
Affiliation(s)
| | | | - Gottfried Baier
- Division of Cell Genetics, Department of Pharmacology and Genetics, Medical University Innsbruck, Innsbruck,Tyrol, Austria
| |
Collapse
|