1
|
Jin H, Li Q, Tang L, Naseem S, Park S, Wang E, Sun B, Manzoor A, Hur SJ, Li X, Choi SH. The L-Ascorbic Acid Increases Proliferation and Differentiation of Yanbian Cattle Skeletal Muscle Satellite Cells by Activating the Akt/mTOR/P70S6K Signaling Pathway. Food Sci Anim Resour 2025; 45:484-503. [PMID: 40093636 PMCID: PMC11907417 DOI: 10.5851/kosfa.2024.e50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 03/19/2025] Open
Abstract
Cell-cultured meat, as a new type of meat food, can effectively mitigate the negative effects of conventional animal husbandry on the environment, health, and animal welfare. Muscle stem cells are the main seed cells for the production of cell-cultured meat, but their weak proliferative capacity in vitro severely limits the large-scale and low-cost production of cell-cultured meat. There is growing evidence that L-ascorbic acid (AA) has the ability to increase the efficiency of muscle stem cell proliferation and differentiation in vitro. However, the role of AA in Yanbian bovine skeletal muscle satellite cells (BSCs) and its molecular mechanisms are unknown. Therefore, in the present study, the promotional effect of AA on the proliferation and differentiation of BSCs was confirmed by the Cell Counting Kit 8 (CCK-8) assay, 5-ethyl-2'-deoxyuridine (EdU) proliferation assay, real-time quantitative PCR (RT-qPCR), immunoprotein blotting (Western blotting) and immunofluorescence assay. RT-qPCR and Western blotting results showed that AA up-regulated the expression of p-Akt, p-mTOR, and p-P70S6K genes and proteins, whereas when the mTOR pathway inhibitor rapamycin was co-treated with AA in BSCs, the expression of p-Akt, p-mTOR, and p-P70S6K genes and proteins was significantly down-regulated. In summary, data suggest that AA regulates the proliferation and differentiation of BSCs by activating the AKT/mTOR/P70S6K signaling pathway. These data provide a practical approach and theoretical basis for the efficient and low-cost manufacture of cell culture meat.
Collapse
Affiliation(s)
- Huaina Jin
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Qiang Li
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Lin Tang
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Sajida Naseem
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Sungkwon Park
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| | - Enze Wang
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Bin Sun
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Abid Manzoor
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Sun Jin Hur
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Xiangzi Li
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Department of Animal Science, Yanbian University, Yanji 133002, China
| | - Seong-Ho Choi
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
2
|
Alvarez-Salazar EK, Cortés-Hernández A, Arteaga-Cruz S, Soldevila G. Induced regulatory T cells as immunotherapy in allotransplantation and autoimmunity: challenges and opportunities. J Leukoc Biol 2024; 116:947-965. [PMID: 38630873 DOI: 10.1093/jleuko/qiae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/14/2024] [Accepted: 02/21/2024] [Indexed: 04/19/2024] Open
Abstract
Regulatory T cells play a crucial role in the homeostasis of the immune response. Regulatory T cells are mainly generated in the thymus and are characterized by the expression of Foxp3, which is considered the regulatory T-cell master transcription factor. In addition, regulatory T cells can be induced from naive CD4+ T cells to express Foxp3 under specific conditions both in vivo (peripheral regulatory T cells) and in vitro (induced regulatory T cells). Both subsets of thymic regulatory T cells and peripheral regulatory T cells are necessary for the establishment of immune tolerance to self and non-self antigens. Although it has been postulated that induced regulatory T cells may be less stable compared to regulatory T cells, mainly due to epigenetic differences, accumulating evidence in animal models shows that induced regulatory T cells are stable in vivo and can be used for the treatment of inflammatory disorders, including autoimmune diseases and allogeneic transplant rejection. In this review, we describe the biological characteristics of induced regulatory T cells, as well as the key factors involved in induced regulatory T-cell transcriptional, metabolic, and epigenetic regulation, and discuss recent advances for de novo generation of stable regulatory T cells and their use as immunotherapeutic tools in different experimental models. Moreover, we discuss the challenges and considerations for the application of induced regulatory T cells in clinical trials and describe the new approaches proposed to achieve in vivo stability, including functional or metabolic reprogramming and epigenetic editing.
Collapse
Affiliation(s)
- Evelyn Katy Alvarez-Salazar
- Department of Immunology and National Laboratory of Flow Cytometry, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, Ciudad Universitaria, Colonia Copilco, Delegación Coyoacan, Apartado Postal 70228, CP 04510 Mexico City, Mexico
| | - Arimelek Cortés-Hernández
- Department of Immunology and National Laboratory of Flow Cytometry, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, Ciudad Universitaria, Colonia Copilco, Delegación Coyoacan, Apartado Postal 70228, CP 04510 Mexico City, Mexico
| | - Saúl Arteaga-Cruz
- Department of Immunology and National Laboratory of Flow Cytometry, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, Ciudad Universitaria, Colonia Copilco, Delegación Coyoacan, Apartado Postal 70228, CP 04510 Mexico City, Mexico
| | - Gloria Soldevila
- Department of Immunology and National Laboratory of Flow Cytometry, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, Ciudad Universitaria, Colonia Copilco, Delegación Coyoacan, Apartado Postal 70228, CP 04510 Mexico City, Mexico
| |
Collapse
|
3
|
Kabelitz D, Cierna L, Juraske C, Zarobkiewicz M, Schamel WW, Peters C. Empowering γδ T-cell functionality with vitamin C. Eur J Immunol 2024; 54:e2451028. [PMID: 38616772 DOI: 10.1002/eji.202451028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Vitamin C (ascorbic acid) is a potent antioxidant and a cofactor for various enzymes including histone demethylases and methylcytosine dioxygenases. Vitamin C also exerts direct cytotoxicity toward selected tumor cells including colorectal carcinoma. Moreover, vitamin C has been shown to impact immune cell differentiation at various levels including maturation and/or functionality of T cells and their progenitors, dendritic cells, B cells, and NK cells. γδ T cells have recently attracted great interest as effector cells for cell-based cancer immunotherapy, due to their HLA-independent recognition of a large variety of tumor cells. While γδ T cells can thus be also applied as an allogeneic off-the-shelf product, it is obvious that the effector function of γδ T cells needs to be optimized to ensure the best possible clinical efficacy. Here we review the immunomodulatory mechanisms of vitamin C with a special focus on how vitamin C enhances the effector function of γδ T cells. We also discuss future directions of how vitamin C can be used in the clinical setting to boost the efficacy of adoptive cell therapies.
Collapse
Affiliation(s)
- Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts University and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
- Institute of Immunology, UKSH Campus Kiel, Kiel, Germany
| | - Lea Cierna
- Institute of Immunology, Christian-Albrechts University and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Claudia Juraske
- Signalling Research Centres BIOSS and CIBSS, and Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Michal Zarobkiewicz
- Institute of Immunology, Christian-Albrechts University and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Wolfgang W Schamel
- Signalling Research Centres BIOSS and CIBSS, and Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Peters
- Institute of Immunology, Christian-Albrechts University and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| |
Collapse
|
4
|
Wang S, Yao Y, Song L, Zhai Z, Li S, Wang Z, Shen L, Zhu Y, Liu W, Gao C. ROS-responsive drug-releasing injectable microgels for ameliorating myocardial infarction. Biomaterials 2024; 307:122534. [PMID: 38518589 DOI: 10.1016/j.biomaterials.2024.122534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/05/2024] [Accepted: 03/11/2024] [Indexed: 03/24/2024]
Abstract
Despite of the recent advances in regulatory T cell (Treg) therapy, a limited number of available cells and specificity at the desired tissue site have severely compromised their efficacy. Herein, an injectable drug-releasing (MTK-TK-drug) microgel system in response to in situ stimulation by reactive oxygen species (ROS) was constructed with a coaxial capillary microfluidic system and UV curing. The spherical microgels with a size of 150 μm were obtained. The MTK-TK-drug microgels efficiently converted the pro-inflammatory Th17 cells into anti-inflammatory regulatory T cells (Treg) cells in vitro, and the ROS-scavenging materials synergistically enhanced the effect by modulating the inflammation microenvironment. Thus, the microgels significantly reduced cardiomyocyte apoptosis and decreased the inflammatory response in the early stages of post-myocardial infarction (MI) in vivo, thereby reducing fibrosis, promoting vascularization, and preserving cardiac function. Overall, our results indicate that the MTK-TK-drug microgels can attenuate the inflammatory response and improve MI therapeutic effects in vivo.
Collapse
Affiliation(s)
- Shuqin Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yuejun Yao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Liang Song
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Zihe Zhai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Shifen Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Zhaolong Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Liyin Shen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yang Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China.
| | - Wenxing Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
5
|
Sasidharan Nair V, Huehn J. Impact of vitamin C on the development, differentiation and functional properties of T cells. Eur J Microbiol Immunol (Bp) 2024; 14:67-74. [PMID: 38446173 PMCID: PMC11097783 DOI: 10.1556/1886.2024.00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Vitamin C plays a multifaceted role in various biological processes and is well-known to facilitate pleiotropic activities in both innate and adaptive immune responses, where the antioxidant capacity of vitamin C is most likely highly relevant since immune responses mainly occur in reducing environments. Beyond its antioxidant properties, vitamin C can enhance the transcription potential of genes by promoting DNA demethylation through ten-eleven-translocation (Tet) methylcytosine dioxygenases, which have been recently demonstrated to be critical for the development and differentiation of T cells. In this minireview, we will provide a broader overview on the impact of vitamin C on signaling and regulatory activities in both innate and adaptive immune cells. Particularly, we will summarize recent findings on the decisive role of finely tuned vitamin C concentrations for T cell development, T helper cell differentiation, and optimal T cell-mediated immune responses.
Collapse
Affiliation(s)
- Varun Sasidharan Nair
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124Braunschweig, Germany
- Genetics and Epigenetics of Behavior, Italian Institute of Technology, Via Morego 30, 16163Genova, Italy
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124Braunschweig, Germany
| |
Collapse
|
6
|
Fowler AA. Vitamin C: Rationale for Its Use in Sepsis-Induced Acute Respiratory Distress Syndrome (ARDS). Antioxidants (Basel) 2024; 13:95. [PMID: 38247519 PMCID: PMC10812524 DOI: 10.3390/antiox13010095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening event that occurs in patients suffering from bacterial, fungal, or viral sepsis. Research performed over the last five decades showed that ARDS is a consequence of severe unrestrained systemic inflammation, which leads to injury of the lung's microvasculature and alveolar epithelium. ARDS leads to acute hypoxic/hypercapnic respiratory failure and death in a significant number of patients hospitalized in intensive care units worldwide. Basic and clinical research performed during the time since ARDS was first described has been unable to construct a pharmacological agent that will combat the inflammatory fire leading to ARDS. In-depth studies of the molecular pharmacology of vitamin C indicate that it can serve as a potent anti-inflammatory agent capable of attenuating the pathobiological events that lead to acute injury of the lungs and other body organs. This analysis of vitamin C's role in the treatment of ARDS includes a focused systematic review of the literature relevant to the molecular physiology of vitamin C and to the past performance of clinical trials using the agent.
Collapse
Affiliation(s)
- Alpha A Fowler
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
7
|
Phadke I, Pouzolles M, Machado A, Moraly J, Gonzalez-Menendez P, Zimmermann VS, Kinet S, Levine M, Violet PC, Taylor N. Vitamin C deficiency reveals developmental differences between neonatal and adult hematopoiesis. Front Immunol 2022; 13:898827. [PMID: 36248829 PMCID: PMC9562198 DOI: 10.3389/fimmu.2022.898827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/02/2022] [Indexed: 11/25/2022] Open
Abstract
Hematopoiesis, a process that results in the differentiation of all blood lineages, is essential throughout life. The production of 1x1012 blood cells per day, including 200x109 erythrocytes, is highly dependent on nutrient consumption. Notably though, the relative requirements for micronutrients during the perinatal period, a critical developmental window for immune cell and erythrocyte differentiation, have not been extensively studied. More specifically, the impact of the vitamin C/ascorbate micronutrient on perinatal as compared to adult hematopoiesis has been difficult to assess in animal models. Even though humans cannot synthesize ascorbate, due to a pseudogenization of the L-gulono-γ-lactone oxidase (GULO) gene, its generation from glucose is an ancestral mammalian trait. Taking advantage of a Gulo-/- mouse model, we show that ascorbic acid deficiency profoundly impacts perinatal hematopoiesis, resulting in a hypocellular bone marrow (BM) with a significant reduction in hematopoietic stem cells, multipotent progenitors, and hematopoietic progenitors. Furthermore, myeloid progenitors exhibited differential sensitivity to vitamin C levels; common myeloid progenitors and megakaryocyte-erythrocyte progenitors were markedly reduced in Gulo-/- pups following vitamin C depletion in the dams, whereas granulocyte-myeloid progenitors were spared, and their frequency was even augmented. Notably, hematopoietic cell subsets were rescued by vitamin C repletion. Consistent with these data, peripheral myeloid cells were maintained in ascorbate-deficient Gulo-/- pups while other lineage-committed hematopoietic cells were decreased. A reduction in B cell numbers was associated with a significantly reduced humoral immune response in ascorbate-depleted Gulo-/- pups but not adult mice. Erythropoiesis was particularly sensitive to vitamin C deprivation during both the perinatal and adult periods, with ascorbate-deficient Gulo-/- pups as well as adult mice exhibiting compensatory splenic differentiation. Furthermore, in the pathological context of hemolytic anemia, vitamin C-deficient adult Gulo-/- mice were not able to sufficiently increase their erythropoietic activity, resulting in a sustained anemia. Thus, vitamin C plays a pivotal role in the maintenance and differentiation of hematopoietic progenitors during the neonatal period and is required throughout life to sustain erythroid differentiation under stress conditions.
Collapse
Affiliation(s)
- Ira Phadke
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Marie Pouzolles
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Alice Machado
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Josquin Moraly
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Pedro Gonzalez-Menendez
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Valérie S. Zimmermann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Sandrina Kinet
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Mark Levine, ; Pierre-Christian Violet, ; Naomi Taylor,
| | - Pierre-Christian Violet
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Mark Levine, ; Pierre-Christian Violet, ; Naomi Taylor,
| | - Naomi Taylor
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- *Correspondence: Mark Levine, ; Pierre-Christian Violet, ; Naomi Taylor,
| |
Collapse
|
8
|
Zaher A, Stephens LM, Miller AM, Hartwig SM, Stolwijk JM, Petronek MS, Zacharias ZR, Wadas TJ, Monga V, Cullen JJ, Furqan M, Houtman JCD, Varga SM, Spitz DR, Allen BG. Pharmacological ascorbate as a novel therapeutic strategy to enhance cancer immunotherapy. Front Immunol 2022; 13:989000. [PMID: 36072595 PMCID: PMC9444023 DOI: 10.3389/fimmu.2022.989000] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
Pharmacological ascorbate (i.e., intravenous infusions of vitamin C reaching ~ 20 mM in plasma) is under active investigation as an adjuvant to standard of care anti-cancer treatments due to its dual redox roles as an antioxidant in normal tissues and as a prooxidant in malignant tissues. Immune checkpoint inhibitors (ICIs) are highly promising therapies for many cancer patients but face several challenges including low response rates, primary or acquired resistance, and toxicity. Ascorbate modulates both innate and adaptive immune functions and plays a key role in maintaining the balance between pro and anti-inflammatory states. Furthermore, the success of pharmacological ascorbate as a radiosensitizer and a chemosensitizer in pre-clinical studies and early phase clinical trials suggests that it may also enhance the efficacy and expand the benefits of ICIs.
Collapse
Affiliation(s)
- Amira Zaher
- Cancer Biology Program, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Laura M. Stephens
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Ann M. Miller
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Stacey M. Hartwig
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Jeffrey M. Stolwijk
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Michael S. Petronek
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Zeb R. Zacharias
- Human Immunology Core & Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Thaddeus J. Wadas
- Department of Radiology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Varun Monga
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Joseph J. Cullen
- Department of Surgery, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Muhammad Furqan
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Jon C. D. Houtman
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Steven M. Varga
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
- *Correspondence: Bryan G. Allen,
| |
Collapse
|
9
|
Jiang Z, Wu C. Reciprocal Interactions Between Regulatory T Cells and Intestinal Epithelial Cells. Front Immunol 2022; 13:951339. [PMID: 35860233 PMCID: PMC9289291 DOI: 10.3389/fimmu.2022.951339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/09/2022] [Indexed: 11/25/2022] Open
Abstract
It has been well established that Foxp3+ regulatory T cells (Treg cells) play a crucial role for immune repression and tolerance, protecting the body from autoimmunity and inflammation. Previous studies indicate that intestinal Treg cells are one specialized population of Treg cells, distinct from those in other organ compartments, both functionally and phenotypically. Specific external and internal signals, particularly the presence of microbiota, shape these Treg cells to better cooperate with the gut ecosystem, controlling intestinal physiology. The integrity of intestinal epithelial barrier represents a key feature of gut immune tolerance, which can be regulated by multiple factors. Emerging evidence suggests that bidirectional interactions between gut epithelium and resident T cells significantly contribute to intestinal barrier function. Understanding how Treg cells regulate intestinal barrier integrity provides insights into immune tolerance-mediated mucosal homeostasis, which can further illuminate potential therapeutic strategies for treating inflammatory bowel disease and colon cancer.
Collapse
Affiliation(s)
- Zhiqiang Jiang
- Sun-Yat Sen University, School of Medicine, Guangzhou, China
- *Correspondence: Zhiqiang Jiang, ; Chuan Wu,
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States
- *Correspondence: Zhiqiang Jiang, ; Chuan Wu,
| |
Collapse
|
10
|
Vitamin C and its therapeutic potential in the management of COVID19. Clin Nutr ESPEN 2022; 50:8-14. [PMID: 35871955 PMCID: PMC9166267 DOI: 10.1016/j.clnesp.2022.05.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 04/01/2022] [Accepted: 05/31/2022] [Indexed: 12/18/2022]
Abstract
COVID19 has emerged as one of the worst pandemics in the history of mankind. Several vaccines have been approved by different government agencies worldwide, but data on their efficacy and safety are limited, and distribution remains a massive challenge. As per WHO, personal immunity is vital for protection against COVID19. Earlier, Vitamin C-mediated pathways have been shown to play critical role in boosting immunity attributed to its antioxidant properties. Recently, the involvement of such pathways in protection against COVID19 has been suggested. The controlled doses of Vitamin C administered through intravenous (IV) injections are being studied for determining its role in the prognosis of COVID19. In this article, we have discussed the potential role of Vitamin C in the management in COVID19 patients and presented recent clinical trials data. Additionally, we have elaborated the possibility of administering Vitamin C through inhalers in order to achieve local high concentration and the challenges of such approach.
Collapse
|
11
|
Chen HY, Hsu M, Lio CWJ. Micro but mighty-Micronutrients in the epigenetic regulation of adaptive immune responses. Immunol Rev 2022; 305:152-164. [PMID: 34820863 PMCID: PMC8766944 DOI: 10.1111/imr.13045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 01/03/2023]
Abstract
Micronutrients are essential small molecules required by organisms in minute quantity for survival. For instance, vitamins and minerals, the two major categories of micronutrients, are central for biological processes such as metabolism, cell replication, differentiation, and immune response. Studies estimated that around two billion humans worldwide suffer from micronutrient deficiencies, also known as "hidden hunger," linked to weakened immune responses. While micronutrients affect the immune system at multiple levels, recent studies showed that micronutrients potentially impact the differentiation and function of immune cells as cofactors for epigenetic enzymes, including the 2-oxoglutarate-dependent dioxygenase (2OGDD) family involved in histone and DNA demethylation. Here, we will first provide an overview of the role of DNA methylation in T cells and B cells, followed by the micronutrients ascorbate (vitamin C) and iron, two critical cofactors for 2OGDD. We will discuss the emerging evidence of these micronutrients could regulate adaptive immune response by influencing epigenetic remodeling.
Collapse
Affiliation(s)
| | | | - Chan-Wang Jerry Lio
- Corresponding author: Chan-Wang Jerry Lio (), Address: 460 W 12 Ave, Columbus, Ohio, USA 43064, Tel: (614)-247-5337
| |
Collapse
|
12
|
Piotrowska M, Gliwiński M, Trzonkowski P, Iwaszkiewicz-Grzes D. Regulatory T Cells-Related Genes Are under DNA Methylation Influence. Int J Mol Sci 2021; 22:7144. [PMID: 34281195 PMCID: PMC8267835 DOI: 10.3390/ijms22137144] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) exert a highly suppressive function in the immune system. Disturbances in their function predispose an individual to autoimmune dysregulation, with a predominance of the pro-inflammatory environment. Besides Foxp3, which is a master regulator of these cells, other genes (e.g., Il2ra, Ctla4, Tnfrsf18, Ikzf2, and Ikzf4) are also involved in Tregs development and function. Multidimensional Tregs suppression is determined by factors that are believed to be crucial in the action of Tregs-related genes. Among them, epigenetic changes, such as DNA methylation, tend to be widely studied over the past few years. DNA methylation acts as a repressive mark, leading to diminished gene expression. Given the role of increased CpG methylation upon Tregs imprinting and functional stability, alterations in the methylation pattern can cause an imbalance in the immune response. Due to the fact that epigenetic changes can be reversible, so-called epigenetic modifiers are broadly used in order to improve Tregs performance. In this review, we place emphasis on the role of DNA methylation of the genes that are key regulators of Tregs function. We also discuss disease settings that have an impact on the methylation status of Tregs and systematize the usefulness of epigenetic drugs as factors able to influence Tregs functions.
Collapse
Affiliation(s)
| | | | | | - Dorota Iwaszkiewicz-Grzes
- Department of Medical Immunology, Medical University of Gdansk, 80-210 Gdańsk, Poland; (M.P.); (M.G.); (P.T.)
| |
Collapse
|
13
|
Blagih J, Hennequart M, Zani F. Tissue Nutrient Environments and Their Effect on Regulatory T Cell Biology. Front Immunol 2021; 12:637960. [PMID: 33868263 PMCID: PMC8050341 DOI: 10.3389/fimmu.2021.637960] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Regulatory T cells (Tregs) are essential for mitigating inflammation. Tregs are found in nearly every tissue and play either beneficial or harmful roles in the host. The availability of various nutrients can either enhance or impair Treg function. Mitochondrial oxidative metabolism plays a major role in supporting Treg differentiation and fitness. While Tregs rely heavily on oxidation of fatty acids to support mitochondrial activity, they have found ways to adapt to different tissue types, such as tumors, to survive in competitive environments. In addition, metabolic by-products from commensal organisms in the gut also have a profound impact on Treg differentiation. In this review, we will focus on the core metabolic pathways engaged in Tregs, especially in the context of tissue nutrient environments, and how they can affect Treg function, stability and differentiation.
Collapse
Affiliation(s)
| | | | - Fabio Zani
- The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
14
|
Hwang JH, Piao H, Jang JY, Lee SK, Han D, Lee GM, Go C, Kim Y, Oh KI, Kang JS, Yan JJ, Yang J. Suppressive effects of vitamin C-treated induced-regulatory T cells on heart allograft rejection under vitamin C-deficient or -sufficient conditions. PLoS One 2021; 16:e0246967. [PMID: 33577562 PMCID: PMC7880463 DOI: 10.1371/journal.pone.0246967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/28/2021] [Indexed: 12/30/2022] Open
Abstract
Foxp3 stability of vitamin C-treated induced-regulatory T cells (V-iTregs) is superior to that of conventional iTregs (C-iTregs). However, the role of V-iTregs in allograft rejection under vitamin C-deficient conditions, such as those seen in humans, remains unclear. We aimed to elucidate the role of vitamin C treatment on generation and maintenance of iTregs from gulo knockout (Gulo-KO) mice as well as wild type (WT) mice, and in vitro and in vivo suppressive effects of V-iTregs on heart allograft rejection in either Gulo-KO or WT recipient mice. Conversion efficiency of iTregs was similar between C- and V-iTregs in both WT and Gulo-KO mice. V-iTregs from WT or Gulo-KO mice showed better in vitro Foxp3 stability than C-iTregs, although there was no difference between WT V-iTregs and Gulo-KO V-iTregs. Furthermore, V-iTregs from WT or Gulo-KO mice suppressed in vitro T cell proliferation better than C-iTregs. Heterotrophic heart transplantation from BALB/c mice to WT or vitamin C-deficient Gulo-KO C57BL/6J mice was performed following adoptive transfer of C- or V-iTregs. V-iTregs as well as C-iTregs prolonged heart allograft survival in WT and Gulo-KO mice. However, there was no difference between the C- and V-iTreg groups. Supplementation of low- or high-dose vitamin C did not induce significant changes in heart allograft survival in Gulo-KO recipients that had received V-iTregs. In conclusion, V-iTregs do not exert better suppressive effects on heart allograft survival than C-iTregs in either WT or vitamin C-deficient recipients.
Collapse
Affiliation(s)
- Ju Hee Hwang
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Honglin Piao
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Medicine, Graduate School, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joon Young Jang
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sun-Kyung Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Medicine, Graduate School, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dongkyu Han
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Gwang-Min Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Cheolhyeon Go
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yejin Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Kwon Ik Oh
- Department of Pathology, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Jae Seung Kang
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Ji-Jing Yan
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jaeseok Yang
- Transplantation Center, Seoul National University Hospital, Seoul, Republic of Korea
- Department of surgery, Seoul National University hospital, Seoul, Republic of Korea
| |
Collapse
|
15
|
Saksida T, Jevtić B, Djedović N, Miljković Đ, Stojanović I. Redox Regulation of Tolerogenic Dendritic Cells and Regulatory T Cells in the Pathogenesis and Therapy of Autoimmunity. Antioxid Redox Signal 2021; 34:364-382. [PMID: 32458699 DOI: 10.1089/ars.2019.7999] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significance: Autoimmune diseases are progressively affecting westernized societies, as the proportion of individuals suffering from autoimmunity is steadily increasing over the past decades. Understanding the role of reactive oxygen species (ROS) in modulation of the immune response in the pathogenesis of autoimmune disorders is of utmost importance. The focus of this review is the regulation of ROS production within tolerogenic dendritic cells (tolDCs) and regulatory T (Treg) cells that have the essential role in the prevention of autoimmune diseases and significant potency in their therapy. Recent Advances: It is now clear that ROS are extremely important for the proper function of both DC and T cells. Antigen processing/presentation and the ability of DC to activate T cells depend upon the ROS availability. Treg differentiation, suppressive function, and stability are profoundly influenced by ROS presence. Critical Issues: Although a plethora of results on the relation between ROS and immune cells exist, it remains unclear whether ROS modulation is a productive way for skewing T cells and DCs toward a tolerogenic phenotype. Also, the possibility of ROS modulation for enhancement of regulatory properties of DC and Treg during their preparation for use in cellular therapy has to be clarified. Future Directions: Studies of DC and T cell redox regulation should allow for the improvement of the therapy of autoimmune diseases. This could be achieved through the direct therapeutic application of ROS modulators in autoimmunity, or indirectly through ROS-dependent enhancement of tolDC and Treg preparation for cell-based immunotherapy. Antioxid. Redox Signal. 34, 364-382.
Collapse
Affiliation(s)
- Tamara Saksida
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Neda Djedović
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
16
|
Kurniawan H, Soriano-Baguet L, Brenner D. Regulatory T cell metabolism at the intersection between autoimmune diseases and cancer. Eur J Immunol 2020; 50:1626-1642. [PMID: 33067808 PMCID: PMC7756807 DOI: 10.1002/eji.201948470] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/28/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022]
Abstract
Regulatory T cells (Tregs) are critical for peripheral immune tolerance and homeostasis, and altered Treg behavior is involved in many pathologies, including autoimmunity and cancer. The expression of the transcription factor FoxP3 in Tregs is fundamental to maintaining their stability and immunosuppressive function. Recent studies have highlighted the crucial role that metabolic reprogramming plays in controlling Treg plasticity, stability, and function. In this review, we summarize how the availability and use of various nutrients and metabolites influence Treg metabolic pathways and activity. We also discuss how Treg-intrinsic metabolic programs define and shape their differentiation, FoxP3 expression, and suppressive capacity. Lastly, we explore how manipulating the regulation of Treg metabolism might be exploited in different disease settings to achieve novel immunotherapies.
Collapse
Affiliation(s)
- Henry Kurniawan
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology and Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Leticia Soriano-Baguet
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology and Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Dirk Brenner
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology and Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
17
|
Masuelli L, Benvenuto M, Focaccetti C, Ciuffa S, Fazi S, Bei A, Miele MT, Piredda L, Manzari V, Modesti A, Bei R. Targeting the tumor immune microenvironment with "nutraceuticals": From bench to clinical trials. Pharmacol Ther 2020; 219:107700. [PMID: 33045254 DOI: 10.1016/j.pharmthera.2020.107700] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2020] [Indexed: 02/06/2023]
Abstract
The occurrence of immune effector cells in the tissue microenvironment during neoplastic progression is critical in determining tumor growth outcomes. On the other hand, tumors may also avoid immune system-mediated elimination by recruiting immunosuppressive leukocytes and soluble factors, which coordinate a tumor microenvironment that counteracts the efficiency of the antitumor immune response. Checkpoint inhibitor therapy results have indicated a way forward via activation of the immune system against cancer. Widespread evidence has shown that different compounds in foods, when administered as purified substances, can act as immunomodulators in humans and animals. Although there is no universally accepted definition of nutraceuticals, the term identifies a wide category of natural compounds that may impact health and disease statuses and includes purified substances from natural sources, plant extracts, dietary supplements, vitamins, phytonutrients, and various products with combinations of functional ingredients. In this review, we summarize the current knowledge on the immunomodulatory effects of nutraceuticals with a special focus on the cancer microenvironment, highlighting the conceptual benefits or drawbacks and subtle cell-specific effects of nutraceuticals for envisioning future therapies employing nutraceuticals as chemoadjuvants.
Collapse
Affiliation(s)
- Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Monica Benvenuto
- Saint Camillus International University of Health and Medical Sciences, via di Sant'Alessandro 8, 00131 Rome, Italy; Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Chiara Focaccetti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy; Department of Human Science and Promotion of the Quality of Life, San Raffaele University Rome, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Sara Ciuffa
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Sara Fazi
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Arianna Bei
- Medical School, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Martino Tony Miele
- Department of Experimental Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Lucia Piredda
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Vittorio Manzari
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy; CIMER, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
18
|
Mitochondrial oxidative phosphorylation is linked to T-cell exhaustion. Aging (Albany NY) 2020; 12:16665-16666. [PMID: 32897246 PMCID: PMC7521505 DOI: 10.18632/aging.103995] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/17/2020] [Indexed: 01/24/2023]
|
19
|
Zhao Y, Hu W, Chen P, Cao M, Zhang Y, Zeng C, Hara H, Cooper DKC, Mou L, Luan S, Gao H. Immunosuppressive and metabolic agents that influence allo‐ and xenograft survival by in vivo expansion of T regulatory cells. Xenotransplantation 2020; 27:e12640. [PMID: 32892428 DOI: 10.1111/xen.12640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/06/2020] [Accepted: 08/17/2020] [Indexed: 12/23/2022]
Affiliation(s)
- Yanli Zhao
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center Institute of Translational Medicine Shenzhen University Health Science Center, Shenzhen University School of Medicine First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s Hospital Shenzhen China
- Department of Medical Laboratory Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | | | - Pengfei Chen
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
- Department of Medical Laboratory Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | - Mengtao Cao
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
- Department of Medical Laboratory Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | - Yingwei Zhang
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | - Changchun Zeng
- Department of Medical Laboratory Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | - Hidetaka Hara
- Xenotransplantation Program Department of Surgery University of Alabama at Birmingham Birmingham AL USA
| | - David K. C. Cooper
- Xenotransplantation Program Department of Surgery University of Alabama at Birmingham Birmingham AL USA
| | - Lisha Mou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center Institute of Translational Medicine Shenzhen University Health Science Center, Shenzhen University School of Medicine First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s Hospital Shenzhen China
| | - Shaodong Luan
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | - Hanchao Gao
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center Institute of Translational Medicine Shenzhen University Health Science Center, Shenzhen University School of Medicine First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s Hospital Shenzhen China
- Department of Medical Laboratory Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| |
Collapse
|
20
|
Li R, Zhang J, Pan S, Yuan Y, Qi H, Shu H, Hu Y, Ren L, Jiang Y, Yuan S. HMGB1 aggravates lipopolysaccharide-induced acute lung injury through suppressing the activity and function of Tregs. Cell Immunol 2020; 356:104192. [PMID: 32853967 DOI: 10.1016/j.cellimm.2020.104192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND CD4+CD25+FoxP3+ T helper cells (Tregs), a subgroup of CD4+ T helper cells, are critical effectors that protect against acute lung injury (ALI) by contact-dependent suppression or releasing anti-inflammatory cytokines including interleukin-10 (IL-10), and transforming growth factor (TGF-β). HMGB1 (High mobility group box 1 protein) was identified as a nuclear non-histone DNA-binding chromosomal protein, which participates in the regulation of lung inflammatory response and pathological processes in ALI. Previous studies have suggested that Tregs overexpresses the HMGB1-recognizing receptor. However, the interaction of HMGB1 with Tregs in ALI is still unclear. OBJECTIVE To investigate whether HMGB1 aggravates ALI by suppressing immunosuppressive function of Tregs. METHODS Anti-HMGB1 antibody and recombinant mouse HMGB1 (rHMGB1) were administered in lipopolysaccharide (LPS)-induced ALI mice and polarized LPS-primed Tregs in vitro. The Tregs pre-stimulated with or without rHMGB1 were adoptively transferred to ALI mice and depleted by Diphtheria toxin (DT). For coculture experiment, isolated Tregs were first pre-stimulated with or without rHMGB1 or anti-HMGB1 antibody, then they were cocultured with bone marrow-derived macrophages (BMMs) under LPS stimulation. RESULTS Tregs protected against acute lung pathological injury. HMGB1 modulated the suppressive function of Tregs as follows: reduction in the number of the cells and the activity of Tregs, the secretion of anti-inflammatory cytokines (IL-10, TGF-β) from Tregs, the production of IL-2 from CD4+ T cells and CD11c+ DCs, and the M2 polarization of macrophages, as well as inducing proinflammatory response of macrophages. CONCLUSIONS HMGB1 could aggravate LPS induced-ALI through suppressing the activity and function of Tregs.
Collapse
Affiliation(s)
- Ruiting Li
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Jiancheng Zhang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Shangwen Pan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Yin Yuan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Hong Qi
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Huaqing Shu
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Yingying Hu
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Lehao Ren
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Yongxiang Jiang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Shiying Yuan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| |
Collapse
|
21
|
Arroyo Hornero R, Hamad I, Côrte-Real B, Kleinewietfeld M. The Impact of Dietary Components on Regulatory T Cells and Disease. Front Immunol 2020; 11:253. [PMID: 32153577 PMCID: PMC7047770 DOI: 10.3389/fimmu.2020.00253] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
The rise in the prevalence of autoimmune diseases in developed societies has been associated with a change in lifestyle patterns. Among other factors, increased consumption of certain dietary components, such as table salt and fatty acids and excessive caloric intake has been associated with defective immunological tolerance. Dietary nutrients have shown to modulate the immune response by a direct effect on the function of immune cells or, indirectly, by acting on the microbiome of the gastrointestinal tract. FOXP3+ regulatory T cells (Tregs) suppress immune responses and are critical for maintaining peripheral tolerance and immune homeostasis, modulating chronic tissue inflammation and autoimmune disease. It is now well-recognized that Tregs show certain degree of plasticity and can gain effector functions to adapt their regulatory function to different physiological situations during an immune response. However, plasticity of Tregs might also result in conversion into effector T cells that may contribute to autoimmune pathogenesis. Yet, which environmental cues regulate Treg plasticity and function is currently poorly understood, but it is of significant importance for therapeutic purposes. Here we review the current understanding on the effect of certain dietary nutrients that characterize Western diets in Treg metabolism, stability, and function. Moreover, we will discuss the role of Tregs linking diet and autoimmunity and the potential of dietary-based interventions to modulate Treg function in disease.
Collapse
Affiliation(s)
- Rebeca Arroyo Hornero
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Ibrahim Hamad
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Beatriz Côrte-Real
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| |
Collapse
|
22
|
Peters C, Kouakanou L, Kabelitz D. A comparative view on vitamin C effects on αβ- versus γδ T-cell activation and differentiation. J Leukoc Biol 2020; 107:1009-1022. [PMID: 32034803 DOI: 10.1002/jlb.1mr1219-245r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/06/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022] Open
Abstract
Vitamin C (VitC) is an essential vitamin that needs to be provided through exogenous sources. It is a potent anti-oxidant, and an essential cofactor for many enzymes including a group of enzymes that modulate epigenetic regulation of gene expression. Moreover, VitC has a significant influence on T-cell differentiation, and can directly interfere with T-cell signaling. Conventional CD4 and CD8 T cells express the αβ TCR and recognize peptide antigens in the context of MHC presentation. The numerically small population of γδ T cells recognizes antigens in an MHC-independent manner. γδ T cells kill a broad variety of malignant cells, and because of their unique features, are interesting candidates for cancer immunotherapy. In this review, we summarize what is known about the influence of VitC on T-cell activation and differentiation with a special focus on γδ T cells. The known mechanisms of action of VitC on αβ T cells are discussed and extrapolated to the effects observed on γδ T-cell activation and differentiation. Overall, VitC enhances proliferation and effector functions of γδ T cells and thus may help to increase the efficacy of γδ T cells applied as cancer immunotherapy in adoptive cell transfer.
Collapse
Affiliation(s)
- Christian Peters
- Institute of Immunology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Léonce Kouakanou
- Institute of Immunology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
23
|
Iamsawat S, Tian L, Daenthanasanmak A, Wu Y, Nguyen HD, Bastian D, Yu XZ. Vitamin C stabilizes CD8+ iTregs and enhances their therapeutic potential in controlling murine GVHD and leukemia relapse. Blood Adv 2019; 3:4187-4201. [PMID: 31856270 PMCID: PMC6929397 DOI: 10.1182/bloodadvances.2019000531] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023] Open
Abstract
Adoptive transfer of induced regulatory T cells (iTregs) can ameliorate graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (allo-HCT). CD4+ iTregs can effectively prevent GVHD but impair the graft-versus-leukemia (GVL) effect, whereas CD8+ iTregs preserve the GVL effect but have limited efficacy in GVHD control because of their instability under inflammatory conditions. Thus, we aimed to stabilize CD8+ iTregs via treatment with vitamin C (Vit C) to improve their efficacy in controlling GVHD. We found that addition of Vit C significantly improved the stability of forkhead box P3 (Foxp3) expression in CD8+ iTregs. Moreover, Vit C-treated CD8+ iTregs exhibited high efficacy in attenuating acute and chronic GVHD. The mechanistic study revealed that addition of Vit C to CD8+ iTreg culture markedly increased DNA demethylation in the conserved noncoding sequence 2 region and, hence, maintained higher Foxp3 expression levels compared with untreated controls. In acute GVHD, Vit C-treated CD8+ iTregs were able to inhibit pathogenic T-cell expansion and differentiation while reducing thymus damage and B-cell activation in cGVHD. Importantly, in contrast to CD4+ iTregs, Vit C-treated CD8+ iTregs retained the ability to control tumor relapse. These results provide a strong rationale to use Vit C in the clinic to stabilize CD8+ iTregs for the control of GVHD and preservation of GVL after allo-HCT.
Collapse
Affiliation(s)
| | - Linlu Tian
- Department of Microbiology and Immunology and
| | | | - Yongxia Wu
- Department of Microbiology and Immunology and
| | | | | | - Xue-Zhong Yu
- Department of Microbiology and Immunology and
- Department of Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
24
|
Nutritional Modulation of Immune and Central Nervous System Homeostasis: The Role of Diet in Development of Neuroinflammation and Neurological Disease. Nutrients 2019; 11:nu11051076. [PMID: 31096592 PMCID: PMC6566411 DOI: 10.3390/nu11051076] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/02/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
The gut-microbiome-brain axis is now recognized as an essential part in the regulation of systemic metabolism and homeostasis. Accumulating evidence has demonstrated that dietary patterns can influence the development of metabolic alterations and inflammation through the effects of nutrients on a multitude of variables, including microbiome composition, release of microbial products, gastrointestinal signaling molecules, and neurotransmitters. These signaling molecules are, in turn, implicated in the regulation of the immune system, either promoting or inhibiting the production of pro-inflammatory cytokines and the expansion of specific leukocyte subpopulations, such as Th17 and Treg cells, which are relevant in the development of neuroinflammatory and neurodegenerative conditions. Metabolic diseases, like obesity and type 2 diabetes mellitus, are related to inadequate dietary patterns and promote variations in the aforementioned signaling pathways in patients with these conditions, which have been linked to alterations in neurological functions and mental health. Thus, maintenance of adequate dietary patterns should be an essential component of any strategy aiming to prevent neurological pathologies derived from systemic metabolic alterations. The present review summarizes current knowledge on the role of nutrition in the modulation of the immune system and its impact in the development of neuroinflammation and neurological disease.
Collapse
|
25
|
Wu JS, Li L, Wang SS, Pang X, Wu JB, Sheng SR, Tang YJ, Tang YL, Zheng M, Liang XH. Autophagy is positively associated with the accumulation of myeloid‑derived suppressor cells in 4‑nitroquinoline‑1‑oxide‑induced oral cancer. Oncol Rep 2018; 40:3381-3391. [PMID: 30272335 PMCID: PMC6196587 DOI: 10.3892/or.2018.6747] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/13/2018] [Indexed: 02/05/2023] Open
Abstract
It has previously been demonstrated that autophagy and inflammation act synergistically to promote carcinogenesis. However, the precise roles of autophagy in multistep oral carcinogenesis are still unclear, particularly regarding its association with tumor inflammation. The present study established a 4NQO-induced oral cancer mouse model and investigated autophagy status in the multistep process of oral carcinogenesis using immunohistochemistry, western blotting and immunofluorescence staining. Furthermore, the number of Gr-1+CD11b+ myeloid derived suppressor cells (MDSCs) and CD4+ Foxp3+ regulatory T cells (Tregs) during oral carcinogenesis and the association with autophagy status was also examined. The results revealed that the expression of autophagy biomarkers, including dihydrosphingosine 1-phosphate phosphatase LCB3 (LC3B), p62/SQSTM1 (p62) and Beclin 1 increased during 4NQO-induced carcinogenesis and in human oral cancer. The number of MDSCs and Tregs also increased during oral carcinogenesis. Furthermore, the expression of LC3B and p62 significantly correlated with the accumulation of MDSCs and the expression of Beclin 1 correlated with the increase of Tregs. These data indicated that autophagy may be activated by the tumor inflammation microenvironment during oral carcinogenesis.
Collapse
Affiliation(s)
- Jia-Shun Wu
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Li
- Department of Stomatology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang 316021, P.R. China
| | - Sha-Sha Wang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xin Pang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jing-Biao Wu
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Su-Rui Sheng
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ya-Jie Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei 430068, P.R. China
| | - Ya-Ling Tang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Min Zheng
- Department of Stomatology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang 316021, P.R. China
| | - Xin-Hua Liang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|