1
|
Hansen B, Sánchez-Castro M, Schintgen L, Khakdan A, Schneider JG, Wilmes P. The impact of fasting and caloric restriction on rheumatoid arthritis in humans: A narrative review. Clin Nutr 2025; 49:178-186. [PMID: 40328175 DOI: 10.1016/j.clnu.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease affecting approximately 1 % of the global population. It is characterized by swollen and painful joints eventually evolving into bone erosion, cartilage degradation and systemic inflammation, that significantly reduce patients' quality of life. While modern pharmacological treatments often lead to symptom improvement, they are also accompanied by substantial side effects, which can further impair patient wellbeing. Dietary interventions, particularly fasting and caloric restriction (CR), have gained increasing attention as adjunctive strategies for RA prevention and treatment. Their anti-inflammatory potential and ability to modulate the gut microbiome render them an attractive option to accompany or modify medical treatment. However, high-quality research on fasting and CR interventions in humans with RA remains limited, and the underlying mechanisms are not yet fully understood. The present narrative review reflects our current knowledge regarding fasting and CR, emphasising their impact on clinical outcomes, potential underlying mechanism and the sustainability of their effects. Evidence suggests that fasting and CR may lead to short-term improvements in RA disease activity, including reductions in inflammatory markers such as C-reactive protein (CRP) and interleukin-6 (IL-6). However, their long-term efficacy remains uncertain due to the limited duration of most studies. Future research should focus on identifying optimal fasting and CR protocols and their feasibility in long-term disease management, along with investigating patient adherence and potential risks associated with fasting interventions.
Collapse
Affiliation(s)
- Bérénice Hansen
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marta Sánchez-Castro
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Lynn Schintgen
- Department of Microbiome Research and Applied Bioinformatics, Institute of Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Arefeh Khakdan
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jochen G Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg; Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg; Department of Internal Medicine II, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany.
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg; Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
2
|
Qiu X, Wen R, Wu F, Mao J, Azad T, Wang Y, Zhu J, Zhou X, Xie H, Hong K, Li B, Zhang L, Wen C. The role of double-negative B cells in the pathogenesis of systemic lupus erythematosus. Autoimmun Rev 2025; 24:103821. [PMID: 40274006 DOI: 10.1016/j.autrev.2025.103821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
B cells are essential to the pathophysiology of systemic lupus erythematosus (SLE), a chronic autoimmune illness. IgD-CD27-double negative B cells (DNB cells) are one of the aberrant B cell subsets linked to SLE that have attracted much scientific interest. There is growing evidence that DNB cells play a significant role in the development of the disease and are strongly linked to the activity of lupus. These cells play a pivotal role in the pathogenesis of SLE by producing a diverse array of autoantibodies, which form immune complexes that drive target organ damage. A comprehensive understanding of SLE pathophysiology necessitates in-depth investigation into DNB cells, not only to elucidate their mechanistic contributions but also to uncover novel therapeutic strategies. According to available data, treatments that target B cells have proven effective in managing SLE; nevertheless, a significant breakthrough in precision medicine for SLE may come from targeting DNB cells specifically. Despite growing interest in DNB cells, their precise characteristics, developmental trajectories, and regulatory mechanisms remain incompletely defined, posing significant challenges to the field. A comprehensive investigation of the regulatory mechanisms governing DNB cell differentiation and expansion in SLE may facilitate novel therapeutic discoveries. This review aims to provide an updated synthesis of current research on DNB cells, with a focus on their origins, developmental trajectories in SLE, and potential as precision therapeutic targets.
Collapse
Affiliation(s)
- Xinying Qiu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China; The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, Changsha 410007, Hunan, China
| | - RuiFan Wen
- Medical School, Hunan University of Chinese Medicine, No.300 Xueshi Road, Hanpu Science & Education District, Changsha, Hunan 410208, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Tasnim Azad
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yang Wang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Junquan Zhu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Xin Zhou
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Haotian Xie
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Kimsor Hong
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Binbin Li
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Liang Zhang
- The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, Changsha 410007, Hunan, China; Department of Nephrology, Rheumatology and Immunology, Hunan Children's Hospital, Changsha 410007, Hunan, China.
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| |
Collapse
|
3
|
Perugino CA, Liu H, Feldman J, Marbourg J, Guy TV, Hui A, Ingram N, Liebaert J, Chaudhary N, Tao W, Jacob-Dolan C, Hauser BM, Mian Z, Nathan A, Zhao Z, Kaseke C, Tano-Menka R, Getz MA, Senjobe F, Berrios C, Ofoman O, Manickas-Hill Z, Wesemann DR, Lemieux JE, Goldberg MB, Nündel K, Moormann A, Marshak-Rothstein A, Larocque RC, Ryan ET, Iafrate JA, Lingwood D, Gaiha G, Charles R, Balazs AB, Pandit A, Naranbhai V, Schmidt AG, Pillai S. Two distinct durable human class-switched memory B cell populations are induced by vaccination and infection. Cell Rep 2025; 44:115472. [PMID: 40173042 DOI: 10.1016/j.celrep.2025.115472] [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: 10/08/2024] [Revised: 01/21/2025] [Accepted: 03/05/2025] [Indexed: 04/04/2025] Open
Abstract
Memory lymphocytes are durable cells that persist in the absence of antigen, but few human B cell subsets have been characterized in terms of durability. The relative durability of eight non-overlapping human B cell sub-populations covering 100% of all human class-switched B cells was interrogated. Only two long-lived B cell populations persisted in the relative absence of antigen. In addition to canonical germinal center-derived switched-memory B cells with an IgD-CD27+CXCR5+ phenotype, a second, non-canonical, but distinct memory population of IgD-CD27-CXCR5+ DN1 B cells was also durable, exhibited a unique TP63-linked transcriptional and anti-apoptotic signature, had low levels of somatic hypermutation, but was more clonally expanded than canonical switched-memory B cells. DN1 B cells likely evolved to preserve immunological breadth and may represent the human counterparts of rodent extrafollicular memory B cells that, unlike canonical memory B cells, can enter germinal centers and facilitate B cell and antibody evolution.
Collapse
Affiliation(s)
- Cory A Perugino
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Division of Rheumatology Allergy and Immunology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hang Liu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Jared Feldman
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Jess Marbourg
- Abbvie Cambridge Research Center, Cambridge, MA 02139, USA
| | - Thomas V Guy
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Anson Hui
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Nicole Ingram
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Julian Liebaert
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Neha Chaudhary
- Abbvie Cambridge Research Center, Cambridge, MA 02139, USA
| | - Weiyang Tao
- Abbvie Cambridge Research Center, Cambridge, MA 02139, USA
| | - Catherine Jacob-Dolan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Monash University, Melbourne, VIC 3800, Australia
| | - Blake M Hauser
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Zayd Mian
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Anusha Nathan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Zezhou Zhao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Clarety Kaseke
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Rhoda Tano-Menka
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Matthew A Getz
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Fernando Senjobe
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Cristhian Berrios
- Department of Pathology, Massachusetts Hospital, Boston, MA 02114, USA
| | - Onosereme Ofoman
- Department of Pathology, Massachusetts Hospital, Boston, MA 02114, USA
| | | | - Duane R Wesemann
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jacob E Lemieux
- Infectious Diseases Division, Massachusetts Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Marcia B Goldberg
- Infectious Diseases Division, Massachusetts Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Kerstin Nündel
- University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ann Moormann
- University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | | - Regina C Larocque
- Infectious Diseases Division, Massachusetts Hospital, Boston, MA 02114, USA
| | - Edward T Ryan
- Infectious Diseases Division, Massachusetts Hospital, Boston, MA 02114, USA
| | - John A Iafrate
- MGH Cancer Center, Massachusetts Hospital, Boston, MA 02114, USA
| | - Daniel Lingwood
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Gaurav Gaiha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Gastrointestinal Unit, Massachusetts Hospital, Boston, MA 02114, USA
| | - Richelle Charles
- Infectious Diseases Division, Massachusetts Hospital, Boston, MA 02114, USA
| | | | | | - Vivek Naranbhai
- MGH Cancer Center, Massachusetts Hospital, Boston, MA 02114, USA; Monash University, Melbourne, VIC 3800, Australia; Center for the AIDS Programme of Research in South Africa, Durban 4001, South Africa
| | - Aaaron G Schmidt
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
4
|
Martinis E, Tonon S, Colamatteo A, La Cava A, Matarese G, Pucillo CEM. B cell immunometabolism in health and disease. Nat Immunol 2025; 26:366-377. [PMID: 39984733 DOI: 10.1038/s41590-025-02102-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 01/15/2025] [Indexed: 02/23/2025]
Abstract
B cells have crucial roles in the initiation and progression of many pathological conditions, and several therapeutic strategies have targeted the function of these cells. The advent of immunometabolism has provided compelling evidence that the metabolic reprogramming of immune cells can dramatically alter physiopathological immune activities. A better knowledge of the metabolic profiles of B cells can provide valuable means for developing therapies tuning defined cell pathways. Here we review the cellular and molecular mechanisms by which immunometabolism controls the physiology and pathophysiology of B cells and discuss the experimental evidence linking B cell metabolism to health, autoimmunity, and cancer. Considering that several metabolic pathways in B cells are involved differently, or even in opposite ways, in health and disease, we discuss how targeted modulation of B cell immunometabolism could be exploited mechanistically to rebalance abnormal B cell functions that have become altered in disease states.
Collapse
Affiliation(s)
| | - Silvia Tonon
- Department of Medicine, University of Udine, Udine, Italy
| | - Alessandra Colamatteo
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Napoli, Italy
| | - Antonio La Cava
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Napoli, Italy
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Giuseppe Matarese
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Napoli, Italy.
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore' - Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy.
| | | |
Collapse
|
5
|
Yee TM, Wang LW. Metabolic Reprogramming in Epstein-Barr Virus Associated Diseases. J Med Virol 2025; 97:e70197. [PMID: 39895469 DOI: 10.1002/jmv.70197] [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: 12/12/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025]
Abstract
Epstein-Barr virus (EBV) is the first human cancer-causing viral pathogen to be discovered; it has been epidemiologically associated with a wide range of diseases, including cancers, autoimmunity, and hyperinflammatory disorders. Its evolutionary success is underpinned by coordinated expression of viral transcription factors (EBV nuclear antigens), signaling proteins (EBV latent membrane proteins), and noncoding RNAs, which orchestrate cell transformation, immune evasion, and dissemination. Each of those activities entails significant metabolic rewiring, which is achieved by viral subversion of key host metabolic regulators such as the mammalian target of rapamycin (mTOR), MYC, and hypoxia-inducible factor (HIF). In this review, we systemically discuss how EBV-encoded factors regulate metabolism to achieve viral persistence and propagation, as well as potential research questions and directions in EBV-driven metabolism.
Collapse
Affiliation(s)
- Tiffany Melanie Yee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Liang Wei Wang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| |
Collapse
|
6
|
Ueno M, Iwata S, Yamagata K, Todoroki Y, Sonomoto K, Nagayasu A, Miyagawa I, Kubo S, Miyazaki Y, Miyata H, Kanda R, Aritomi T, Nakayamada S, Tanaka Y. Induction of interleukin 21 receptor expression via enhanced intracellular metabolism in B cells and its relevance to the disease activity in systemic lupus erythematosus. RMD Open 2024; 10:e004567. [PMID: 39740932 PMCID: PMC11749818 DOI: 10.1136/rmdopen-2024-004567] [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: 05/25/2024] [Accepted: 11/26/2024] [Indexed: 01/02/2025] Open
Abstract
OBJECTIVE To elucidate the association between the changes in intracellular metabolism in the early stage of B cell activation and systemic lupus erythematosus (SLE) pathogenesis. METHODS CD19+ or CD19+CD27- (naïve) cells from the peripheral blood of healthy controls and lupus patients were cultured under different stimuli. The changes in intracellular metabolism and signalling pathways in these cells were evaluated. RESULTS Stimulation with CpG (Toll-like receptor 9 (TLR9) ligand) in vitro induced enhanced interleukin 21 (IL-21) receptor expression in CD19+CD27- cells after 24 hours. The addition of IL-21 to the CpG stimulation enhanced the extracellular acidification rate, which indicates glycolysis, within 30 min. IL-21 receptor (IL-21R) expression induced by CpG stimulation was selectively inhibited by 2-deoxy-D-glucose (hexokinase 2 (HK2) inhibitor) and heptelidic acid (glyceraldehyde 3-phosphate dehydrogenase (GAPDH) inhibitor). RNA immunoprecipitation with anti-GAPDH antibody revealed that CpG stimulation dissociated the binding between IL-21R messenger RNA (mRNA) and GAPDH under no stimulation. HK2 and GAPDH expression were higher in CD19+CD27- cells of lupus patients than in those of healthy controls, and GAPDH expression was correlated with the plasmocyte count and disease activity score. CONCLUSION IL-21R mRNA-GAPDH binding dissociation associated with rapid glycolytic enhancement by the TLR9 ligand in B cells may induce plasmocyte differentiation through IL-21 signals and be involved in exacerbating SLE.
Collapse
Affiliation(s)
- Masanobu Ueno
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Shigeru Iwata
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
- Department of Rheumatology and Clinical Immunology, Wakayama Medical University, Wakayama, Japan
| | - Kaoru Yamagata
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Yasuyuki Todoroki
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
- Department of Molecular Targeted Therapies, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Koshiro Sonomoto
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
- Department of Clinical Nursing, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Atsushi Nagayasu
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Ippei Miyagawa
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Satoshi Kubo
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
- Department of Molecular Targeted Therapies, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Yusuke Miyazaki
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Hiroko Miyata
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Ryuichiro Kanda
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Takafumi Aritomi
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| |
Collapse
|
7
|
Martin J, Cheng Q, Laurent SA, Thaler FS, Beenken AE, Meinl E, Krönke G, Hiepe F, Alexander T. B-Cell Maturation Antigen (BCMA) as a Biomarker and Potential Treatment Target in Systemic Lupus Erythematosus. Int J Mol Sci 2024; 25:10845. [PMID: 39409173 PMCID: PMC11476889 DOI: 10.3390/ijms251910845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
The BAFF-APRIL system is crucial for the pathogenesis of systemic lupus erythematosus (SLE) by promoting B cell survival, differentiation and the maintenance of humoral autoimmunity. Here, we investigated the relationship of BCMA expression on B cell subsets with its ligands BAFF and APRIL, together with soluble BCMA, and with clinical and serologic variables in a cohort of 100 SLE patients (86 under conventional and 14 under belimumab therapy) and 30 healthy controls (HCs) using multicolor flow cytometry and ELISA. We found that BCMA expression in SLE patients was significantly increased on all B cell subsets compared to HCs, with all examined components of the BAFF-APRIL system being upregulated. BCMA expression was significantly increased on switched and unswitched memory B cells compared to naïve B cells, both in HCs and SLE. BCMA expression on B cells correlated with plasmablast frequencies, serum anti-dsDNA antibodies and complement consumption, while soluble BCMA correlated with plasmablast frequency, highlighting its potential as a clinical biomarker. Belimumab treatment significantly reduced BCMA expression on most B cell subsets and soluble TACI and contributed to the inhibition of almost the entire BAFF-APRIL system and restoration of B cell homeostasis. These results provide insights into the complex dysregulation of the BAFF-APRIL system in SLE and highlight the therapeutic potential of targeting its components, particularly BCMA, in addition to its use as a biomarker for disease activity.
Collapse
MESH Headings
- Humans
- Lupus Erythematosus, Systemic/drug therapy
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Lupus Erythematosus, Systemic/blood
- B-Cell Maturation Antigen/metabolism
- B-Cell Maturation Antigen/immunology
- Biomarkers/blood
- Female
- Adult
- Male
- B-Cell Activating Factor/blood
- B-Cell Activating Factor/metabolism
- Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
- Tumor Necrosis Factor Ligand Superfamily Member 13/blood
- Middle Aged
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Antinuclear/blood
- Antibodies, Antinuclear/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- B-Lymphocytes/drug effects
- B-Lymphocyte Subsets/metabolism
- B-Lymphocyte Subsets/immunology
- Case-Control Studies
Collapse
Affiliation(s)
- Jonas Martin
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (J.M.); (Q.C.); (A.E.B.); (G.K.); (F.H.)
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Qingyu Cheng
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (J.M.); (Q.C.); (A.E.B.); (G.K.); (F.H.)
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Sarah A. Laurent
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 80336 Munich, Germany (F.S.T.); (E.M.)
| | - Franziska S. Thaler
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 80336 Munich, Germany (F.S.T.); (E.M.)
- Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Martinsried, Germany
| | - Anne Elisabeth Beenken
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (J.M.); (Q.C.); (A.E.B.); (G.K.); (F.H.)
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 80336 Munich, Germany (F.S.T.); (E.M.)
- Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Martinsried, Germany
| | - Gerhard Krönke
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (J.M.); (Q.C.); (A.E.B.); (G.K.); (F.H.)
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Falk Hiepe
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (J.M.); (Q.C.); (A.E.B.); (G.K.); (F.H.)
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (J.M.); (Q.C.); (A.E.B.); (G.K.); (F.H.)
- Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, 10117 Berlin, Germany
| |
Collapse
|
8
|
Abir AH, Weckwerth L, Wilhelm A, Thomas J, Reichardt CM, Munoz L, Völkl S, Appelt U, Mroz M, Niesner R, Hauser A, Sophie Fischer R, Pracht K, Jäck HM, Schett G, Krönke G, Mielenz D. Metabolic profiling of single cells by exploiting NADH and FAD fluorescence via flow cytometry. Mol Metab 2024; 87:101981. [PMID: 38971403 PMCID: PMC11300934 DOI: 10.1016/j.molmet.2024.101981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/08/2024] Open
Abstract
OBJECTIVE The metabolism of different cells within the same microenvironment can differ and dictate physiological or pathological adaptions. Current single-cell analysis methods of metabolism are not label-free. METHODS The study introduces a label-free, live-cell analysis method assessing endogenous fluorescence of NAD(P)H and FAD in surface-stained cells by flow cytometry. RESULTS OxPhos inhibition, mitochondrial uncoupling, glucose exposure, genetic inactivation of glucose uptake and mitochondrial respiration alter the optical redox ratios of FAD and NAD(P)H as measured by flow cytometry. Those alterations correlate strongly with measurements obtained by extracellular flux analysis. Consequently, metabolically distinct live B-cell populations can be resolved, showing that human memory B-cells from peripheral blood exhibit a higher glycolytic flexibility than naïve B cells. Moreover, the comparison of blood-derived B- and T-lymphocytes from healthy donors and rheumatoid arthritis patients unleashes rheumatoid arthritis-associated metabolic traits in human naïve and memory B-lymphocytes. CONCLUSIONS Taken together, these data show that the optical redox ratio can depict metabolic differences in distinct cell populations by flow cytometry.
Collapse
Affiliation(s)
- Ariful Haque Abir
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Leonie Weckwerth
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Artur Wilhelm
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jana Thomas
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Clara M Reichardt
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Luis Munoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
| | - Uwe Appelt
- Flow cytometry core unit, Friedrich-Alexander-Universität Erlangen-Nürnberg, Glückstr. 6, 91054 Erlangen, Germany
| | - Markus Mroz
- Flow cytometry core unit, Friedrich-Alexander-Universität Erlangen-Nürnberg, Glückstr. 6, 91054 Erlangen, Germany
| | - Raluca Niesner
- Deutsches Rheumaforschungszentrum Berlin, Biophysikalische Analytik, Charitéplatz 1, 10117 Berlin, Germany; Freie Universität Berlin, Dynamisches und funktionelles in vivo Imaging, Adresse: Oertzenweg 19b, 14163 Berlin, Germany
| | - Anja Hauser
- Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Deutsches Rheumaforschungszentrum Berlin, Immundynamik, Charitéplatz 1, 10117 Berlin, Germany
| | - Rebecca Sophie Fischer
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Katharina Pracht
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany.
| |
Collapse
|
9
|
Noble J, Cabezas L, Truffot A, Dumolard L, Jouve T, Malvezzi P, Rostaing L, Dard C, Saas P, Cravedi P, Macek-Jilkova Z. Glycolysis Changes in Alloreactive Memory B Cells in Highly Sensitized Kidney Transplant Recipients Undergoing Desensitization Therapy. Transpl Int 2024; 37:13029. [PMID: 39081904 PMCID: PMC11287219 DOI: 10.3389/ti.2024.13029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024]
Abstract
Despite the growing use of desensitization strategies, hyperimmune patients remain at high risk of antibody-mediated rejection suggesting that, even when donor-specific antibodies (DSA) are effectively depleted, anti-donor specific B cells persist. We included 10 highly sensitized recipients that underwent desensitization with plasmapheresis and B cell depletion prior to kidney transplantation. We quantified changes in DSA (luminex), total B-cell subsets (flow cytometry), anti-donor HLA B cells (fluorospot), and single-cell metabolism in serially collected samples before desensitization, at the time of transplant, and at 6 and 12 months thereafter. Desensitization was associated with a decrease in DSA and total memory B cell and naive B cell percentage, while plasma cells and memory anti-donor HLA circulating B cells persisted up to 12 months after transplant. At 12-month post-transplantation, memory B cells increased their glycolytic capacity, while proliferative KI67+ plasma cells modified their metabolism by increasing fatty acid and amino acid oxidation capacity and decreasing their glucose dependence. Despite effective DSA depletion, anti-donor B cells persist in kidney transplant recipients. Due to the reliance of these cells on glycolysis, glycolysis-targeting therapies might represent a valuable treatment strategy.
Collapse
Affiliation(s)
- Johan Noble
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lara Cabezas
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Aurelie Truffot
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Virology Department, University Hospital Grenoble, Grenoble, France
| | - Lucile Dumolard
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Thomas Jouve
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Paolo Malvezzi
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
| | - Lionel Rostaing
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Céline Dard
- EFS, Recherche et Développement, Grenoble, France
| | - Philippe Saas
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
- EFS, Recherche et Développement, Grenoble, France
| | - Paolo Cravedi
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zuzana Macek-Jilkova
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
| |
Collapse
|
10
|
Wang S, Yang N, Zhang H. Metabolic dysregulation of lymphocytes in autoimmune diseases. Trends Endocrinol Metab 2024; 35:624-637. [PMID: 38355391 DOI: 10.1016/j.tem.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 02/16/2024]
Abstract
Lymphocytes are crucial for protective immunity against infection and cancers; however, immune dysregulation can lead to autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Metabolic adaptation controls lymphocyte fate; thus, metabolic reprogramming can contribute to the pathogenesis of autoimmune diseases. Here, we summarize recent advances on how metabolic reprogramming determines the autoreactive and proinflammatory nature of lymphocytes in SLE and RA, unraveling molecular mechanisms and providing therapeutic targets for human autoimmune diseases.
Collapse
Affiliation(s)
- Shuyi Wang
- Department of Rheumatology and Clinical Immunology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Niansheng Yang
- Department of Rheumatology and Clinical Immunology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Hui Zhang
- Department of Rheumatology and Clinical Immunology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
11
|
Khameneh SC, Razi S, Lashanizadegan R, Akbari S, Sayaf M, Haghani K, Bakhtiyari S. MicroRNA-mediated metabolic regulation of immune cells in cancer: an updated review. Front Immunol 2024; 15:1424909. [PMID: 39007129 PMCID: PMC11239499 DOI: 10.3389/fimmu.2024.1424909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/12/2024] [Indexed: 07/16/2024] Open
Abstract
The study of immunometabolism, which examines how immune cells regulate their metabolism to maintain optimal performance, has become an important area of focus in cancer immunology. Recent advancements in this field have highlighted the intricate connection between metabolism and immune cell function, emphasizing the need for further research. MicroRNAs (miRNAs) have gained attention for their ability to post-transcriptionally regulate gene expression and impact various biological processes, including immune function and cancer progression. While the role of miRNAs in immunometabolism is still being explored, recent studies have demonstrated their significant influence on the metabolic activity of immune cells, such as macrophages, T cells, B cells, and dendritic cells, particularly in cancer contexts. Disrupted immune cell metabolism is a hallmark of cancer progression, and miRNAs have been linked to this process. Understanding the precise impact of miRNAs on immune cell metabolism in cancer is essential for the development of immunotherapeutic approaches. Targeting miRNAs may hold potential for creating groundbreaking cancer immunotherapies to reshape the tumor environment and improve treatment outcomes. In summary, the recognition of miRNAs as key regulators of immune cell metabolism across various cancers offers promising potential for refining cancer immunotherapies. Further investigation into how miRNAs affect immune cell metabolism could identify novel therapeutic targets and lead to the development of innovative cancer immunotherapies.
Collapse
Affiliation(s)
| | - Sara Razi
- Vira Ideators of Modern Science, Tehran, Iran
- Vira Pioneers of Modern Science (VIPOMS), Tehran, Iran
| | | | | | - Masoud Sayaf
- Department of Cellular and Molecular Biology, Faculty of Basic Sciences, Azad University Central Tehran Branch, Tehran, Iran
| | - Karimeh Haghani
- Department of Clinical Biochemistry, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Salar Bakhtiyari
- Department of Clinical Biochemistry, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, United States
| |
Collapse
|
12
|
Fouza A, Fylaktou A, Tagkouta A, Daoudaki M, Vagiotas L, Kasimatis E, Stangou M, Xochelli A, Nikolaidou V, Katsanos G, Tsoulfas G, Skoura L, Papagianni A, Antoniadis N. Evaluation of Regulatory B Cell Subpopulations CD24++CD38++, CD24++CD27+, Plasmablasts and Their Correlation with T Regs CD3+CD4+CD25+FOXP3+ in Dialysis Patients and Early Post-Transplant Rejection-Free Kidney Recipients. J Clin Med 2024; 13:3080. [PMID: 38892795 PMCID: PMC11173263 DOI: 10.3390/jcm13113080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Background: B and T regulatory cells, also known as Bregs and Tregs, are involved in kidney transplantation. The purpose of this study is to monitor changes in the frequency and absolute numbers of Tregs (CD3+CD4+CD25+FoxP3+), transitional Bregs (tBregs) (CD24++CD38++), memory Bregs (mBregs) (CD24++CD27+), and plasmablasts before (T0) and six months (T6) after transplantation. Additionally, we aim to investigate any correlation between Tregs and tBregs, mBregs, or plasmablasts and their relationship with graft function. Methods: Flow cytometry was used to immunophenotype cells from 50 kidney recipients who did not experience rejection. Renal function was assessed using the estimated glomerular filtration rate (eGFR). Results: At T6, there was a significant decrease in the frequency of Tregs, plasmablasts, and tBregs, as well as in the absolute number of tBregs. The frequency of mBregs, however, remained unchanged. Graft function was found to have a positive correlation with the frequency of tBregs and plasmablasts. A significant correlation was observed between the frequency and absolute number of tBregs only when the eGFR was greater than 60 but not at lower values. At an eGFR greater than 60, there was a positive correlation between the absolute numbers of Tregs and mBregs but not between Tregs and tBregs. No correlation was observed for any cell population in dialysis patients. Conclusions: The data show a correlation between the frequency and absolute number of tBregs and the absolute number of Tregs and mBregs with good renal function in the early post-transplant period.
Collapse
Affiliation(s)
- Ariadni Fouza
- Department of Transplant Surgery, Center for Research and Innovation in Solid Organ Transplantation School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, 54642 Thessaloniki, Greece; (L.V.); (G.K.); (G.T.); (N.A.)
| | - Asimina Fylaktou
- National Peripheral Histocompatibility Center, Department of Immunology, Ippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (A.X.); (V.N.)
| | - Anneta Tagkouta
- Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Department of Hygiene, Social-Preventive Medicine & Medical Statistics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Daoudaki
- Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Lampros Vagiotas
- Department of Transplant Surgery, Center for Research and Innovation in Solid Organ Transplantation School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, 54642 Thessaloniki, Greece; (L.V.); (G.K.); (G.T.); (N.A.)
| | - Efstratios Kasimatis
- 1st Department of Nephrology, School of Medicine, Aristotle University of Thessaloniki, Ippokration General Hospital, 54642 Thessaloniki, Greece; (E.K.); (M.S.); (A.P.)
| | - Maria Stangou
- 1st Department of Nephrology, School of Medicine, Aristotle University of Thessaloniki, Ippokration General Hospital, 54642 Thessaloniki, Greece; (E.K.); (M.S.); (A.P.)
| | - Aliki Xochelli
- National Peripheral Histocompatibility Center, Department of Immunology, Ippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (A.X.); (V.N.)
| | - Vasiliki Nikolaidou
- National Peripheral Histocompatibility Center, Department of Immunology, Ippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (A.X.); (V.N.)
| | - Georgios Katsanos
- Department of Transplant Surgery, Center for Research and Innovation in Solid Organ Transplantation School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, 54642 Thessaloniki, Greece; (L.V.); (G.K.); (G.T.); (N.A.)
| | - Georgios Tsoulfas
- Department of Transplant Surgery, Center for Research and Innovation in Solid Organ Transplantation School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, 54642 Thessaloniki, Greece; (L.V.); (G.K.); (G.T.); (N.A.)
| | - Lemonia Skoura
- Microbiology Laboratory, Department of Immunology, AHEPA University Hospital, 54636 Thessaloniki, Greece;
| | - Aikaterini Papagianni
- 1st Department of Nephrology, School of Medicine, Aristotle University of Thessaloniki, Ippokration General Hospital, 54642 Thessaloniki, Greece; (E.K.); (M.S.); (A.P.)
| | - Nikolaos Antoniadis
- Department of Transplant Surgery, Center for Research and Innovation in Solid Organ Transplantation School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, 54642 Thessaloniki, Greece; (L.V.); (G.K.); (G.T.); (N.A.)
| |
Collapse
|
13
|
Chung MKY, Gong L, Kwong DL, Lee VH, Lee AW, Guan X, Kam N, Dai W. Functions of double-negative B cells in autoimmune diseases, infections, and cancers. EMBO Mol Med 2023; 15:e17341. [PMID: 37272217 PMCID: PMC10493577 DOI: 10.15252/emmm.202217341] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 06/06/2023] Open
Abstract
Most mature B cells can be divided into four subtypes based on the expression of the surface markers IgD and CD27: IgD+ CD27- naïve B cells, IgD+ CD27+ unswitched memory B cells, IgD- CD27+ switched memory B cells, and IgD- CD27- double-negative (DN) B cells. Despite their small population size in normal peripheral blood, DN B cells play integral roles in various diseases. For example, they generate autoimmunity in autoimmune conditions, while these cells may generate both autoimmune and antipathogenic responses in COVID-19, or act in a purely antipathogenic capacity in malaria. Recently, DN B cells have been identified in nasopharyngeal carcinoma and non-small-cell lung cancers, where they may play an immunosuppressive role. The distinct functions that DN B cells play in different diseases suggest that they are a heterogeneous B-cell population. Therefore, further study of the mechanisms underlying the involvement of DN B cells in these diseases is essential for understanding their pathogenesis and the development of therapeutic strategies. Further research is thus warranted to characterize the DN B-cell population in detail.
Collapse
Affiliation(s)
- Michael King Yung Chung
- Department of Clinical Oncology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Lanqi Gong
- Department of Clinical Oncology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized TherapyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
| | - Dora Lai‐Wan Kwong
- Department of Clinical Oncology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized TherapyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
| | - Victor Ho‐Fun Lee
- Department of Clinical Oncology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized TherapyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
| | - Ann Wing‐Mui Lee
- Department of Clinical Oncology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized TherapyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
| | - Xin‐Yuan Guan
- Department of Clinical Oncology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized TherapyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
| | - Ngar‐Woon Kam
- Department of Clinical Oncology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Laboratory for Synthetic Chemistry and Chemical BiologyHong Kong (SAR)China
| | - Wei Dai
- Department of Clinical Oncology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized TherapyThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
| |
Collapse
|
14
|
Álvarez Gómez JA, Salazar-Camarena DC, Román-Fernández IV, Ortiz-Lazareno PC, Cruz A, Muñoz-Valle JF, Marín-Rosales M, Espinoza-García N, Sagrero-Fabela N, Palafox-Sánchez CA. BAFF system expression in double negative 2, activated naïve and activated memory B cells in systemic lupus erythematosus. Front Immunol 2023; 14:1235937. [PMID: 37675114 PMCID: PMC10478082 DOI: 10.3389/fimmu.2023.1235937] [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: 06/06/2023] [Accepted: 07/28/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction B cell activating factor (BAFF) has an important role in normal B cell development. The aberrant expression of BAFF is related with the autoimmune diseases development like Systemic Lupus Erythematosus (SLE) for promoting self-reactive B cells survival. BAFF functions are exerted through its receptors BAFF-R (BR3), transmembrane activator calcium modulator and cyclophilin ligand interactor (TACI) and B cell maturation antigen (BCMA) that are reported to have differential expression on B cells in SLE. Recently, atypical B cells that express CD11c have been associated with SLE because they are prone to develop into antibody-secreting cells, however the relationship with BAFF remains unclear. This study aims to analyze the BAFF system expression on CXCR5- CD11c+ atypical B cell subsets double negative 2 (DN2), activated naïve (aNAV), switched memory (SWM) and unswitched memory (USM) B cells. Methods Forty-five SLE patients and 15 healthy subjects (HS) were included. Flow cytometry was used to evaluate the expression of the receptors in the B cell subpopulations. Enzyme-linked immunosorbent assay (ELISA) was performed to quantify the soluble levels of BAFF (sBAFF) and IL-21. Results We found increased frequency of CXCR5- CD11c+ atypical B cell subpopulations DN2, aNAV, SWM and USM B cells in SLE patients compared to HS. SLE patients had increased expression of membrane BAFF (mBAFF) and BCMA receptor in classic B cell subsets (DN, NAV, SWM and USM). Also, the CXCR5+ CD11c- DN1, resting naïve (rNAV), SWM and USM B cell subsets showed higher mBAFF expression in SLE. CXCR5- CD11c+ atypical B cell subpopulations DN2, SWM and USM B cells showed strong correlations with the expression of BAFF receptors. The atypical B cells DN2 in SLE showed significant decreased expression of TACI, which correlated with higher IL-21 levels. Also, lower expression of TACI in atypical B cell DN2 was associated with high disease activity. Discussion These results suggest a participation of the BAFF system in CXCR5- CD11c+ atypical B cell subsets in SLE patients. Decreased TACI expression on atypical B cells DN2 correlated with high disease activity in SLE patients supporting the immunoregulatory role of TACI in autoimmunity.
Collapse
Affiliation(s)
- Jhonatan Antonio Álvarez Gómez
- Doctorado en Ciencias en Biología Molecular en Medicina (DCBMM), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Diana Celeste Salazar-Camarena
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Ilce Valeria Román-Fernández
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Pablo César Ortiz-Lazareno
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Alvaro Cruz
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - José Francisco Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Miguel Marín-Rosales
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
- Hospital General de Occidente, Secretaría de Salud Jalisco, Guadalajara, Jalisco, Mexico
| | - Noemí Espinoza-García
- Doctorado en Ciencias en Biología Molecular en Medicina (DCBMM), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Nefertari Sagrero-Fabela
- Doctorado en Ciencias Biomédicas (DCB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Claudia Azucena Palafox-Sánchez
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
15
|
Qi J, Liu J, Zhao X, Huang H, Tang Y, Li X. IL-27 enhances peripheral B cell glycolysis of rheumatoid arthritis patients via activating mTOR signaling. Int Immunopharmacol 2023; 121:110532. [PMID: 37354782 DOI: 10.1016/j.intimp.2023.110532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/22/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
Our previous study found that increased serum IL-27 could promote rheumatoid arthritis (RA) B cell dysfunction via activating mTOR signaling pathway. This study aimed to explore the effects of IL-27 on B cell metabolism and clarify the mechanisms via which IL-27 enhancing glycolysis to induce B cells hyperactivation. Peripheral CD19+ B cells were purified from healthy controls (HC) and RA patients and then cultured with or without anti-CD40/CpG and glycolysis inhibitor 2-deoxy-D-glucose (2-DG) or mTOR inhibitor rapamycin. Furthermore, the isolated CD19+ B cells were treated by HC serum or RA serum in the presence and absence of recombinant human IL-27 or anti-IL-27 neutralizing antibodies or 2-DG or rapamycin. The B cell glycolysis level, proliferation, differentiation and inflammatory actions were detected by qPCR, flow cytometry or ELISA. We found that the glycolysis in RA B cells was increased significantly compared with HC B cells. Glycolysis inhibition downregulated the proliferation, differentiation, and inflammatory actions of RA B cells. RA serum and IL-27 promoted B cell glycolysis, which could be obviously rescued by anti-IL-27 antibodies or mTOR inhibitor rapamycin. Our results suggest that the enhanced cellular glycolysis of RA B cells induced by IL-27 may contribute to B cells hyperactivation through activating the mTOR signaling pathway.
Collapse
Affiliation(s)
- Jingjing Qi
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, Liaoning, People's Republic of China
| | - Jiaqing Liu
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, Liaoning, People's Republic of China
| | - Xiangge Zhao
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, Liaoning, People's Republic of China
| | - Huina Huang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, Liaoning, People's Republic of China
| | - Yawei Tang
- Department of Flow Cytometry Center, Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116044, Liaoning, People's Republic of China.
| | - Xia Li
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, Liaoning, People's Republic of China.
| |
Collapse
|
16
|
Gao Y, Zhou J, Huang Y, Wang M, Zhang Y, Zhang F, Gao Y, Zhang Y, Li H, Sun J, Xie Z. Jiedu-Quyu-Ziyin Fang (JQZF) inhibits the proliferation and activation of B cells in MRL/lpr mice via modulating the AKT/mTOR/c-Myc signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023:116625. [PMID: 37236380 DOI: 10.1016/j.jep.2023.116625] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiedu-Quyu-Ziyin Fang (JQZF) is a new herbal formula improved based on "Sheng Ma Bie Jia Tang" in the Golden Chamber, has been proved to be effective in the treatment of SLE. The ability of JQZF to prevent lymphocyte growth and survival has been demonstrated in earlier investigations. However, the specific mechanism of JQZF on SLE has not been fully investigated. AIM OF THE STUDY To reveal the potential mechanisms of JQZF inhibiting B cell proliferation and activation in MRL/lpr mice. MATERIALS AND METHODS MRL/lpr mice were treated with low-dose, high-dose JQZF and normal saline for 6 weeks. The effect of JQZF on disease improvement in MRL/lpr mice was studied using enzyme-linked immunosorbent assay (ELISA), histopathological staining, serum biochemical parameters and urinary protein levels. The changes of B lymphocyte subsets in the spleen were analyzed by flow cytometry. The contents of ATP and PA in B lymphocytes from the spleens of mice were determined by ATP content assay kit and PA assay kit. Raji cells (a B lymphocyte line) were selected as the cell model in vitro. The effects of JQZF on the proliferation and apoptosis of B cells were detected by flow cytometry and CCK8. The effect of JQZF on the AKT/mTOR/c-Myc signaling pathway in B cells were detected via western blot. RESULTS JQZF, especially at high dose, significantly improved the disease development of MRL/lpr mice. Flow cytometry results showed that JQZF affected the proliferation and activation of B cells. In addition, JQZF inhibited the production of ATP and PA in B lymphocytes. In vitro cell experiments further confirmed that JQZF can inhibit Raji proliferation and promote cell apoptosis through AKT/mTOR/c-Myc signaling pathway. CONCLUSION JQZF may affect the proliferation and activation of B cells by inhibiting the AKT/mTOR/c-Myc signaling pathway.
Collapse
Affiliation(s)
- YiNi Gao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - JiaWang Zhou
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yao Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - MeiJiao Wang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yi Zhang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - FengQi Zhang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yan Gao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - YiYang Zhang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - HaiChang Li
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Jing Sun
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| | - ZhiJun Xie
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
17
|
Iwata S, Hajime Sumikawa M, Tanaka Y. B cell activation via immunometabolism in systemic lupus erythematosus. Front Immunol 2023; 14:1155421. [PMID: 37256149 PMCID: PMC10225689 DOI: 10.3389/fimmu.2023.1155421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an inflammatory autoimmune disease involving multiple organs in which B cells perform important functions such as antibody and cytokine production and antigen presentation. B cells are activated and differentiated by the primary B cell receptor, co-stimulatory molecule signals-such as CD40/CD40L-, the Toll-like receptors 7,9, and various cytokine signals. The importance of immunometabolism in the activation, differentiation, and exerting functions of B cells and other immune cells has been widely reported in recent years. However, the regulatory mechanism of immunometabolism in B cells and its involvement in SLE pathogenesis remain elusive. Similarly, the importance of the PI3K-Akt-mTOR signaling pathway, glycolytic system, and oxidative phosphorylation has been demonstrated in the mechanisms of B cell immunometabolic activation, mainly in mouse studies. However, the activation of the mTOR pathway in B cells in patients with SLE, the induction of plasmablast differentiation through metabolic and transcription factor regulation by mTOR, and the involvement of this phenomenon in SLE pathogenesis are unclear. In our studies using activated B cells derived from healthy donors and from patients with SLE, we observed that methionine, an essential amino acid, is important for mTORC1 activation. Further, we observed that splenic tyrosine kinase and mTORC1 activation synergistically induce EZH2 expression and plasmablasts by suppressing BACH2 expression through epigenomic modification. Additionally, we identified another mechanism by which the glutaminolysis-induced enhancement of mitochondrial function promotes plasmablast differentiation in SLE. In this review, we focused on the SLE exacerbation mechanisms related to the activation of immune cells-especially B cells-and immunometabolism and reported the latest findings in the field.
Collapse
Affiliation(s)
- Shigeru Iwata
- Department of Rheumatology and Clinical Immunology, Wakayama Medical University, Wakayama, Japan
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Maiko Hajime Sumikawa
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
18
|
Luo X, Wu X, Wang A, Chen Y, Peng Y, Deng C, Zhao L, Yang H, Zhou J, Peng L, Wu Q, Li M, Zhao Y, Zeng X, Zhang W, Fei Y. mTORC1-GLUT1-mediated glucose metabolism drives hyperactivation of B cells in primary Sjogren's syndrome. Immunology 2023; 168:432-443. [PMID: 36155926 DOI: 10.1111/imm.13580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 09/07/2022] [Indexed: 11/28/2022] Open
Abstract
Primary Sjögren's syndrome (pSS) is a chronic systemic autoimmune disease characterized by B cell hyperactivation and hypergrammaglobulinemia. Currently, the role of metabolic pathways in the B cells of pSS patients is poorly defined. Here, we showed that upon cytosine phosphate-guanine (CpG)/sCD40L/IL-4 stimulation, B cells proportionally increased glycolysis and oxygen consumption, and compared with B cells from healthy controls (HCs), B cells from pSS patients exhibited higher glycolysis capacity and maximal oxidative respiration (OXPHOS). We also found that glucose transporter 1 (GLUT1) expression in B cells from pSS patients was significantly higher than that in B cells from HCs. Treatment with 2-deoxy-d-glucose (2-DG) inhibited the activation of B cells in pSS patients. Both 2-DG and Metformin inhibited the proliferation, formation of plasma/plasmablasts and decreased the IgG and IgM levels in the supernatants of B cells from pSS patients. Furthermore, inhibition of mTORC1 by rapamycin had an effect similar to that of 2-DG, suppressing B cell activation, proliferation and antibody production. Taken together, we demonstrated that B cells from pSS patients are more metabolically active than those from HCs and suggested that the mTORC1-GLUT1 glycolysis pathways were the major drivers of B cell hyperactivation and autoantibody production in pSS patients.
Collapse
Affiliation(s)
- Xuan Luo
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China.,Department of Rheumatology, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xunyao Wu
- Clinical Biobank, Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Anqi Wang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Yingying Chen
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Yu Peng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Chuiwen Deng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Lidan Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Huaxia Yang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Jiaxin Zhou
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Linyi Peng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Qingjun Wu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Mengtao Li
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Yan Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Wen Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Yunyun Fei
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| |
Collapse
|
19
|
Iwasaki Y, Takeshima Y, Nakano M, Okubo M, Ota M, Suzuki A, Kochi Y, Okamura T, Endo T, Miki I, Sakurada K, Yamamoto K, Fujio K. Combined plasma metabolomic and transcriptomic analysis identify histidine as a biomarker and potential contributor in SLE pathogenesis. Rheumatology (Oxford) 2023; 62:905-913. [PMID: 35689621 DOI: 10.1093/rheumatology/keac338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES To investigate metabolite alterations in the plasma of SLE patients to identify novel biomarkers and provide insight into SLE pathogenesis. METHODS Patients with SLE (n = 41, discovery cohort and n = 37, replication cohort), healthy controls (n = 30 and n = 29) and patients with RA (n = 19, disease control) were recruited. Metabolic profiles of the plasma samples were analysed using liquid chromatography-time-of-flight mass spectrometry and capillary electrophoresis-time-of-flight mass spectrometry. Transcriptome data was analysed using RNA-sequencing for 18 immune cell subsets. The importance of histidine (His) in plasmablast differentiation was investigated by using mouse splenic B cells. RESULTS We demonstrate that a specific amino acid combination including His can effectively distinguish between SLE patients and healthy controls. Random forest and partial least squares-discriminant analysis identified His as an effective classifier for SLE patients. A decrease in His plasma levels correlated with damage accrual independent of prednisolone dosage and type I IFN signature. The oxidative phosphorylation signature in plasmablasts negatively correlated with His levels. We also showed that plasmablast differentiation induced by innate immune signals was dependent on His. CONCLUSIONS Plasma His levels are a potential biomarker for SLE patients and are associated with damage accrual. Our data suggest the importance of His as a pathogenic metabolite in SLE pathogenesis.
Collapse
Affiliation(s)
- Yukiko Iwasaki
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo.,Department of Palliative Medicine.,Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama
| | - Yusuke Takeshima
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo.,Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo
| | - Masahiro Nakano
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo.,Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama
| | - Mai Okubo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo
| | - Mineto Ota
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo.,Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo
| | - Akari Suzuki
- Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama
| | - Yuta Kochi
- Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama.,Department of Genomic Function and Diversity, Tokyo Medical and Dental University, Tokyo
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo.,Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo
| | - Takaho Endo
- Medical Sciences Innovation Hub Program, Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama
| | - Ichiro Miki
- Medical Sciences Innovation Hub Program, Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama
| | - Kazuhiro Sakurada
- Medical Sciences Innovation Hub Program, Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama.,Department of Extended Intelligence for Medicine, The Ishii-Ishibashi Laboratory, Keio University School of Medicine Graduate School of Medicine, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo
| |
Collapse
|
20
|
The oxidative phosphorylation inhibitor IM156 suppresses B-cell activation by regulating mitochondrial membrane potential and contributes to the mitigation of systemic lupus erythematosus. Kidney Int 2023; 103:343-356. [PMID: 36332729 DOI: 10.1016/j.kint.2022.09.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/09/2022] [Accepted: 09/22/2022] [Indexed: 11/15/2022]
Abstract
Current treatment strategies for autoimmune diseases may not sufficiently control aberrant metabolism in B-cells. To address this concern, we investigated a biguanide derivative, IM156, as a potential regulator for B-cell metabolism in vitro and in vivo on overactive B-cells stimulated by the pro-inflammatory receptor TLR-9 agonist CpG oligodeoxynucleotide, a mimic of viral/bacterial DNA. Using RNA sequencing, we analyzed the B-cell transcriptome expression, identifying the major molecular pathways affected by IM156 in vivo. We also evaluated the anti-inflammatory effects of IM156 in lupus-prone NZB/W F1 mice. CD19+B-cells exhibited higher mitochondrial mass and mitochondrial membrane potential compared to T-cells and were more susceptible to IM156-mediated oxidative phosphorylation inhibition. In vivo, IM156 inhibited mitochondrial oxidative phosphorylation, cell cycle progression, plasmablast differentiation, and activation marker levels in CpG oligodeoxynucleotide-stimulated mouse spleen B-cells. Interestingly, IM156 treatment significantly increased overall survival, reduced glomerulonephritis and inhibited B-cell activation in the NZB/W F1 mice. Thus, our data indicated that IM156 suppressed the mitochondrial membrane potentials of activated B-cells in mice, contributing to the mitigation of lupus activity. Hence, IM156 may represent a therapeutic alternative for autoimmune disease mediated by B-cell hyperactivity.
Collapse
|
21
|
Peripheral immunophenotypes associated with the flare in the systemic lupus erythematosus patients with low disease activity state. Clin Immunol 2022; 245:109166. [DOI: 10.1016/j.clim.2022.109166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/08/2021] [Accepted: 10/14/2022] [Indexed: 11/22/2022]
|
22
|
Shanley LC, Fitzgerald HK, O’Rourke SA, Dunne A. Endogenous drivers of altered immune cell metabolism. Exp Biol Med (Maywood) 2022; 247:2192-2200. [PMID: 36511089 PMCID: PMC9899986 DOI: 10.1177/15353702221134093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Dysregulated metabolism has long been recognized as a feature of many metabolic disorders. However, recent studies demonstrating that metabolic reprogramming occurs in immune cells have led to a growing interest in the relationship between metabolic rewiring and immune-mediated disease pathogeneses. It is clear now that immune cell subsets engage in different metabolic pathways depending on their activation and/or maturation state. As a result, it may be possible to modulate metabolic reprogramming for clinical benefit. In this review, we provide an overview of immune cell metabolism with focus on endogenous drivers of metabolic reprogramming given their link to a number of immune-mediated disorders.
Collapse
Affiliation(s)
- Lianne C Shanley
- School of Biochemistry &
Immunology, Trinity College, University of Dublin, Dublin 2, Ireland
- Centre for Advanced Materials and
Bioengineering Research (AMBER), Trinity College Dublin, Dublin 2,
Ireland
| | - Hannah K Fitzgerald
- School of Biochemistry &
Immunology, Trinity College, University of Dublin, Dublin 2, Ireland
| | - Sinead A O’Rourke
- School of Biochemistry &
Immunology, Trinity College, University of Dublin, Dublin 2, Ireland
- School of Engineering, Trinity
College, University of Dublin, Dublin 2, Ireland
| | - Aisling Dunne
- School of Biochemistry &
Immunology, Trinity College, University of Dublin, Dublin 2, Ireland
- Centre for Advanced Materials and
Bioengineering Research (AMBER), Trinity College Dublin, Dublin 2,
Ireland
- School of Medicine, Trinity
College, University of Dublin, Dublin 2, Ireland
- Aisling Dunne.
| |
Collapse
|
23
|
Corneth OBJ, Neys SFH, Hendriks RW. Aberrant B Cell Signaling in Autoimmune Diseases. Cells 2022; 11:cells11213391. [PMID: 36359789 PMCID: PMC9654300 DOI: 10.3390/cells11213391] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/24/2022] [Indexed: 11/30/2022] Open
Abstract
Aberrant B cell signaling plays a critical in role in various systemic and organ-specific autoimmune diseases. This is supported by genetic evidence by many functional studies in B cells from patients or specific animal models and by the observed efficacy of small-molecule inhibitors. In this review, we first discuss key signal transduction pathways downstream of the B cell receptor (BCR) that ensure that autoreactive B cells are removed from the repertoire or functionally silenced. We provide an overview of aberrant BCR signaling that is associated with inappropriate B cell repertoire selection and activation or survival of peripheral B cell populations and plasma cells, finally leading to autoantibody formation. Next to BCR signaling, abnormalities in other signal transduction pathways have been implicated in autoimmune disease. These include reduced activity of several phosphates that are downstream of co-inhibitory receptors on B cells and increased levels of BAFF and APRIL, which support survival of B cells and plasma cells. Importantly, pathogenic synergy of the BCR and Toll-like receptors (TLR), which can be activated by endogenous ligands, such as self-nucleic acids, has been shown to enhance autoimmunity. Finally, we will briefly discuss therapeutic strategies for autoimmune disease based on interfering with signal transduction in B cells.
Collapse
|
24
|
Bystrom J, Taher TE, Henson SM, Gould DJ, Mageed RA. Metabolic requirements of Th17 cells and of B cells: Regulation and defects in health and in inflammatory diseases. Front Immunol 2022; 13:990794. [PMCID: PMC9614365 DOI: 10.3389/fimmu.2022.990794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
The immune system protects from infections and cancer through complex cellular networks. For this purpose, immune cells require well-developed mechanisms of energy generation. However, the immune system itself can also cause diseases when defective regulation results in the emergence of autoreactive lymphocytes. Recent studies provide insights into how differential patterns of immune cell responses are associated with selective metabolic pathways. This review will examine the changing metabolic requirements of Th17 cells and of B cells at different stages of their development and activation. Both cells provide protection but can also mediate diseases through the production of autoantibodies and the production of proinflammatory mediators. In health, B cells produce antibodies and cytokines and present antigens to T cells to mount specific immunity. Th17 cells, on the other hand, provide protection against extra cellular pathogens at mucosal surfaces but can also drive chronic inflammation. The latter cells can also promote the differentiation of B cells to plasma cells to produce more autoantibodies. Metabolism-regulated checkpoints at different stages of their development ensure the that self-reactive B cells clones and needless production of interleukin (IL-)17 are limited. The metabolic regulation of the two cell types has some similarities, e.g. the utility of hypoxia induced factor (HIF)1α during low oxygen tension, to prevent autoimmunity and regulate inflammation. There are also clear differences, as Th17 cells only are vulnerable to the lack of certain amino acids. B cells, unlike Th17 cells, are also dependent of mechanistic target of rapamycin 2 (mTORC2) to function. Significant knowledge has recently been gained, particularly on Th17 cells, on how metabolism regulates these cells through influencing their epigenome. Metabolic dysregulation of Th17 cells and B cells can lead to chronic inflammation. Disease associated alterations in the genome can, in addition, cause dysregulation to metabolism and, thereby, result in epigenetic alterations in these cells. Recent studies highlight how pathology can result from the cooperation between the two cell types but only few have so far addressed the key metabolic alterations in such settings. Knowledge of the impact of metabolic dysfunction on chronic inflammation and pathology can reveal novel therapeutic targets to treat such diseases.
Collapse
Affiliation(s)
- Jonas Bystrom
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- *Correspondence: Jonas Bystrom, ; Taher E. Taher,
| | - Taher E. Taher
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Jonas Bystrom, ; Taher E. Taher,
| | - Sian M. Henson
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - David J. Gould
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Rizgar A. Mageed
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
25
|
Mao Z, Tan Y, Tao J, Li L, Yu F, Zhao M. mTORC1 activation induced proximal tubular damage via the pentose phosphate pathway in lupus nephritis. Free Radic Biol Med 2022; 189:91-101. [PMID: 35863688 DOI: 10.1016/j.freeradbiomed.2022.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/03/2022] [Accepted: 07/13/2022] [Indexed: 10/17/2022]
Abstract
More recent studies suggested that metabolic disorders could contribute to the pathogenesis of systemic lupus erythematosus (SLE) and lupus nephritis (LN). The present work aimed at identifying metabolic reprogramming in the kidney of lupus nephritis via proteomics and investigating the potential regulatory mechanism. The proteomic studies on the renal biopsies revealed that the pentose phosphate pathway (PPP) was significantly enriched in the kidneys of LN patients compared with normal controls (NCs). Immunohistochemical stanning of glucose-6-phosphate dehydrogenase (G6PD), the key rate-limiting enzyme of PPP, verify the results of proteomics. We found that G6PD was highly expressed in the kidneys of LN patients and correlated with several clinicopathological indices. The univariate Cox regression analysis (HR, 95%CI, 1.877 (1.059-3.328), P = 0.031) and Kaplan-Meier (KM) analysis (P = 0.028) suggested that high G6PD expression in the tubulointerstitial area was a risk factor for worse prognosis. Moreover, the Gene set enrichment analysis (GSEA) demonstrated that the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway ranked first in the kidneys of LN patients with high G6PD expression and G6PD was co-localized with mTORC1 activation in the tubule. Immunoglobulin G (IgG) isolated from LN patients significantly activated the mTORC1 pathway and increased G6PD expression, G6PD activity, NADPH production, NADPH oxidase 2 (NOX2) expression, reactive oxygen species (ROS) production, and cell apoptosis in tubule cells in vitro. The above phenotypes were partially rescued after the addition of rapamycin or knock-down of G6PD. Overall, our study suggested that renal G6PD expression was associated with the overall enhanced disease activity and worse renal prognosis. mTORC1 activation might be involved in IgG-LN-induced tubular damage via PPP.
Collapse
Affiliation(s)
- Zhaomin Mao
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100034, PR China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR China
| | - Juan Tao
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR China
| | - Linlin Li
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR China
| | - Feng Yu
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR China; Department of Nephrology, Peking University International Hospital, Beijing, 102206, PR China.
| | - Minghui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100034, PR China
| |
Collapse
|
26
|
Chakraborty S, Khamaru P, Bhattacharyya A. Regulation of immune cell metabolism in health and disease: Special focus on T and B cell subsets. Cell Biol Int 2022; 46:1729-1746. [PMID: 35900141 DOI: 10.1002/cbin.11867] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 04/03/2022] [Accepted: 07/05/2022] [Indexed: 12/15/2022]
Abstract
Metabolism is a dynamic process and keeps changing from time to time according to the demand of a particular cell to meet its bio-energetic requirement. Different immune cells rely on distinct metabolic programs which allow the cell to balance its requirements for energy, molecular biosynthesis, and effector activity. In the aspect of infection and cancer immunology, effector T and B cells get exhausted and help tumor cells to evade immunosurveillance. On the other hand, T cells become hyperresponsive in the scenario of autoimmune diseases. In this article, we have explored the uniqueness and distinct metabolic features of key CD4+ T and B helper cell subsets, CD4+ T, B regulatory cell subsets and CD8+ T cells regarding health and disease. Th1 cells rely on glycolysis and glutaminolysis; inhibition of these metabolic pathways promotes Th1 cells in Treg population. However, Th2 cells are also dependent on glycolysis but an abundance of lactate within TME shifts their metabolic dependency to fatty acid metabolism. Th17 cells depend on HIF-1α mediated glycolysis, ablation of HIF-1α reduces Th17 cells but enhance Treg population. In contrast to effector T cells which are largely dependent on glycolysis for their differentiation and function, Treg cells mainly rely on FAO for their function. Therefore, it is of utmost importance to understand the metabolic fates of immune cells and how it facilitates their differentiation and function for different disease models. Targeting metabolic pathways to restore the functionality of immune cells in diseased conditions can lead to potent therapeutic measures.
Collapse
Affiliation(s)
- Sayan Chakraborty
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Poulomi Khamaru
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| |
Collapse
|
27
|
Zhao L, Hu X, Xiao F, Zhang X, Zhao L, Wang M. Mitochondrial impairment and repair in the pathogenesis of systemic lupus erythematosus. Front Immunol 2022; 13:929520. [PMID: 35958572 PMCID: PMC9358979 DOI: 10.3389/fimmu.2022.929520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022] Open
Abstract
Nucleic acid autoantibodies, increase type I interferon (IFN-α) levels, and immune cell hyperactivation are hallmarks of systemic lupus erythematosus (SLE). Notably, immune cell activation requires high level of cellular energy that is predominately generated by the mitochondria. Mitochondrial reactive oxygen species (mROS), the byproduct of mitochondrial energy generation, serves as an essential mediator to control the activation and differentiation of cells and regulate the antigenicity of oxidized nucleoids within the mitochondria. Recently, clinical trials on normalization of mitochondrial redox imbalance by mROS scavengers and those investigating the recovery of defective mitophagy have provided novel insights into SLE prophylaxis and therapy. However, the precise mechanism underlying the role of oxidative stress-related mitochondrial molecules in skewing the cell fate at the molecular level remains unclear. This review outlines distinctive mitochondrial functions and pathways that are involved in immune responses and systematically delineates how mitochondrial dysfunction contributes to SLE pathogenesis. In addition, we provide a comprehensive overview of damaged mitochondrial function and impaired metabolic pathways in adaptive and innate immune cells and lupus-induced organ tissues. Furthermore, we summarize the potential of current mitochondria-targeting drugs for SLE treatment. Developing novel therapeutic approaches to regulate mitochondrial oxidative stress is a promising endeavor in the search for effective treatments for systemic autoimmune diseases, particularly SLE.
Collapse
Affiliation(s)
- Like Zhao
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianda Hu
- Beijing Tibetan Hospital, China Tibetology Research Center, Beijing, China
| | - Fei Xiao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- *Correspondence: Min Wang, ; Lidan Zhao,
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Min Wang, ; Lidan Zhao,
| |
Collapse
|
28
|
Horisberger A, Humbel M, Fluder N, Bellanger F, Fenwick C, Ribi C, Comte D. Measurement of circulating CD21 -CD27 - B lymphocytes in SLE patients is associated with disease activity independently of conventional serological biomarkers. Sci Rep 2022; 12:9189. [PMID: 35654865 PMCID: PMC9163192 DOI: 10.1038/s41598-022-12775-4] [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: 12/24/2021] [Accepted: 05/16/2022] [Indexed: 12/17/2022] Open
Abstract
Determining disease activity in systemic lupus erythematosus (SLE) patients is challenging and limited by the lack of reliable biomarkers. Abnormally activated B cells play a key role in the pathogenesis of SLE, but their measure in clinical practice is currently not recommended. Here, we studied peripheral B cells to identify a valid biomarker. We analyzed peripheral B cells in a discovery cohort of 30 SLE patients compared to 30 healthy controls (HC) using mass cytometry and unsupervised clustering analysis. The relevant B cell populations were subsequently studied by flow cytometry in a validation cohort of 63 SLE patients, 28 autoimmune diseases controls and 39 HC. Our data show an increased frequency of B cell populations with activated phenotype in SLE compared to healthy and autoimmune diseases controls. These cells uniformly lacked the expression of CD21 and CD27. Measurement of CD21−CD27− B cells in the blood identified patients with active disease and their frequency correlated with disease severity. Interestingly, we did not observe an increase in the frequency of CD21−CD27− B cells in patients with clinically inactive disease but with elevated conventional biomarkers (anti-dsDNA and complement levels). Accordingly, measurement of CD21−CD27− B cells represents a robust and easily accessible biomarker to assess the activity of the disease in SLE patients.
Collapse
Affiliation(s)
- Alice Horisberger
- Department of Medicine, Service of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, 46, Rue du Bugnon, 1011, Lausanne, Switzerland
| | - Morgane Humbel
- Department of Medicine, Service of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, 46, Rue du Bugnon, 1011, Lausanne, Switzerland
| | - Natalia Fluder
- Department of Medicine, Service of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, 46, Rue du Bugnon, 1011, Lausanne, Switzerland
| | - Florence Bellanger
- Department of Medicine, Service of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, 46, Rue du Bugnon, 1011, Lausanne, Switzerland
| | - Craig Fenwick
- Department of Medicine, Service of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, 46, Rue du Bugnon, 1011, Lausanne, Switzerland
| | - Camillo Ribi
- Department of Medicine, Service of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, 46, Rue du Bugnon, 1011, Lausanne, Switzerland
| | - Denis Comte
- Department of Medicine, Service of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, 46, Rue du Bugnon, 1011, Lausanne, Switzerland.
| |
Collapse
|
29
|
Wu X, Peng Y, Li J, Zhang P, Liu Z, Lu H, Peng L, Zhou J, Fei Y, Zeng X, Zhao Y, Zhang W. Single-Cell Sequencing of Immune Cell Heterogeneity in IgG4-Related Disease. Front Immunol 2022; 13:904288. [PMID: 35693817 PMCID: PMC9184520 DOI: 10.3389/fimmu.2022.904288] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/28/2022] [Indexed: 12/26/2022] Open
Abstract
Background The IgG4-related disease (IgG4-RD) is an immune-mediated disorder with fibrotic manifestations. However, the transcriptional profiles of immune cell subsets at single-cell level are unknown. Herein, single-cell sequencing was used to assess the specific cell subpopulations and pathways in peripheral blood mononuclear cells (PBMCs) of IgG4-RD. Methods Single-cell sequencing was performed using the PBMCs from four patients with IgG4-RD and three healthy controls (HCs). Functional enrichment and cell analysis were performed through re-clustering of PBMCs to assess functional pathways and intercellular communication networks in IgG4-RD. Western blot and flow cytometry were used to verify sequencing and functional enrichment results. Results Four major cell types and 21 subtypes were identified. Further subclustering demonstrated that plasma B-cell proportions increased with increasing glycolysis/gluconeogenesis activity in IgG4-RD. Re-clustering of myeloid cells showed that EGR1 and CD36 expressions were significantly increased in CD14+ monocytes of IgG4-RD, as validated by Western blot analysis. Moreover, tumor necrosis factor (TNF) production pathways were positively regulated in CD14+ monocytes of IgG4-RD. In vitro stimulation showed that CD14+ monocytes of IgG4-RD could secrete higher levels of TNF-α . Notably, the proportions of CD8 central memory T (TCM) and TIGIT+ CD8 cytotoxic T (CTL) increased in patients with IgG4-RD compared with HCs. Further interaction analysis showed that B cell activation factor (BAFF) signaling pathways were enriched from myeloid cells subsets to B cells. Conclusion This study enhances the understanding of the cellular heterogeneity and transcriptional features involved in the pathogenesis of IgG4-RD, providing key clinical implications.
Collapse
Affiliation(s)
- Xunyao Wu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- Clinical Biobank, Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Peng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Jieqiong Li
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Panpan Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Zheng Liu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Hui Lu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Linyi Peng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Jiaxin Zhou
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Yunyun Fei
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Yan Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- *Correspondence: Yan Zhao, ; Wen Zhang,
| | - Wen Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- *Correspondence: Yan Zhao, ; Wen Zhang,
| |
Collapse
|
30
|
Tzovara I, Papadatou I, Tzanoudaki M, Spoulou V. Development of a novel flow cytometry method for detecting pneumococcal‐specific B cells
1. Cytometry A 2022; 101:588-596. [DOI: 10.1002/cyto.a.24654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/16/2022] [Accepted: 05/05/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Irene Tzovara
- Department of Infectious Diseases ‐ Immunobiology and Vaccinology Research Lab “Aghia Sophia” Children's Hospital, 1st Department of Pediatrics ‐ National and Kapodistrian University of Athens Athens Greece
| | - Ioanna Papadatou
- Department of Infectious Diseases ‐ Immunobiology and Vaccinology Research Lab “Aghia Sophia” Children's Hospital, 1st Department of Pediatrics ‐ National and Kapodistrian University of Athens Athens Greece
| | - Marianna Tzanoudaki
- Department of Immunology & Histocompatibility, Specific Reference Centre for Primary Immunodeficiencies‐Paediatric Immunology “Aghia Sophia” Children's Hospital Athens Greece
| | - Vasiliki Spoulou
- Department of Infectious Diseases ‐ Immunobiology and Vaccinology Research Lab “Aghia Sophia” Children's Hospital, 1st Department of Pediatrics ‐ National and Kapodistrian University of Athens Athens Greece
| |
Collapse
|
31
|
Xu S, Guo Y, Luo T, Jiang P, Yan Z, He Y, Fu L, Liu H, Gao Z, Wang D, Sun Z, Yang X, Pan W, Sun F. Transcriptomic Profiles of Splenic CD19 + B Cells in Mice Chronically Infected With the Larval Echinococcus granulosus. Front Vet Sci 2022; 9:848458. [PMID: 35548052 PMCID: PMC9082817 DOI: 10.3389/fvets.2022.848458] [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: 01/04/2022] [Accepted: 03/28/2022] [Indexed: 11/26/2022] Open
Abstract
Background We previously reported that the larval Echinococcus granulosus (E. granulosus) infection can expand the population of regulatory B cells in mice, thereby inhibiting the anti-infective immunity. However, the underlying mechanism is still largely unknown. This study further investigated the holistic transcriptomic profiles of total splenic B cells following the chronic infection of the parasite. Methods The infection model of larval E. granulosus was established by intraperitoneal inoculation with 2000 protoscolexes. Magnetic-Activated Cell Separation (MACS) was used to isolate the total splenic B cells. RNA sequencing was performed to screen the differentially expressed genes (DEGs) after infection. The expression of selected DEGs was verified using qRT-PCR. Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and Co-expression network analysis were applied to predict these DEGs' underlying biological processes, pathways, and interactions respectively. Results A total of 413 DEGs were identified in larval E. granulosus infected B cells, including 303 up- and 110 down-regulated genes. Notably, most DEGs related to inflammation and chemotaxis were significantly upregulated after infection. In line with these changes, significant expression upregulation of DEGs associated with fatty acid oxidation, lipid synthesis, lipolysis, lipid transport, and cholesterol biosynthesis, were observed in infected B cells. Co-expression network analysis showed an intimate interaction between these DEGs associated with immune and metabolism. Conclusions The present study revealed that the larval E. granulosus infection induces metabolic reprogramming of B cells, which provides a novel clue to clarify the immunoregulatory mechanism of B cells in parasitic infection.
Collapse
Affiliation(s)
- Shiping Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Yuxin Guo
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Tiancheng Luo
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Pengfei Jiang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Ziyi Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Yan He
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Linlin Fu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Hua Liu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai, China
- National Health Commission Key Laboratory of Parasite and Vector Biology, Shanghai, China
- World Health Organization Collaborating Centre for Tropical Diseases, Shanghai, China
- National Center for International Research on Tropical Diseases, Shanghai, China
| | - Zixuan Gao
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Dingmin Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Zhengxiu Sun
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Fenfen Sun
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, China
| |
Collapse
|
32
|
SLC7A5 expression is up-regulated in peripheral blood T and B lymphocytes of systemic lupus erythematosus patients, associating with renal damage. Clin Immunol 2022; 237:108987. [DOI: 10.1016/j.clim.2022.108987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/16/2022] [Accepted: 03/23/2022] [Indexed: 12/21/2022]
|
33
|
Sun W, Li P, Cai J, Ma J, Zhang X, Song Y, Liu Y. Lipid Metabolism: Immune Regulation and Therapeutic Prospectives in Systemic Lupus Erythematosus. Front Immunol 2022; 13:860586. [PMID: 35371016 PMCID: PMC8971568 DOI: 10.3389/fimmu.2022.860586] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous disease characterized by the production of abnormal autoantibodies and immune complexes that can affect the organ and organ systems, particularly the kidneys and the cardiovascular system. Emerging evidence suggests that dysregulated lipid metabolism, especially in key effector cells, such as T cells, B cells, and innate immune cells, exerts complex effects on the pathogenesis and progression of SLE. Beyond their important roles as membrane components and energy storage, different lipids can also modulate different cellular processes, such as proliferation, differentiation, and survival. In this review, we summarize altered lipid metabolism and the associated mechanisms involved in the pathogenesis and progression of SLE. Furthermore, we discuss the recent progress in the role of lipid metabolism as a potential therapeutic target in SLE.
Collapse
Affiliation(s)
- Wei Sun
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Southeast University, Nanjing, China
| | - Pengchong Li
- Department of Rheumatology and Clinical Immunology, The Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Beijing, China
- Department of Gastroenterology, Beijing Friendship Hospital, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease center, Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Capital Medical University, Beijing, China
| | - Jianping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontolog, Beijing, China
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Southeast University, Nanjing, China
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
- *Correspondence: Yudong Liu, ; Yong Song,
| | - Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Center of Biotherapy, Beijing Hospital, National Center of Gerontolog, Beijing, China
- *Correspondence: Yudong Liu, ; Yong Song,
| |
Collapse
|
34
|
Iwata S, Tanaka Y. Association of Viral Infection With the Development and Pathogenesis of Systemic Lupus Erythematosus. Front Med (Lausanne) 2022; 9:849120. [PMID: 35280878 PMCID: PMC8914279 DOI: 10.3389/fmed.2022.849120] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/03/2022] [Indexed: 12/27/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that causes multiple organ damage in women of childbearing age and has a relapsing-remitting course. SLE is caused by the interaction between genetic and environmental factors, however, its underlying triggers remain unknown. Among the environmental factors, the involvement of infections as a trigger for SLE, especially those of viral etiology, has been widely reported. Human endogenous retroviruses (HERVs) may put patients at a genetic predisposition to SLE, while the Epstein-Barr virus (EBV) may play a role as an environmental factor that triggers the development of SLE. It has been suggested that EBV-infected B-cells may become resistant to apoptosis, resulting in the activation, proliferation, and antibody production of autoreactive B-cells, which cause tissue damage in SLE. However, the interaction between the virus and immune cells, as well as the impact of the virus on the differentiation and dysfunction of immune cells, remain unclear. In this review, we focus on the relationship between the development and pathogenesis of SLE and viral infections, as well as the mechanism of SLE exacerbation via activation of immune cells, such as B-cells, based on the latest findings.
Collapse
|
35
|
iNKT cells can effectively inhibit IL-6 production by B cells in systemic sclerosis. Cytotherapy 2022; 24:482-488. [PMID: 35181242 DOI: 10.1016/j.jcyt.2021.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is a connective tissue disease with poorly understood pathogenesis and limited treatment options. Patient mortality is rooted predominantly in the development of pulmonary and cardiac complications. The overactivated immune system is assumed to sustain the inflammatory signature of this autoimmune disease. Here, we investigate the potential of immunoregulatory invariant natural killer T (iNKT) cells to inhibit proinflammatory B cell responses in an in vitro model of inflammation. METHODS B cells from healthy volunteers (n = 17) and patients with SSc (n = 15) were used for functional testing upon lipopolysaccharide (LPS) stimulation in a co-culture system with third-party iNKT cells. Cytokine production was measured with antibody-based immunoassays (ELISA) and intracellular cytokine staining. RESULTS iNKT cells strongly inhibited the production of proinflammatory interleukin-6 by B cells upon stimulation with LPS in both healthy volunteers and patients with SSc. In a Transwell assay, cell contact between B cells and iNKT cells proved necessary for this inhibitory effect. Similarly, blocking of CD1d on the surface of B cells abolished the immunoregulatory effect of iNKT cells on B cells. B cell subsets with higher expression of CD1d, namely unswitched memory B cells, were more susceptible to iNKT cell inhibition. CONCLUSION Our in vitro data underline the potential of iNKT cells in the control of SSc and provide a rationale for the use of novel iNKT cell-based therapeutic strategies in the context of autoimmune diseases.
Collapse
|
36
|
Mao Z, Tan Y, Tao J, Li L, Wang H, Yu F, Perl A, Zhao M. Renal mTORC1 activation is associated with disease activity and prognosis in lupus nephritis. Rheumatology (Oxford) 2022; 61:3830-3840. [PMID: 35040950 DOI: 10.1093/rheumatology/keac037] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/11/2022] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE This study was initiated to evaluate mammalian target of rapamycin (mTOR) activation in renal tissue of lupus nephritis (LN) patients. METHODS This retrospective study included 187 LN patients, 20 diabetic nephropathy (DN) patients, 10 minimal change disease (MCD) patients, and 10 normal controls (NCs). 7 of 187 LN patients had repeated renal biopsies. mTORC1/2 activation was evaluated by immunohistochemistry and multiplexed immunofluorescence. The association of mTORC1/2 activation with the clinicopathologic indices and prognostic outcomes was analysed among 187 LN patients. Proteomics was performed in renal biopsies of 20 LN patients. Proteomics was employed to comprehensively evaluate the impact of mTOR activation on intrarenal gene expression. RESULTS mTORC1/2 was significantly activated in podocytes, mesangial cells, endothelial cells and tubular epithelial cells of LN patients as compared with those with MCD or NC. The glomerular mTORC1 activation was higher in LN patients compared with DN patients. mTORC1, but not mTORC2, activation strongly correlated with serum albumin, complement C3, proteinuria, and the following pathological biomarkers of LN: crescent formation, interstitial inflammation and fibrosis. Moreover, mTORC1 activation was identified as a prognostic marker in LN patients. Bioinformatic analyses of proteomics and immunohistochemical data unveiled increased complement activation, antigen presentation, and phagocytosis in LN patients with mTORC1 activation. CONCLUSION Renal mTORC1 activation could be a biomarker to reveal disease activity and predict clinical prognosis in LN patients.
Collapse
Affiliation(s)
- Zhaomin Mao
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100034, PR. China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China
| | - Juan Tao
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China
| | - Linlin Li
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China
| | - Hui Wang
- Laboratory of Electron Microscopy, Peking University First Hospital, Beijing, PR China
| | - Feng Yu
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China.,Department of Nephrology, Peking University International Hospital, Beijing, 102206, PR. China
| | - Andras Perl
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, New York, Syracuse, NY, 13210, USA
| | - Minghui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100034, PR. China
| |
Collapse
|
37
|
Zhang Z, Cai J, Hao S, Li C, Chen J, Li T, Feng X. Transcriptomic analysis of spleen B cell revealed the molecular basis of bursopentin on B cell differentiation. Vet Res 2022; 53:109. [PMID: 36517897 PMCID: PMC9753308 DOI: 10.1186/s13567-022-01123-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/30/2022] [Indexed: 12/23/2022] Open
Abstract
The bursa of Fabricius, the acknowledged humoral immune organ unique to birds, plays a vital role in B cell development. Bursopentin (BP5) derived from the bursa is reported to induce the development and formation of B cells. However, the mechanism of BP5 on B cell differentiation is still unclear. In this paper, total B lymphocytes from mice immunized with H9N2 subtype AIV vaccine were stimulated with BP5. The results show that BP5 at the experimental dosages promoted B cell differentiation, including the total B cells, activated B cells, differentiated B cells, mature B cells and plasma cells. Then, the in vivo immune experiment proved that the percentages of activated and differentiated B cells from mice immunized with AIV vaccine and 0.25 mg/mL BP5 were increased. To investigate the molecular mechanism of BP5 on B cell differentiation, the gene expression profiles of B cells purified from the spleen cells of mice immunized with AIV vaccine and BP5 were detected following RNA sequencing technology. The results show that BP5 at 0.05 and 0.25 mg/mL induced the enrichment of various biological functions, and stimulated five common significant enrichment pathways in B cells from the immunized mice. Additionally, 120 and 59 differentially expressed genes (DEG) represented transcriptional factors in B cells following 0.05 and 0.25 mg/mL BP5 immunization, respectively. In summary, these results suggest that BP5 regulates various gene expression involved in regulation of B cell development, which provides the knowledge required for additional studies on B cell differentiation in response to bursal-derived peptides and also provides an important experimental basis for improving vaccine immunity.
Collapse
Affiliation(s)
- Ze Zhang
- grid.27871.3b0000 0000 9750 7019Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China ,grid.27871.3b0000 0000 9750 7019MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Jiaxi Cai
- grid.27871.3b0000 0000 9750 7019Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China ,grid.27871.3b0000 0000 9750 7019MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Shanshan Hao
- grid.27871.3b0000 0000 9750 7019Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China ,grid.27871.3b0000 0000 9750 7019MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Chenfei Li
- grid.27871.3b0000 0000 9750 7019Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China ,grid.27871.3b0000 0000 9750 7019MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Jiajing Chen
- grid.27871.3b0000 0000 9750 7019Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China ,grid.27871.3b0000 0000 9750 7019MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Tongtong Li
- grid.27871.3b0000 0000 9750 7019Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China ,grid.27871.3b0000 0000 9750 7019MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Xiuli Feng
- grid.27871.3b0000 0000 9750 7019Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China ,grid.27871.3b0000 0000 9750 7019MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| |
Collapse
|
38
|
Alves RW, da Silva EM, Doretto-Silva L, Andrade-Oliveira V. Metabolic Pathways in Immune Cells Commitment and Fate. ESSENTIAL ASPECTS OF IMMUNOMETABOLISM IN HEALTH AND DISEASE 2022:53-82. [DOI: 10.1007/978-3-030-86684-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
39
|
Mielle J, Morel J, ElHmioui J, Combe B, Macia L, Dardalhon V, Taylor N, Audo R, Daien C. Glutamine promotes the generation of B10 + cells via the mTOR/GSK3 pathway. Eur J Immunol 2021; 52:418-430. [PMID: 34961940 DOI: 10.1002/eji.202149387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 11/23/2021] [Accepted: 12/20/2021] [Indexed: 11/06/2022]
Abstract
Alterations in cell metabolism can shift the differentiation of immune cells towards a regulatory or inflammatory phenotype, thus opening up new therapeutic opportunities for immune-related diseases. Indeed, growing knowledge on T cell metabolism has revealed differences in the metabolic programs of suppressive regulatory T cells (Tregs) as compared to inflammatory Th1 and Th17 cells. In addition to Tregs, IL-10-producing regulatory B cells are crucial for maintaining tolerance, inhibiting inflammation and autoimmunity. Yet, the metabolic networks regulating diverse B lymphocyte responses are not well known. Here, we show that glutaminase blockade decreased downstream mTOR activation and attenuated IL-10 secretion. Direct suppression of mTOR activity by rapamycin selectively impaired IL-10 production by B cells whereas secretion was restored upon GSK3 inhibition. Mechanistically, we found mTORC1 activation leads to GSK3 inhibition, identifying a key signalling pathway regulating IL-10 secretion by B lymphocytes. Thus, our results identify glutaminolysis and the mTOR/GSK3 signalling axis, as critical regulators of the generation of IL-10 producing B cells with regulatory functions. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Julie Mielle
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Department of Rheumatology, CHU de Montpellier, Montpellier, France
| | - Jacques Morel
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Department of Rheumatology, CHU de Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM, CNRS UMR, Montpellier, France
| | - Jamila ElHmioui
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Bernard Combe
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Department of Rheumatology, CHU de Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM, CNRS UMR, Montpellier, France
| | - Laurence Macia
- Charles Perkins Centre, the University of Sydney, Sydney, Australia.,School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Rachel Audo
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Department of Rheumatology, CHU de Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM, CNRS UMR, Montpellier, France
| | - Claire Daien
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Department of Rheumatology, CHU de Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM, CNRS UMR, Montpellier, France
| |
Collapse
|
40
|
Immune Memory in Aging: a Wide Perspective Covering Microbiota, Brain, Metabolism, and Epigenetics. Clin Rev Allergy Immunol 2021; 63:499-529. [PMID: 34910283 PMCID: PMC8671603 DOI: 10.1007/s12016-021-08905-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 11/06/2022]
Abstract
Non-specific innate and antigen-specific adaptive immunological memories are vital evolutionary adaptations that confer long-lasting protection against a wide range of pathogens. Adaptive memory is established by memory T and B lymphocytes following the recognition of an antigen. On the other hand, innate immune memory, also called trained immunity, is imprinted in innate cells such as macrophages and natural killer cells through epigenetic and metabolic reprogramming. However, these mechanisms of memory generation and maintenance are compromised as organisms age. Almost all immune cell types, both mature cells and their progenitors, go through age-related changes concerning numbers and functions. The aging immune system renders the elderly highly susceptible to infections and incapable of mounting a proper immune response upon vaccinations. Besides the increased infectious burden, older individuals also have heightened risks of metabolic and neurodegenerative diseases, which have an immunological component. This review discusses how immune function, particularly the establishment and maintenance of innate and adaptive immunological memory, regulates and is regulated by epigenetics, metabolic processes, gut microbiota, and the central nervous system throughout life, with a focus on old age. We explain in-depth how epigenetics and cellular metabolism impact immune cell function and contribute or resist the aging process. Microbiota is intimately linked with the immune system of the human host, and therefore, plays an important role in immunological memory during both homeostasis and aging. The brain, which is not an immune-isolated organ despite former opinion, interacts with the peripheral immune cells, and the aging of both systems influences the health of each other. With all these in mind, we aimed to present a comprehensive view of the aging immune system and its consequences, especially in terms of immunological memory. The review also details the mechanisms of promising anti-aging interventions and highlights a few, namely, caloric restriction, physical exercise, metformin, and resveratrol, that impact multiple facets of the aging process, including the regulation of innate and adaptive immune memory. We propose that understanding aging as a complex phenomenon, with the immune system at the center role interacting with all the other tissues and systems, would allow for more effective anti-aging strategies.
Collapse
|
41
|
Zhang M, Iwata S, Sonomoto K, Ueno M, Fujita Y, Anan J, Miyazaki Y, Ohkubo N, Sumikawa MH, Todoroki Y, Miyata H, Nagayasu A, Kanda R, Trimova G, Lee S, Nakayamada S, Sakata K, Tanaka Y. mTOR activation in CD8+ cells contributes to disease activity of rheumatoid arthritis and increases therapeutic response to TNF inhibitors. Rheumatology (Oxford) 2021; 61:3010-3022. [PMID: 34791054 DOI: 10.1093/rheumatology/keab834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/09/2021] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE This study aimed to understand the role of mTOR in CD8+ cells in the pathogenicity of rheumatoid arthritis (RA) and the changes after treatment with biologic drugs. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated from 17 healthy controls and 86 patients with RA. Phosphorylation of mTOR (p-mTOR) and its clinical relevance were evaluated. The role of mTOR in CD8+ cells was also examined in vitro. RESULTS Patients with RA who had a moderate or high disease activity, were biologic-naïve, and were refractory to MTX were enrolled in this study. The p-mTOR levels in CD8+ cells were higher in patients with RA than in healthy controls, and they positively correlated with the disease activity in such patients. However, after one year of treatment with TNF inhibitors, the p-mTOR levels in CD8+ cells were suppressed and showed a positive correlation with the treatment response, which was not observed in the abatacept-treatment group. In vitro stimulation of CD8+ cells with anti-CD3 and anti-CD28 antibodies induced mTOR phosphorylation and increased the production of granzyme B, GNLY, TNF-α, and IFN-γ but decreased the production of granzyme K. However, on treatment with TNF inhibitors, p-mTOR levels in CD8+ cells and granzyme B production decreased, while granzyme K production increased. The production of GNLY and IFN-γ was not affected by the TNF inhibitors. CONCLUSION These results suggested that mTOR activation in CD8+ cells may be a novel evaluation marker for RA disease activity and a predictive marker of therapeutic response to TNF inhibitors.
Collapse
Affiliation(s)
- Mingzeng Zhang
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.,Department of Hematology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shigeru Iwata
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Koshiro Sonomoto
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masanobu Ueno
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuya Fujita
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Junpei Anan
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.,Mitsubishi Tanabe Pharma, Yokohama, Kanagawa, Japan
| | - Yusuke Miyazaki
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Naoaki Ohkubo
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Maiko Hajime Sumikawa
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yasuyuki Todoroki
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hiroko Miyata
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Atsushi Nagayasu
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Ryuichiro Kanda
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Gulzhan Trimova
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.,Department of Clinical Subjects, High School of Medicine, Faculty of Medicine and Health care, Al-Farabi Kazakh National University
| | - Seunghyun Lee
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kei Sakata
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.,Mitsubishi Tanabe Pharma, Yokohama, Kanagawa, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
42
|
Iperi C, Bordron A, Dueymes M, Pers JO, Jamin C. Metabolic Program of Regulatory B Lymphocytes and Influence in the Control of Malignant and Autoimmune Situations. Front Immunol 2021; 12:735463. [PMID: 34650560 PMCID: PMC8505885 DOI: 10.3389/fimmu.2021.735463] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/10/2021] [Indexed: 11/17/2022] Open
Abstract
Metabolic pathways have been studied for a while in eukaryotic cells. During glycolysis, glucose enters into the cells through the Glut1 transporter to be phosphorylated and metabolized generating ATP molecules. Immune cells can use additional pathways to adapt their energetic needs. The pentose phosphate pathway, the glutaminolysis, the fatty acid oxidation and the oxidative phosphorylation generate additional metabolites to respond to the physiological requirements. Specifically, in B lymphocytes, these pathways are activated to meet energetic demands in relation to their maturation status and their functional orientation (tolerance, effector or regulatory activities). These metabolic programs are differentially involved depending on the receptors and the co-activation molecules stimulated. Their induction may also vary according to the influence of the microenvironment, i.e. the presence of T cells, cytokines … promoting the expression of particular transcription factors that direct the energetic program and modulate the number of ATP molecule produced. The current review provides recent advances showing the underestimated influence of the metabolic pathways in the control of the B cell physiology, with a particular focus on the regulatory B cells, but also in the oncogenic and autoimmune evolution of the B cells.
Collapse
Affiliation(s)
| | - Anne Bordron
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France
| | - Maryvonne Dueymes
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France.,Service d'Odontologie, CHU de Brest, Brest, France
| | - Jacques-Olivier Pers
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France.,Service d'Odontologie, CHU de Brest, Brest, France
| | - Christophe Jamin
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France.,Laboratoire d'Immunologie et Immunothérapie, CHU de Brest, Brest, France
| |
Collapse
|
43
|
Yang L, Li N, Yang D, Chen A, Tang J, Jing Y, Kang D, Jiang P, Dai X, Luo L, Chen Q, Chang J, Liu J, Gu H, Huang Y, Chen Q, Li Z, Zhu Y, Miller H, Chen Y, Qiu L, Mei H, Hu Y, Gong Q, Liu C. CCL2 regulation of MST1-mTOR-STAT1 signaling axis controls BCR signaling and B-cell differentiation. Cell Death Differ 2021; 28:2616-2633. [PMID: 33879857 PMCID: PMC8408168 DOI: 10.1038/s41418-021-00775-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 02/01/2023] Open
Abstract
Chemokines are important regulators of the immune system, inducing specific cellular responses by binding to receptors on immune cells. In SLE patients, decreased expression of CCL2 on mesenchymal stem cells (MSC) prevents inhibition of B-cell proliferation, causing the characteristic autoimmune phenotype. Nevertheless, the intrinsic role of CCL2 on B-cell autoimmunity is unknown. In this study using Ccl2 KO mice, we found that CCL2 deficiency enhanced BCR signaling by upregulating the phosphorylation of the MST1-mTORC1-STAT1 axis, which led to reduced marginal zone (MZ) B cells and increased germinal center (GC) B cells. The abnormal differentiation of MZ and GC B cells were rescued by in vivo inhibition of mTORC1. Additionally, the inhibition of MST1-mTORC1-STAT1 with specific inhibitors in vitro also rescued the BCR signaling upon antigenic stimulation. The deficiency of CCL2 also enhanced the early activation of B cells including B-cell spreading, clustering and signalosome recruitment by upregulating the DOCK8-WASP-actin axis. Our study has revealed the intrinsic role and underlying molecular mechanism of CCL2 in BCR signaling, B-cell differentiation, and humoral response.
Collapse
Affiliation(s)
- Lu Yang
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Li
- grid.410654.20000 0000 8880 6009Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Di Yang
- grid.488412.3Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Ministry of Education Key Laboratory of Child Development and Disorder, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Anwei Chen
- grid.488412.3Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Ministry of Education Key Laboratory of Child Development and Disorder, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China ,grid.488412.3Department of Dermatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlong Tang
- grid.33199.310000 0004 0368 7223Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yukai Jing
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danqing Kang
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Panpan Jiang
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Dai
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Luo
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyue Chen
- grid.410654.20000 0000 8880 6009Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Jiang Chang
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ju Liu
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Gu
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanmei Huang
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianglin Chen
- grid.410654.20000 0000 8880 6009Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Zhenzhen Li
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingzi Zhu
- grid.33199.310000 0004 0368 7223Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- grid.94365.3d0000 0001 2297 5165Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Yan Chen
- grid.413390.cThe Second Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Liru Qiu
- grid.33199.310000 0004 0368 7223Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Mei
- grid.33199.310000 0004 0368 7223Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- grid.33199.310000 0004 0368 7223Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quan Gong
- grid.410654.20000 0000 8880 6009Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Chaohong Liu
- grid.33199.310000 0004 0368 7223Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Iwata S, Tanaka Y. Therapeutic perspectives on the metabolism of lymphocytes in patients with rheumatoid arthritis and systemic lupus erythematosus. Expert Rev Clin Immunol 2021; 17:1121-1130. [PMID: 34351835 DOI: 10.1080/1744666x.2021.1964957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The activation of autoreactive T- and B-cells and production of autoantibodies by B cells are involved in the pathogenesis of autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Recently, the concept of 'immunometabolism' has attracted significant attention. Immune cells produce large amounts of energy in the form of ATP and biosynthesize biological components such as nucleic acids and lipids via metabolic reprogramming to activate, differentiate, and exert their functions. AREAS COVERED While the mechanisms underlying the metabolism of CD4+ T cells in SLE have been extensively studied, the metabolic changes underlying B cell activation, differentiation, and function remain unclear. Drugs targeting mTOR and AMPK, such as sirolimus, rapamycin, and metformin, have shown some efficacy and tolerability in clinical trials on patients with SLE, but have not led to breakthroughs. In this review, we summarize the current knowledge on the immunometabolic mechanisms involved in SLE and RA and discuss the potential novel therapeutic drugs. EXPERT OPINION The intensity of activation of different immune cells and their metabolic kinetics vary in different autoimmune diseases; thus, understanding the disease- and cell-specific metabolic mechanisms may help in the development of clinically effective immunometabolism-targeting drugs.
Collapse
Affiliation(s)
- Shigeru Iwata
- The First Department of Internal Medicine, Assistant Professor, University of Occupational and Environmental Health, Japan, School of Medicine, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, Professor and Chairman, Deputy Director, University of Occupational and Environmental Health, Japan, the University Hospital, School of Medicine, Kitakyushu, Japan
| |
Collapse
|
45
|
Regulation of humoral immune response by HIF-1α-dependent metabolic reprogramming of the germinal center reaction. Cell Immunol 2021; 367:104409. [PMID: 34246872 DOI: 10.1016/j.cellimm.2021.104409] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/18/2021] [Accepted: 06/29/2021] [Indexed: 11/23/2022]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) has been implicated in the regulation of many genes responsible for aerobic glycolysis; however, the role of HIF-1α in B-cell metabolism has not been well defined. Here, we analyzed patterns of gene expression and oxygen consumption rates in B-cell subpopulations from humans and mice and described a model of HIF-1α-mediated B-cell metabolic reprogramming during the germinal center (GC) reaction. Importantly, we found that HIF-1α was highly expressed in GC B-cells, and HIF-1α deficiency in B-cells impaired a functional GC reaction, resulting in defective class-switch recombination and generation of high-affinity plasma cells. These results identified an important role of HIF-1α in regulating humoral immunity through metabolic reprogramming during the GC response. This newly discovered metabolic character of GC B-cells will advance our understanding of GC biology and B-cell lymphomagenesis.
Collapse
|
46
|
Abstract
B cells are central to the pathogenesis of multiple autoimmune diseases, through antigen presentation, cytokine secretion, and the production of autoantibodies. During development and differentiation, B cells undergo drastic changes in their physiology. It is emerging that these are accompanied by equally significant shifts in metabolic phenotype, which may themselves also drive and enforce the functional properties of the cell. The dysfunction of B cells during autoimmunity is characterised by the breaching of tolerogenic checkpoints, and there is developing evidence that the metabolic state of B cells may contribute to this. Determining the metabolic phenotype of B cells in autoimmunity is an area of active study, and is important because intervention by metabolism-altering therapeutic approaches may represent an attractive treatment target.
Collapse
Affiliation(s)
- Iwan G. A. Raza
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Alexander J. Clarke
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
47
|
Qi F, Zhang W, Huang J, Fu L, Zhao J. Single-Cell RNA Sequencing Analysis of the Immunometabolic Rewiring and Immunopathogenesis of Coronavirus Disease 2019. Front Immunol 2021; 12:651656. [PMID: 33936072 PMCID: PMC8079812 DOI: 10.3389/fimmu.2021.651656] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/29/2021] [Indexed: 12/16/2022] Open
Abstract
Although immune dysfunction is a key feature of coronavirus disease 2019 (COVID-19), the metabolism-related mechanisms remain elusive. Here, by reanalyzing single-cell RNA sequencing data, we delineated metabolic remodeling in peripheral blood mononuclear cells (PBMCs) to elucidate the metabolic mechanisms that may lead to the progression of severe COVID-19. After scoring the metabolism-related biological processes and signaling pathways, we found that mono-CD14+ cells expressed higher levels of glycolysis-related genes (PKM, LDHA and PKM) and PPP-related genes (PGD and TKT) in severe patients than in mild patients. These genes may contribute to the hyperinflammation in mono-CD14+ cells of patients with severe COVID-19. The mono-CD16+ cell population in COVID-19 patients showed reduced transcription levels of genes related to lysine degradation (NSD1, KMT2E, and SETD2) and elevated transcription levels of genes involved in OXPHOS (ATP6V1B2, ATP5A1, ATP5E, and ATP5B), which may inhibit M2-like polarization. Plasma cells also expressed higher levels of the OXPHOS gene ATP13A3 in COVID-19 patients, which was positively associated with antibody secretion and survival of PCs. Moreover, enhanced glycolysis or OXPHOS was positively associated with the differentiation of memory B cells into plasmablasts or plasma cells. This study comprehensively investigated the metabolic features of peripheral immune cells and revealed that metabolic changes exacerbated inflammation in monocytes and promoted antibody secretion and cell survival in PCs in COVID-19 patients, especially those with severe disease.
Collapse
Affiliation(s)
- Furong Qi
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China.,Shenzhen Research Center for Communicable Disease Diagnosis and Treatment of Chinese Academy of Medical Science, Shenzhen, China
| | - Wenbo Zhang
- Trinity School of Durham and Chapel Hill, Durham, NC, United States
| | - Jialu Huang
- Electronic and Computer Engineering, China North Vehicle Research Institute, Beijing, China
| | - Lili Fu
- Center for Life Sciences, Tsinghua University, Beijing, China
| | - Jinfang Zhao
- Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
48
|
Iwata S, Zhang M, Hajime M, Ohkubo N, Sonomoto K, Torimoto K, Kitanaga Y, Trimova G, Todoroki Y, Miyata H, Ueno M, Nagayasu A, Kanda R, Nakano K, Nakayamada S, Sakata K, Tanaka Y. Pathological role of activated mTOR in CXCR3+ memory B cells of rheumatoid arthritis. Rheumatology (Oxford) 2021; 60:5452-5462. [PMID: 33693564 DOI: 10.1093/rheumatology/keab229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/23/2021] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVES B cells play an important pathological role in rheumatoid arthritis (RA). In this study, we investigated the role of metabolic regulator mTOR in B cells and its relevance to the pathology of RA. METHODS Peripheral blood mononuclear cells were isolated from 31 normal subjects and 86 RA patients and the gated B cells were assessed for mTOR phosphorylation and chemokine receptor expression. In vitro studies on peripheral blood B cells isolated from the control and RA patients investigated the molecular mechanisms. RESULTS Higher concentrations of CXCL10 (CXCR3 ligands) and lower percentages of CXCR3+ memory B cells were present in the peripheral blood of RA patients relative to the control. RA patients with high CXCL10 concentrations had smaller percentage of CXCR3+ memory B cells and high disease activity. One-year treatment with TNF inhibitors increased the percentage of CXCR3+ memory B cells and reduced serum CXCL10 concentrations. mTOR phosphorylation in B cells was further enhanced in RA patients, compared to the control, and was selectively enhanced in CXCR3+ memory B cells. mTOR phosphorylation in CXCR3+ memory B cells correlated with disease activity. In vitro, mTOR phosphorylation in B cells enhanced IL-6 production and increased RANKL expression. CONCLUSION mTOR activation in CXCR3+ memory B cells of RA patients is associated with disease activity, mediated through IL-6 production and RANKL expression. The obtained results also suggest that TNF inhibitors mediate an impact on the association between CXCL10 and mTOR activated CXCR3+ memory B cells.
Collapse
Affiliation(s)
- Shigeru Iwata
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Mingzeng Zhang
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
- Department of Hematology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Maiko Hajime
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Naoaki Ohkubo
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Koshiro Sonomoto
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Keiichi Torimoto
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Yukihiro Kitanaga
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
- Astellas Pharma, Inc., Tsukuba, Japan
| | - Gulzhan Trimova
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
- Department of Clinical Subjects, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Yasuyuki Todoroki
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Hiroko Miyata
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Masanobu Ueno
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Atsushi Nagayasu
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Ryuichiro Kanda
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Kazuhisa Nakano
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Shingo Nakayamada
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Kei Sakata
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
- Mitsubishi Tanabe Pharma Corp, Yokohama, Kanagawa, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
49
|
Su YJ, Wang PW, Weng SW. The Role of Mitochondria in Immune-Cell-Mediated Tissue Regeneration and Ageing. Int J Mol Sci 2021; 22:2668. [PMID: 33800867 PMCID: PMC7961648 DOI: 10.3390/ijms22052668] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/03/2021] [Indexed: 12/25/2022] Open
Abstract
During tissue injury events, the innate immune system responds immediately to alarms sent from the injured cells, and the adaptive immune system subsequently joins in the inflammatory reaction. The control mechanism of each immune reaction relies on the orchestration of different types of T cells and the activators, antigen-presenting cells, co-stimulatory molecules, and cytokines. Mitochondria are an intracellular signaling organelle and energy plant, which supply the energy requirement of the immune system and maintain the system activation with the production of reactive oxygen species (ROS). Extracellular mitochondria can elicit regenerative effects or serve as an activator of the immune cells to eliminate the damaged cells. Recent clarification of the cytosolic escape of mitochondrial DNA triggering innate immunity underscores the pivotal role of mitochondria in inflammation-related diseases. Human mesenchymal stem cells could transfer mitochondria through nanotubular structures to defective mitochondrial DNA cells. In recent years, mitochondrial therapy has shown promise in treating heart ischemic events, Parkinson's disease, and fulminating hepatitis. Taken together, these results emphasize the emerging role of mitochondria in immune-cell-mediated tissue regeneration and ageing.
Collapse
Affiliation(s)
- Yu-Jih Su
- Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, 123, Dapi Road, Niaosong District, Kaohsiung 833, Taiwan; (Y.-J.S.); (P.-W.W.)
| | - Pei-Wen Wang
- Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, 123, Dapi Road, Niaosong District, Kaohsiung 833, Taiwan; (Y.-J.S.); (P.-W.W.)
| | - Shao-Wen Weng
- Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, 123, Dapi Road, Niaosong District, Kaohsiung 833, Taiwan; (Y.-J.S.); (P.-W.W.)
| |
Collapse
|
50
|
Abstract
A large and growing body of evidence supports functions of enzymes that regulate or effect cellular metabolism in governing the development, survival, and effector functions of immune cells—especially T cells, macrophages, and dendritic cells. Among these proteins, adenosine monophosphate-activated protein kinase (AMPK) is a conserved ATP and nutrient sensor that regulates multiple metabolic pathways to promote energy homeostasis. Although AMPK had been shown to regulate aspects of CD4+ and CD8+ T cell biology, its function in B lymphocytes has been less clear. Here, we review recent advances in our understanding of the role of AMPK in the metabolism, function, and maintenance of the B lineage.
Collapse
|