1
|
Shen X, Zhang F, Tang C, Soković M, Mišić D, Xu H, Ye Y, Liu J. Advances in Sampling and Analytical Techniques for Single-Cell Metabolomics: Exploring Cellular Heterogeneity. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2025; 39:e10045. [PMID: 40223194 DOI: 10.1002/rcm.10045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025]
Abstract
Single-cell metabolomics is an emerging and powerful technology that uncovers intercellular heterogeneity and reveals microenvironmental dynamics in both physiological and pathological conditions. This technology enables detailed observations of cellular interactions, providing valuable insights into processes such as aging, immune responses, and disease development. Despite significant advances, the need for detailed discussions on sampling and analytical methods in single-cell metabolomics continues to grow, with increasing focus on selecting the most suitable techniques for diverse research objectives. This review addresses these challenges by exploring key sampling and analytical strategies used in single-cell metabolomics. We focus on three main approaches: the capture and isolation of specific cell types, the precise aspiration of individual cells, and in situ mass spectrometry imaging. These methods are critically assessed to highlight strategies for achieving accurate metabolite detection at the single-cell level across diverse research applications.
Collapse
Affiliation(s)
- Xinxin Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fangyuan Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Chunping Tang
- China-Serbia "Belt and Road" Joint Laboratory for Natural Products and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Marina Soković
- China-Serbia "Belt and Road" Joint Laboratory for Natural Products and Drug Discovery, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Danijela Mišić
- China-Serbia "Belt and Road" Joint Laboratory for Natural Products and Drug Discovery, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Ye
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- China-Serbia "Belt and Road" Joint Laboratory for Natural Products and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jia Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
2
|
Oh JM, Guo T, Begum HM, Marty SE, Sha L, Kilic C, Zhou H, Dou Y, Shen K. A micro-metabolic rewiring assay for assessing hypoxia-associated cancer metabolic heterogeneity. Bioact Mater 2025; 48:493-509. [PMID: 40093303 PMCID: PMC11910375 DOI: 10.1016/j.bioactmat.2025.02.030] [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: 11/02/2024] [Revised: 01/11/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Cancer metabolism plays an essential role in therapeutic resistance, where significant inter- and intra-tumoral heterogeneity exists. Hypoxia is a prominent driver of metabolic rewiring behaviors and drug responses. Recapitulating the hypoxic landscape in the tumor microenvironment thus offers unique insights into heterogeneity in metabolic rewiring and therapeutic responses, to inform better treatment strategies. There remains a lack of scalable tools that can readily interface with imaging platforms and resolve the heterogeneous behaviors in hypoxia-associated metabolic rewiring. Here we present a micro-metabolic rewiring (μMeRe) assay that provides the scalability and resolution needed to characterize the metabolic rewiring behaviors of different cancer cells in the context of hypoxic solid tumors. Our assay generates hypoxia through cellular metabolism without external gas controls, enabling the characterization of cell-specific intrinsic ability to drive hypoxia and undergo metabolic rewiring. We further developed quantitative metrics that measure the metabolic plasticity through phenotypes and gene expression. As a proof-of-concept, we evaluated the efficacy of a metabolism-targeting strategy in mitigating hypoxia- and metabolic rewiring-induced chemotherapeutic resistance. Our study and the scalable platform thus lay the foundation for designing more effective cancer treatments tailored toward specific metabolic rewiring behaviors.
Collapse
Affiliation(s)
- Jeong Min Oh
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Tianze Guo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Hydari Masuma Begum
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Saci-Elodie Marty
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Liang Sha
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Cem Kilic
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Hao Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Yali Dou
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
- USC Stem Cell, University of Southern California, Los Angeles, CA, 90033, USA
| |
Collapse
|
3
|
Krug A, Ernst LM, Mhaidly R, Ramis J, Gusta MF, Bastus NG, Martinez-Turtos A, Tosolini M, Di Mascio L, Tari G, Boyer L, Gaulard P, Lemonnier F, Ricci JE, Verhoeyen E, Puntes V. Scavenging Reactive Oxygen Species by Cerium Oxide Nanoparticles Prevents Death in a Peripheral T Cell Lymphoma Preclinical Mouse Model. ACS NANO 2025; 19:18644-18660. [PMID: 40346022 DOI: 10.1021/acsnano.5c02860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Cancer cell survival and proliferation are correlated with increased metabolic activity and consequent oxidative stress, driving metabolic shifts that interfere with the immune response to malignant cells. This is the case of high-energy-demanding angioimmunoblastic T cell lymphoma (AITL), a highly aggressive cancer with poor survival rates, where malignant CD4+ PD-1high T cells show increased mitochondrial activity and Reactive oxygen species (ROS) accumulation. Here, we report that administration of ROS scavenging cerium oxide (CeO2) nanoparticles in an AITL preclinical mouse model leads to their preferential accumulation in the spleen, where the CD4+ PD-1high T cells driving malignancy were significantly reduced. This was accompanied by activation of previously exhausted cytotoxic CD8+ T cells, restoring their potent antitumor function. As a result, survival rates dramatically increase with no observed toxicity to healthy cells or tissues. Overall, it highlights the correlation between increased energy demand, increased mitochondrial mass, increased PD-1 expression, increased ROS production, and immune suppression and how this vicious loop can be stopped by scavenging ROS.
Collapse
Affiliation(s)
- Adrien Krug
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
| | - Lena M Ernst
- Vall d'Hebron Research Institute (VHIR), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona 08193, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona 08010, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Rana Mhaidly
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
| | - Joana Ramis
- Vall d'Hebron Research Institute (VHIR), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona 08193, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona 08010, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Muriel F Gusta
- Vall d'Hebron Research Institute (VHIR), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona 08193, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona 08010, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Neus G Bastus
- Vall d'Hebron Research Institute (VHIR), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona 08193, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona 08010, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | | | - Marie Tosolini
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse 31100, France
| | - Léa Di Mascio
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
| | - Gamze Tari
- INSERMU955; Unité hémopathies lymphoïdes, Hôpitaux Universitaires Henri Mondor, Assistance publique des Hôpitaux de Paris, Université Paris-Est Créteil; Institut Mondor de Recherche Biomédicale, Créteil F-94010, France
| | - Laurent Boyer
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
| | - Philippe Gaulard
- département de pathologie, AP-HP, Groupe hospitalo-universitaire Chenevier Mondor, Créteil F-94010, France
- Service Unité Hémopathies Lymphoides, AP-HP, Groupe hospitalo-universitaire Chenevier Mondor, Créteil F-94010, France
| | - François Lemonnier
- INSERMU955; Unité hémopathies lymphoïdes, Hôpitaux Universitaires Henri Mondor, Assistance publique des Hôpitaux de Paris, Université Paris-Est Créteil; Institut Mondor de Recherche Biomédicale, Créteil F-94010, France
| | - Jean-Ehrland Ricci
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
| | - Els Verhoeyen
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
- CIRI, Université de Lyon; INSERM U1111; ENS de Lyon; University Lyon1; CNRS UMR5308, Lyon 69007, France
| | - Victor Puntes
- Vall d'Hebron Research Institute (VHIR), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona 08193, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona 08010, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
4
|
Schimmer S, Kerkmann L, Kahlert N, Jubeh SA, Werner T, Corkish C, Prendeville H, Finlay DK, Sutter K, Dittmer U, Littwitz-Salomon E. Dietary lipid overload creates a suppressive environment that impedes the antiviral functions of NK cells. iScience 2025; 28:112396. [PMID: 40352719 PMCID: PMC12063142 DOI: 10.1016/j.isci.2025.112396] [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: 10/21/2024] [Revised: 02/03/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Natural killer (NK) cells are innate immune cells able to recognize and eliminate virus-infected cells. NK cell activity strongly correlates with a metabolic reprogramming and breakdown of fatty acids by β-oxidation during virus infections. However, there is limited knowledge regarding the impact of obesity on antiviral NK cell functions. Here, employing the Friend retrovirus mouse model, we show that the cytotoxicity and cytokine production of NK cells was impaired in obesity, leading to higher viral loads. NK cells suppression in obesity was mediated by activated Tregs. Furthermore, obese mice that were switched back to a regular diet showed complete recovery of the NK cell activity. Interestingly, feeding mice with a high-fat diet (HFD) for just ten days caused NK cell dysfunction and increased retroviral burden. This study is the first to link the detrimental impact of an obesity-induced immunosuppressive microenvironment with NK cell dysfunction during an acute retroviral infection.
Collapse
Affiliation(s)
- Simone Schimmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Leonie Kerkmann
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Nele Kahlert
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Shahd al Jubeh
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tanja Werner
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carrie Corkish
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Hannah Prendeville
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
| | - David K. Finlay
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Elisabeth Littwitz-Salomon
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
5
|
Duan Q, Wang J, Sun L, Chen Z, Li W, Liu X, Zhang A, Liu Y, Zhang L. Malonate promotes CD8 + T cell memory formation via protein malonylation. Cell Mol Immunol 2025:10.1038/s41423-025-01294-7. [PMID: 40369188 DOI: 10.1038/s41423-025-01294-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
Protein malonylation represents a recently identified posttranslational modification whose role in CD8+ T cell differentiation and functionality remains incompletely understood. In this study, we demonstrate that enhancing protein malonylation through sodium malonate (SM) treatment promotes CD8+ T cell memory formation in response to bacterial infection, subsequently potentiating recall responses. Comparative metabolomic analysis between SM-treated and control CD8+ T cells revealed significant metabolic alterations associated with protein malonylation. We present the first comprehensive proteomic analysis of lysine malonylation in murine CD8+ T cells, identifying 77 malonylation sites across 64 proteins involved in diverse cellular processes, particularly metabolic pathways. Malonylation of STAT6 was confirmed via the use of a specific chemical probe. Notably, we established that malonylation at the lysine 374 site of STAT6 results in increased TCF1 expression, due to alleviated transcriptional repression of TCF1 by STAT6. Collectively, our findings provide compelling evidence that protein malonylation plays a significant role in regulating CD8+ T cell memory formation.
Collapse
Affiliation(s)
- Qianqian Duan
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Jiajia Wang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China
| | - Liang Sun
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Zihan Chen
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Wenhui Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China
| | - Xiaowei Liu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China
| | - Aijun Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
| | - Yong Liu
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| | - Lianjun Zhang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China.
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
6
|
Kao YS, Lauterbach M, Lopez Krol A, Distler U, Godoy GJ, Klein M, Argüello RJ, Boukhallouk F, Vallejo Fuente S, Braband KL, Nurbekova A, Romero M, Mamareli P, Silva L, Damasceno LEA, Rampoldi F, Berod L, Lynch L, Hiller K, Sparwasser T. Metabolic reprogramming of interleukin-17-producing γδ T cells promotes ACC1-mediated de novo lipogenesis under psoriatic conditions. Nat Metab 2025:10.1038/s42255-025-01276-z. [PMID: 40360755 DOI: 10.1038/s42255-025-01276-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 03/13/2025] [Indexed: 05/15/2025]
Abstract
Metabolic reprogramming determines γδ T cell fate during thymic development; however, the metabolic requirements of interleukin (IL)-17A-producing γδ T cells (γδT17 cells) under psoriatic conditions are unclear. Combining high-throughput techniques, including RNA sequencing, SCENITH, proteomics and stable isotope tracing, we demonstrated that psoriatic inflammation caused γδT17 cells to switch toward aerobic glycolysis. Under psoriatic conditions, γδT17 cells upregulated ATP-citrate synthase to convert citrate to acetyl-CoA, linking carbohydrate metabolism and fatty acid synthesis (FAS). Accordingly, we used a pharmacological inhibitor, Soraphen A, which blocks acetyl-CoA carboxylase (ACC), to impair FAS in γδT17 cells, reducing their intracellular lipid stores and ability to produce IL-17A under psoriatic conditions in vitro. We pinpointed the pathogenic role of ACC1 in γδT17 cells in vivo by genetic ablation, ameliorating inflammation in a psoriatic mouse model. Furthermore, ACC inhibition limited human IL-17A-producing γδT17 cells. Targeting ACC1 to attenuate pathogenic γδT17 cell function has important implications for psoriasis management.
Collapse
Affiliation(s)
- Yu-San Kao
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA.
| | - Mario Lauterbach
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Aleksandra Lopez Krol
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ute Distler
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Gloria Janet Godoy
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Klein
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Rafael Jose Argüello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Fatima Boukhallouk
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sara Vallejo Fuente
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kathrin Luise Braband
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Assel Nurbekova
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Monica Romero
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Panagiota Mamareli
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Luana Silva
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Luis Eduardo Alves Damasceno
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, São Paulo, Brazil
| | - Francesca Rampoldi
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- DKFZ German Cancer Research Center, Heidelberg, Germany
| | - Luciana Berod
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lydia Lynch
- Molecular Biology, Princeton University, Princeton, NJ, USA
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
7
|
Slattery K, Yao CH, Mylod E, Scanlan J, Scott B, Crowley JP, McGowan O, McManus G, Brennan M, O'Brien K, Glennon K, Corry E, Treacy A, Argüello RJ, Gardiner CM, Haigis MC, Brennan DJ, Lynch L. Uptake of lipids from ascites drives NK cell metabolic dysfunction in ovarian cancer. Sci Immunol 2025; 10:eadr4795. [PMID: 40344087 DOI: 10.1126/sciimmunol.adr4795] [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: 07/02/2024] [Revised: 11/26/2024] [Accepted: 04/16/2025] [Indexed: 05/11/2025]
Abstract
High-grade serous ovarian cancer (HGSOC) remains an urgent unmet clinical need, with more than 70% of patients presenting with metastatic disease. Many patients develop large volumes of ascites, which promotes metastasis and is associated with poor therapeutic response and survival. Immunotherapy trials have shown limited success, highlighting the need to better understand HGSOC immunology. Here, we analyzed cytotoxic lymphocytes [natural killer (NK), T, and innate T cells] from patients with HGSOC and observed widespread dysfunction across primary and metastatic sites. Although nutrient rich, ascites was immunosuppressive for all lymphocyte subsets. NK cell dysfunction was driven by uptake of polar lipids, with associated dysregulation in lipid storage. Phosphatidylcholine was a key immunosuppressive metabolite, disrupting NK cell membrane order and cytotoxicity. Blocking lipid uptake through SR-B1 protected NK cell antitumor functions in ascites. These findings offer insights into immune suppression in HGSOC and have important implications for the design of future immunotherapies.
Collapse
Affiliation(s)
- Karen Slattery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Cong-Hui Yao
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Eimear Mylod
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - John Scanlan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Barry Scott
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Joseph Patrick Crowley
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Orla McGowan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Gavin McManus
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Martin Brennan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Katie O'Brien
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kate Glennon
- UCD-Gynaecological Oncology Group, School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | - Edward Corry
- UCD-Gynaecological Oncology Group, School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | - Ann Treacy
- UCD-Gynaecological Oncology Group, School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | - Rafael J Argüello
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Clair M Gardiner
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Donal J Brennan
- UCD-Gynaecological Oncology Group, School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
- Systems Biology Ireland, UCD School of Medicine, Belfield, Dublin, Ireland
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| |
Collapse
|
8
|
Corda PO, Silva JV, Almeida CR, Pierre P, Fardilha M. De Novo Protein Synthesis Occurs Through the Cytoplasmic Translation Machinery in Mammalian Spermatozoa. J Cell Physiol 2025; 240:e70038. [PMID: 40373039 DOI: 10.1002/jcp.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/02/2025] [Accepted: 04/10/2025] [Indexed: 05/17/2025]
Abstract
The current hypothesis suggests that translation occurs in capacitated spermatozoa through mitochondrial ribosomes. Mitochondrial translation has several particularities, which rise some questions about how mitochondrial ribosomes can ensure sperm translation activity. Here, we aimed to elucidate if cytoplasmic translation occurs in mammalian spermatozoa. A bioinformatic workflow was performed to identify translation-related proteins in human spermatozoa and their association with cytoplasmic translation. The surface sensing of translation (SUnSET) method was used to measure translation activity in capacitated human and bovine spermatozoa. Two translation inhibitors, cycloheximide (CHX, cytoplasmic) and D-chloramphenicol (D-CP, mitochondrial) were used to identify which ribosomes were active in sperm. To spot newly synthesized proteins, puromycin-peptides were immunoprecipitated and analysed by mass spectrometry. A second approach was performed using translation inhibitors and analysing the sperm proteome by mass spectrometry. Bioinformatic analysis revealed that human spermatozoa possess 510 translation proteins, which were enriched for cytoplasmic mRNA translation. CHX decreased translation activity in mammalian sperm, whereas no effect was observed after D-CP treatment. Nine proteins were immunoprecipitated and identified as newly synthesized in capacitated bovine spermatozoa. CHX and D-CP decreased the level of 22 proteins that were replaced, or de novo translated during capacitation. New proteins were associated with relevant processes for sperm physiology. Both translation inhibitors decreased sperm rapid progressive motility and increased sperm immotility. Our results proved sperm translation occurs through cytoplasmic translation machinery in capacitated bovine and human spermatozoa. These results also support that sperm translation is required during capacitation to produce relevant proteins for sperm functions.
Collapse
Affiliation(s)
- Pedro O Corda
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Aveiro, Portugal
| | - Joana Vieira Silva
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Aveiro, Portugal
| | - Catarina R Almeida
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Aveiro, Portugal
| | - Philippe Pierre
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Aveiro, Portugal
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, Bouches-du-Rhône, France
| | - Margarida Fardilha
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Aveiro, Portugal
| |
Collapse
|
9
|
Haring E, Buescher JM, Apostolova P. Metabolism in hematology: Technological advances open new perspectives on disease biology and treatment. Hemasphere 2025; 9:e70134. [PMID: 40390870 PMCID: PMC12086526 DOI: 10.1002/hem3.70134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 05/21/2025] Open
Abstract
The term metabolism refers to the multi-faceted biochemical reactions within a cell or an organism that occur to maintain energy homeostasis, cell growth, and oxidative balance. Cells possess a high metabolic plasticity, allowing them to adapt to the dynamic requirements of their functional state and environment. Deregulated cellular metabolism is a hallmark of many diseases, including benign and malignant hematological conditions. In the last decade, multiple technological innovations in the metabolism field have made in-depth metabolic analysis broadly applicable. Such studies are shedding new light on normal and malignant hematopoiesis and open avenues to a better understanding of the biology of hematological diseases. In this review, we will first give a brief overview of central metabolic processes. Furthermore, we discuss the most commonly used methods to study metabolism. We begin by elaborating on the use of next-generation sequencing to detect metabolism-related genomic mutations and study transcriptional signatures. Furthermore, we discuss methods for measuring protein expression, such as mass spectrometry (MS), flow cytometry, and cytometry time-of-flight. Next, we describe the use of nuclear magnetic resonance spectroscopy, MS, and flow cytometry for metabolite quantification. Finally, we highlight functional assays to probe metabolic pathways in real-time. We illustrate how these technologies and their combination have advanced our understanding of the role of metabolism. Our goal is to provide hematologists with a comprehensive guide to modern techniques in metabolism research, their benefits and disadvantages, and how they guide our understanding of disease and potentially future personalized therapy decisions.
Collapse
Affiliation(s)
- Eileen Haring
- Department of BiomedicineUniversity Hospital Basel, University of BaselBaselSwitzerland
| | - Joerg M. Buescher
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Petya Apostolova
- Department of BiomedicineUniversity Hospital Basel, University of BaselBaselSwitzerland
- Division of HematologyUniversity Hospital BaselBaselSwitzerland
| |
Collapse
|
10
|
Huang Y, Xie X, Liu M, Zhang Y, Yang J, Yang W, Hu Y, Qi S, Feng Y, Liu G, Lu S, Peng X, Ye J, Ma S, Sun J, Wang L, Hu L, Wang L, Zhu X, Cheng H, Sun Z, Chen J, Dong F, Zhang Y, Cheng T. Restoring mitochondrial function promotes hematopoietic reconstitution from cord blood following cryopreservation-related functional decline. J Clin Invest 2025; 135:e183607. [PMID: 40036065 PMCID: PMC12043090 DOI: 10.1172/jci183607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 02/27/2025] [Indexed: 03/06/2025] Open
Abstract
Umbilical cord blood (UCB) plays substantial roles in hematopoietic stem cell (HSC) transplantation and regenerative medicine. UCB is usually cryopreserved for years before use. It remains unclear whether and how cryopreservation affects UCB function. We constructed a single-cell transcriptomics profile of CD34+ hematopoietic stem and progenitor cells (HSPCs) and mononuclear cells (MNCs) from fresh and cryopreserved UCB stored for 1, 5, 10, and 19 years. Compared with fresh UCB, cryopreserved HSCs and multipotent progenitors (MPPs) exhibited more active cell-cycle and lower expression levels of HSC and multipotent progenitor signature genes. Hematopoietic reconstitution of cryopreserved HSPCs gradually decreased during the first 5 years but stabilized thereafter, aligning with the negative correlation between clinical neutrophil engraftment and cryopreservation duration of UCB. Cryopreserved HSPCs also showed reduced megakaryocyte generation. In contrast, cryopreserved NK cells and T cells maintained a capacity for cytokine production and cytotoxicity comparable to that of fresh cells. Mechanistically, cryopreserved HSPCs exhibited elevated ROS, reduced ATP synthesis, and abnormal mitochondrial distribution, which collectively led to attenuated hematopoietic reconstitution. These effects could be ameliorated by sulforaphane (SF). Together, we elucidate the negative effect of cryopreservation on UCB HSPCs and identify SF as a mitigation strategy, broadening the temporal window and scope for clinical applications of cryopreserved UCB.
Collapse
Affiliation(s)
- Yaojin Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaowei Xie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mengyao Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yawen Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Junye Yang
- Tianjin Cord Blood Stem Cell Bank, Tianjin, China
- Union Stem Cell Genetic Engineering Co., Ltd., Tianjin, China
| | - Wenling Yang
- Tianjin Cord Blood Stem Cell Bank, Tianjin, China
- Union Stem Cell Genetic Engineering Co., Ltd., Tianjin, China
| | - Yu Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Saibing Qi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yahui Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Guojun Liu
- Shandong Qilu Stem Cell Engineering Co., Ltd., Jinan, China
| | - Shihong Lu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xuemei Peng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jinhui Ye
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shihui Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jiali Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Linping Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lin Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zimin Sun
- Department of Hematology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, China
| | - Junren Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Fang Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yingchi Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of the Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
11
|
Woodworth KE, Froom ZSCS, Osborne ND, Rempe CN, Wheeler B, Medd K, Callaghan NI, Qian H, Acharya AP, Charron C, Davenport Huyer L. Development of Itaconate Polymers Microparticles for Intracellular Regulation of Pro-Inflammatory Macrophage Activation. Adv Healthc Mater 2025; 14:e2405257. [PMID: 40183748 DOI: 10.1002/adhm.202405257] [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: 12/27/2024] [Revised: 03/12/2025] [Indexed: 04/05/2025]
Abstract
Itaconate (IA) is an endogenous metabolite and a potent regulator of the innate immune system. It's use in immunomodulatory therapies has faced limitations due to challenges in controlled delivery and requirements of high extracellular concentrations for internalization of the highly polar small molecule to achieve its intracellular therapeutic activity. Microparticle (MP)-based delivery strategies are a promising approach for intracellular delivery of small molecule metabolites through macrophage phagocytosis and subsequent intracellular polymer degradation-based delivery. Toward the goal of intracellular delivery of IA, degradable polyester polymer- (poly(dodecyl itaconate)) based IA polymer microparticles (IA-MPs) are generated using an emulsion method, forming micron-scale (≈1.5 µm) degradable microspheres. IA-MPs are characterized with respect to their material properties and IA release kinetics to inform particle fabrication. Treatment of murine bone marrow-derived macrophages with an optimized particle concentration of 0.1 mg million-1 cells enables phagocytosis-mediated internalization and low levels of cytotoxicity. Flow cytometry demonstrates IA-MP-specific regulation of IA-sensitive inflammatory targets. Metabolic analyses demonstrate that IA-MP internalization inhibits oxidative metabolism and induced glycolytic reliance, consistent with the established mechanism of IA-associated inhibition of succinate dehydrogenase. This development of IA-based polymer microparticles provides a basis for additional innovative metabolite-based microparticle drug delivery systems for the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Kaitlyn E Woodworth
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Natasha D Osborne
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Christian N Rempe
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Brenden Wheeler
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Kyle Medd
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Neal I Callaghan
- Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Huikang Qian
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Abhinav P Acharya
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Carlie Charron
- Department of Chemistry, Faculty of Science, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Department of Surgery, Nova Scotia Health, Halifax, NS, B3H 4R2, Canada
| |
Collapse
|
12
|
Ogger PP, Murray PJ. Dissecting inflammation in the immunemetabolomic era. Cell Mol Life Sci 2025; 82:182. [PMID: 40293552 PMCID: PMC12037969 DOI: 10.1007/s00018-025-05715-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Abstract
The role of immune metabolism, specific metabolites and cell-intrinsic and -extrinsic metabolic states across the time course of an inflammatory response are emerging knowledge. Targeted and untargeted metabolomic analysis is essential to understand how immune cells adapt their metabolic program throughout an immune response. In addition, metabolomic analysis can aid to identify pathophysiological patterns in inflammatory disease. Here, we discuss new metabolomic findings within the transition from inflammation to resolution, focusing on three key programs of immunity: Efferocytosis, IL-10 signaling and trained immunity. Particularly the tryptophan-derived metabolite kynurenine was identified as essential for efferocytosis and inflammation resolution as well as a potential biomarker in diverse inflammatory conditions. In summary, metabolomic analysis and integration with transcriptomic and proteomic data, high resolution imaging and spatial information is key to unravel metabolic drivers and dependencies during inflammation and progression to tissue-repair.
Collapse
Affiliation(s)
- Patricia P Ogger
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Peter J Murray
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| |
Collapse
|
13
|
Ziogas A, Novakovic B, Ventriglia L, Galang N, Tran KA, Li W, Matzaraki V, van Unen N, Schlüter T, Ferreira AV, Moorlag SJCFM, Koeken VACM, Moyo M, Li X, Baltissen MPA, Martens JHA, Li Y, Divangahi M, Joosten LAB, Mhlanga MM, Netea MG. Long-term histone lactylation connects metabolic and epigenetic rewiring in innate immune memory. Cell 2025:S0092-8674(25)00400-3. [PMID: 40318634 DOI: 10.1016/j.cell.2025.03.048] [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: 04/26/2024] [Revised: 01/08/2025] [Accepted: 03/31/2025] [Indexed: 05/07/2025]
Abstract
Trained immunity, a de facto innate immune memory characterized by enhanced responsiveness to future challenges, is underpinned by epigenetic and metabolic rewiring. In individuals vaccinated with Bacille Calmette-Guérin (BCG), lactate release was associated with enhanced cytokine responsiveness upon restimulation. Trained monocytes/macrophages are characterized by lactylation of histone H3 at lysine residue 18(H3K18la), mainly at distal regulatory regions. Histone lactylation was positively associated with active chromatin and gene transcription, persisted after the elimination of the training stimulus, and was strongly associated with "trained" gene transcription in response to a secondary stimulus. Increased lactate production upon induction of trained immunity led to enhanced production of proinflammatory cytokines, a process associated with histone lactylation. Pharmacological inhibition of lactate production or histone lactylation blocked trained immunity responses, while polymorphisms of LDHA and EP300 genes modulated trained immunity. Long-term histone lactylation persisted in vivo 90 days after vaccination with BCG, highlighting H3K18la as an epigenetic mark of innate immune memory.
Collapse
Affiliation(s)
- Athanasios Ziogas
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands.
| | - Boris Novakovic
- Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Australia
| | - Lorenzo Ventriglia
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Hepatogastroenterology Division, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Noriko Galang
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Kim A Tran
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - Wenchao Li
- Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Vasiliki Matzaraki
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nienke van Unen
- Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Titus Schlüter
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anaísa V Ferreira
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Simone J C F M Moorlag
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Valerie A C M Koeken
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Research Centre Innovations in Care, Rotterdam University of Applied Sciences, Rotterdam, the Netherlands
| | - Mthabisi Moyo
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Xiaolin Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marijke P A Baltissen
- Department of Molecular Biology, Faculty of Science, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Yang Li
- Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hanover Medical School, Hannover, Germany; Lower Saxony center for artificial intelligence and causal methods in medicine (CAIMed), Hannover, Germany
| | - Maziar Divangahi
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Musa M Mhlanga
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany.
| |
Collapse
|
14
|
Roubanis A, Hilaire M, Le Teuff M, Devergne O, Sparwasser T, Berod L, Salomon BL. A new method to measure cell metabolism of rare cells in vivo reveals a high oxidative phosphorylation dependence of lung T cells. Immunol Cell Biol 2025. [PMID: 40268295 DOI: 10.1111/imcb.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/28/2025] [Accepted: 03/09/2025] [Indexed: 04/25/2025]
Abstract
Regulation of cellular metabolism is a central element governing the fate and function of T cells. However, the in vivo metabolic characteristics of rare cells, such as nonlymphoid tissue T cells, are poorly understood because of experimental limitations. Most techniques measuring cell metabolism require large cell numbers. The recent SCENITH method allows for studying the metabolism of rare cells by flow cytometry. However, this technique requires cells to be isolated and cultured ex vivo, which may alter their metabolism. Here, we propose a new experimental approach, called in vivo SCENITH, to investigate the cellular metabolism of T cells in vivo at a steady state in the spleen and lungs. For this purpose, we administered the metabolic modulators directly in mice, instead of applying these reagents ex vivo, as in the classical SCENITH method. Whereas ex vivo manipulation impacted the viability and phenotype of T cells, this toxic effect was not observed in the in vivo SCENITH. We observed that conventional and regulatory T cells shared similar metabolic profiles. Importantly, whereas spleen T cells used both oxidative phosphorylation and glycolysis, the metabolism of T cells in the lungs was mainly based on oxidative phosphorylation. Finally, metabolic inhibitors that interfere with protein translation and energy availability downregulated Foxp3 expression in regulatory T cells. These results describe an expansion of SCENITH that allows to measure the metabolic profile of rare cells in vivo, revealing a high dependence on oxidative phosphorylation of lung T cells.
Collapse
Affiliation(s)
- Aristeidis Roubanis
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), INSERM, CNRS, Sorbonne Université, Paris, France
| | - Morgane Hilaire
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), INSERM, CNRS, Sorbonne Université, Paris, France
| | - Morgane Le Teuff
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM - CNRS - University Toulouse III, Toulouse, France
| | - Odile Devergne
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), INSERM, CNRS, Sorbonne Université, Paris, France
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene and Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Luciana Berod
- Institute of Medical Microbiology and Hygiene and Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Benoît L Salomon
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), INSERM, CNRS, Sorbonne Université, Paris, France
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM - CNRS - University Toulouse III, Toulouse, France
| |
Collapse
|
15
|
Herbst M, Köksal H, Brunn S, Zanetti D, Domocos I, De Stefani V, Gatti M, Vivalda F, Pereira P, Nater M, Cecconi V, Sartori AA, van den Broek M. Cancer-cell-derived cGAMP limits the activity of tumor-associated CD8 + T cells. Cell Rep 2025; 44:115510. [PMID: 40178978 DOI: 10.1016/j.celrep.2025.115510] [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: 11/05/2024] [Revised: 02/19/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Using a mouse tumor model with inducible cancer-cell-intrinsic cyclic GMP-AMP (cGAMP) synthase (cGAS) expression, we show that cancer-cell-derived cGAMP is essential and sufficient to trigger a sustained type I interferon response within the tumor microenvironment. This leads to improved CD8+ T cell-dependent tumor restriction. However, cGAMP limits the proliferation, survival, and function of stimulator of IFN genes (STING)-expressing, but not of STING-deficient, CD8+ T cells. In vivo, STING deficiency in CD8+ T cells enhances tumor restriction. Consequently, cancer-cell-derived cGAMP both drives and limits the anti-tumor potential of CD8+ T cells. Mechanistically, T cell-intrinsic STING is associated with pro-apoptotic and antiproliferative gene signatures. Our findings suggest that STING signaling acts as a checkpoint in CD8+ T cells that balances tumor immunity.
Collapse
Affiliation(s)
- Michael Herbst
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Hakan Köksal
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Silvan Brunn
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Dominik Zanetti
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Ioana Domocos
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Viola De Stefani
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Marco Gatti
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Francesca Vivalda
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Paulo Pereira
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Marc Nater
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Alessandro A Sartori
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
16
|
Suen TK, Al B, Ulas T, Reusch N, Bahrar H, Bekkering S, Bhat J, Kabelitz D, Schultze JL, van de Veerdonk FL, van Lennep JR, Riksen NP, Joosten LAB, Netea MG, Placek K. Human γδ T Cell Function Is Impaired Upon Mevalonate Pathway Inhibition. Immunology 2025. [PMID: 40264329 DOI: 10.1111/imm.13931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/17/2025] [Accepted: 04/06/2025] [Indexed: 04/24/2025] Open
Abstract
Vδ2 T cells, a predominant human peripheral γδ T cell population, are a promising candidate for the development of immunotherapies against cancer and infected cells. Aminobisphosphonate drugs, such as zoledronate, are commonly used to expand Vδ2 T cells. Yet, such in vitro generated cells have limited efficacy in the clinic. We found that despite inducing excessive proliferation of Vδ2 T cells, zoledronate impaired their effector function and caused the upregulation of the inhibitory receptor TIM3. This effect was due to the inhibition of mevalonate metabolism and dysregulation of downstream biological processes such as protein prenylation and intracellular signalling. In vitro and in vivo inhibition of mevalonate metabolism with zoledronate, statins, and 6-fluoromevalonate, as well as genetic deficiency of the mevalonate kinase, all resulted in compromised cytokine and cytotoxic molecule production by Vδ2 T cells. Impaired Vδ2 T cell function was accompanied by transcriptome and kinome changes. Our findings reveal the importance of mevalonate metabolism for the proper functioning of Vδ2 T cells. This observation provides important considerations for improving their therapeutic use and has repercussions for patients with statin or aminobisphosphonate treatments.
Collapse
Affiliation(s)
- Tsz Kin Suen
- Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Burcu Al
- Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Thomas Ulas
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics at the DZNE and the University of Bonn, Bonn, Germany
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Nico Reusch
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics at the DZNE and the University of Bonn, Bonn, Germany
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Harsh Bahrar
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jaydeep Bhat
- Institute of Immunology, University of Kiel, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dieter Kabelitz
- Institute of Immunology, University of Kiel, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Joachim L Schultze
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics at the DZNE and the University of Bonn, Bonn, Germany
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Frank L van de Veerdonk
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Katarzyna Placek
- Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
17
|
Fonseca-Pereira D, Bae S, Clay SL, Michaud M, MacDonald MH, Glickman JN, Garrett WS. The metabolic sensor LKB1 regulates ILC3 homeostasis and mitochondrial function. Cell Rep 2025; 44:115456. [PMID: 40120107 DOI: 10.1016/j.celrep.2025.115456] [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: 07/19/2024] [Revised: 02/06/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are tissue-resident cells that sense environmental cues, control infections, and promote tissue homeostasis at mucosal surfaces. The metabolic sensor liver kinase B1 (LKB1) integrates intracellular stress, metabolism, and mitochondrial function to promote the development and effector functions of a variety of immune cells; however, the role of LKB1 in ILC3 function was unknown. Here, we show that LKB1 is crucial for adult ILC3 homeostasis, cytokine production, and mitochondrial function. ILC3-specific LKB1 deletion resulted in a reduced number of ILC3s and interleukin-22 (IL-22) production. LKB1-deficient ILC3s had decreased survival, mitochondrial dysfunction, cytoplasmic lipid accumulation, and altered bioenergetics. Using LKB1 downstream kinase modulators, we found that LKB1 regulation of ILC3 survival and IL-22 production requires signaling through microtubule affinity-regulating kinases (MARKs). Mechanistically, LKB1 deficiency resulted in increased reactive oxygen species (ROS) production and NFAT2 and PD-1 expression. Our work reveals that metabolic regulation of enteric ILC3 function by an LKB1-dependent signaling network is crucial for intestinal immunity and tissue homeostasis.
Collapse
Affiliation(s)
- Diogo Fonseca-Pereira
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Sena Bae
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Slater L Clay
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Monia Michaud
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Meghan H MacDonald
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Jonathan N Glickman
- Gastrointestinal Pathology, Massachusetts General Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA; Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Medical Sciences, Harvard Medical School, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
18
|
Willett BAS, Thompson SB, Chen V, Dareshouri A, Jackson CL, Brunetti T, D'Alessandro A, Klarquist J, Nemkov T, Kedl RM. Mitochondrial protein OPA1 is required for the expansion of effector CD8 T cells. Cell Rep 2025; 44:115610. [PMID: 40261796 DOI: 10.1016/j.celrep.2025.115610] [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: 02/05/2025] [Revised: 03/14/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025] Open
Abstract
Short-lived effector cells are characterized metabolically by a highly glycolytic state, driving energy and biomass acquisition, whereas memory-fated T cells have historically been described as meeting these requirements through mitochondrial metabolism. Here, we show that the mitochondrial protein optic atrophy 1 (OPA1) is critical for rapidly dividing CD8 T cells in vivo, the requirement for which is most pronounced in effector CD8 T cells. More specifically, OPA1 supports proper cell cycle initiation and progression and the viability and survival of CD8 T cells during clonal expansion. Use of mice deficient in the mitochondrial membrane fusion proteins Mitofusin 1 and 2 (MFN1/2) in both in vivo proliferation/differentiation assays and ex vivo metabolic analysis indicates that the requirement for OPA1 during cell division supersedes its role in mitochondrial fusion. We conclude that OPA1 is critical for the generation and accumulation of short-lived effector cells that arise during the response to infection.
Collapse
Affiliation(s)
- Benjamin A S Willett
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Scott B Thompson
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vincent Chen
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anza Dareshouri
- Department of Cell and Developmental Biology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Conner L Jackson
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tonya Brunetti
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry & Molecular Genetics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jared Klarquist
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Travis Nemkov
- Department of Biochemistry & Molecular Genetics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
19
|
Young KA, Hosseini M, Mistry JJ, Morganti C, Mills TS, Cai X, James BT, Nye GJ, Fournier NR, Voisin V, Chegini A, Schimmer AD, Bader GD, Egan G, Mansour MR, Challen GA, Pietras EM, Fisher-Wellman KH, Ito K, Chan SM, Trowbridge JJ. Elevated mitochondrial membrane potential is a therapeutic vulnerability in Dnmt3a-mutant clonal hematopoiesis. Nat Commun 2025; 16:3306. [PMID: 40240771 PMCID: PMC12003737 DOI: 10.1038/s41467-025-57238-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/14/2025] [Indexed: 04/18/2025] Open
Abstract
The competitive advantage of mutant hematopoietic stem and progenitor cells (HSPCs) underlies clonal hematopoiesis (CH). Drivers of CH include aging and inflammation; however, how CH-mutant cells gain a selective advantage in these contexts is an unresolved question. Using a murine model of CH (Dnmt3aR878H/+), we discover that mutant HSPCs sustain elevated mitochondrial respiration which is associated with their resistance to aging-related changes in the bone marrow microenvironment. Mutant HSPCs have DNA hypomethylation and increased expression of oxidative phosphorylation gene signatures, increased functional oxidative phosphorylation capacity, high mitochondrial membrane potential (Δψm), and greater dependence on mitochondrial respiration compared to wild-type HSPCs. Exploiting the elevated Δψm of mutant HSPCs, long-chain alkyl-TPP molecules (MitoQ, d-TPP) selectively accumulate in the mitochondria and cause reduced mitochondrial respiration, mitochondrial-driven apoptosis and ablate the competitive advantage of HSPCs ex vivo and in vivo in aged recipient mice. Further, MitoQ targets elevated mitochondrial respiration and the selective advantage of human DNMT3A-knockdown HSPCs, supporting species conservation. These data suggest that mitochondrial activity is a targetable mechanism by which CH-mutant HSPCs gain a selective advantage over wild-type HSPCs.
Collapse
Affiliation(s)
| | - Mohsen Hosseini
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Claudia Morganti
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Departments of Cell Biology, Oncology and Medicine, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Taylor S Mills
- Division of Hematology, Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | | | | | | | | | - Veronique Voisin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada
| | - Ali Chegini
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Grace Egan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Marc R Mansour
- UCL Cancer Institute, Department of Developmental Biology and Cancer, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Grant A Challen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric M Pietras
- Division of Hematology, Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Kelsey H Fisher-Wellman
- East Carolina University, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, Department of Physiology, Greenville, NC, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Departments of Cell Biology, Oncology and Medicine, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Steven M Chan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | |
Collapse
|
20
|
Nour MA, Rajabivahid M, Mehdi MSS, Tahmasebi S, Dashtgol SN, Dehghani-Ghorbi M, Vanan AG, Ghorbaninezhad F. A new era in melanoma immunotherapy: focus on DCs metabolic reprogramming. Cancer Cell Int 2025; 25:149. [PMID: 40234886 PMCID: PMC12001691 DOI: 10.1186/s12935-025-03781-3] [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: 12/11/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
Melanoma, being one of the most dangerous forms of skin cancer, is characterized by its aggressive and metastatic nature, with the potential to develop resistance to various treatments. This resistance makes the disease challenging to treat, emphasizing the need for new treatment strategies. Within the tumor microenvironment (TME), melanoma cells exploit metabolic shifts, particularly glycolysis, to create an immunosuppressive TME that prevents dendritic cells (DCs) from functioning properly. Essential metabolic alterations such as lactate and lipid accumulation, and lack of tryptophan disrupt DC maturation, antigen presentation, and T cell activation. In recent years, melanoma immunotherapy has increasingly focused on reprogramming the metabolism of DCs. This review paper aims to provide insights into the metabolic suppression of melanoma-associated DCs, allowing the design of therapeutic strategies based on metabolic interventions to promote or restore DC function. This contribution reviews the metabolic reprogramming of DCs as a new approach for melanoma immunotherapy.
Collapse
Affiliation(s)
- Mina Afrashteh Nour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mansour Rajabivahid
- Department of Internal Medicine, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Marjan Sadat Seyed Mehdi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Nasirzadeh Dashtgol
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Mahmoud Dehghani-Ghorbi
- Hematology-Oncology Department, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farid Ghorbaninezhad
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Behan-Bush RM, Schrodt MV, Kilburg E, Liszewski JN, Bitterlich LM, English K, Klingelhutz AJ, Ankrum JA. Polychlorinated biphenyls induce immunometabolic switch of antiinflammatory macrophages toward an inflammatory phenotype. PNAS NEXUS 2025; 4:pgaf100. [PMID: 40191133 PMCID: PMC11969150 DOI: 10.1093/pnasnexus/pgaf100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/28/2025] [Indexed: 04/09/2025]
Abstract
Polychlorinated biphenyls (PCBs) are a group of environmental toxicants associated with increased risk of diabetes, obesity, and metabolic syndrome. These metabolic disorders are characterized by systemic and local inflammation within adipose tissue, the primary site of PCB accumulation. These inflammatory changes arise when resident adipose tissue macrophages undergo phenotypic plasticity-switching from an antiinflammatory to an inflammatory phenotype. Thus, we sought to assess whether PCB exposure drives macrophage phenotypic switching. We investigated how human monocyte-derived macrophages polarized toward an M1, M2a, or M2c phenotype were impacted by exposure to Aroclor 1254, a PCB mixture found at high levels in school air. We showed that PCB exposure not only exacerbates the inflammatory phenotype of M1 macrophages but also shifts both M2a and M2c cells toward a more inflammatory phototype in both a dose- and time-dependent manner. Additionally, we show that PCB exposure leads to significant metabolic changes. M2 macrophages exposed to PCBs exhibit increased reliance on aerobic glycolysis and reduced capacity for fatty acid and amino acid oxidation-both indicators of an inflammatory macrophage phenotype. Collectively, these results demonstrate that PCBs promote immunometabolic macrophage plasticity toward a more M1-like phenotype, thereby suggesting that PCBs exacerbate metabolic diseases by altering the inflammatory environment in adipose tissue.
Collapse
Affiliation(s)
- Riley M Behan-Bush
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Michael V Schrodt
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Elizabeth Kilburg
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Jesse N Liszewski
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Laura M Bitterlich
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland W23 F2H6
- Department of Biology, Maynooth University, Maynooth, Ireland W23 F2H6
| | - Karen English
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland W23 F2H6
- Department of Biology, Maynooth University, Maynooth, Ireland W23 F2H6
| | - Aloysius J Klingelhutz
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
22
|
Almeida L, Bacon A, Ghorasaini M, van der Ham AJ, Toes REM, Giera M, Everts B. IgA2 ACPA Drives a Hyper-Inflammatory Phenotype in Macrophages via ATP Synthase and COX2. Eur J Immunol 2025; 55:e202451586. [PMID: 40170376 PMCID: PMC11962234 DOI: 10.1002/eji.202451586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 04/03/2025]
Abstract
IgA can form immune complexes (ICs) and activate myeloid cells via Fc alpha receptor-mediated signalling to secrete pro-inflammatory cytokines. It was previously described that of the two IgA subclasses (IgA1 and IgA2), IgA2 is more inflammatory than IgA1. However, the mechanisms underlying this differential pro-inflammatory potential remain poorly defined. Using anti-citrullinated protein IgA1 and IgA2 antibodies (ACPA) that are commonly found in rheumatoid arthritis (RA) patients and linked to chronic inflammation, we show here that, in macrophages, IgA2-ICs boost TLR-induced TNF and IL6 secretion, COX2 expression, and production of COX2-dependent lipid mediators to a higher level than IgA1-ICs. Metabolically, we found the amplification of TLR-induced cytokine production and COX2 induction by IgA2-ICs to be dependent on mitochondrial ATP synthesis, but not glycolysis. Finally, we found the potentiation of TLR-induced cytokine production by IgA-ICs to be COX2-dependent. Together this work points towards a key role for mitochondrial ATP synthesis in driving COX2 expression and subsequent IgA2-IC-dependent potentiation of TLR-induced cytokine production by macrophages. As such, our work provides new insights into the mechanisms underlying IgA2-induced inflammation in the context of RA. Thus, this may hold novel clues to be explored as therapeutic possibilities to target antibody-driven inflammation in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Luís Almeida
- Centre for Infectious DiseasesLeiden University Medical CentreLeidenThe Netherlands
| | - Alice Bacon
- Department of RheumatologyLeiden University Medical CentreLeidenThe Netherlands
| | - Mohan Ghorasaini
- Centre for Proteomics and MetabolomicsLeiden University Medical CentreThe Netherlands
| | - Alwin J. van der Ham
- Centre for Infectious DiseasesLeiden University Medical CentreLeidenThe Netherlands
| | - René E. M. Toes
- Department of RheumatologyLeiden University Medical CentreLeidenThe Netherlands
| | - Martin Giera
- Centre for Proteomics and MetabolomicsLeiden University Medical CentreThe Netherlands
| | - Bart Everts
- Centre for Infectious DiseasesLeiden University Medical CentreLeidenThe Netherlands
| |
Collapse
|
23
|
Kervella M, Bertile F, Bouillaud F, Criscuolo F. The cell origin of reactive oxygen species and its implication for evolutionary trade-offs. Open Biol 2025; 15:240312. [PMID: 40237040 PMCID: PMC12001088 DOI: 10.1098/rsob.240312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/08/2025] [Accepted: 02/09/2025] [Indexed: 04/17/2025] Open
Abstract
The allocation of resources in animals is shaped by adaptive trade-offs aimed at maximizing fitness. At the heart of these trade-offs, lies metabolism and the conversion of food resources into energy, a process mostly occurring in mitochondria. Yet, the conversion of nutrients to utilizable energy molecules (adenosine triphosphate) inevitably leads to the by-production of reactive oxygen species (ROS) that may cause damage to important biomolecules such as proteins or lipids. The 'ROS theory of ageing' has thus proposed that the relationship between lifespan and metabolic rate may be mediated by ROS production. However, the relationship is not as straightforward as it may seem: not only are mitochondrial ROS crucial for various cellular functions, but mitochondria are also actually equipped with antioxidant systems, and many extra-mitochondrial sources also produce ROS. In this review, we discuss how viewing the mitochondrion as a regulator of cellular oxidative homeostasis, not merely a ROS producer, may provide new insights into the role of oxidative stress in the reproduction-survival trade-off. We suggest several avenues to test how mitochondrial oxidative buffering capacity might complement current bioenergetic and evolutionary studies.
Collapse
|
24
|
Alonso S, Kaur H, Jia L, Nguyen MU, Laguerta A, Fong A, Skariah N, Argüello RJ, Verzi MP, Swamy M, Lau KS, Edelblum KL. Microbiota promote enhanced CD39 expression in γδ intraepithelial lymphocytes through the activation of TCR and IL-15 signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.22.644616. [PMID: 40196668 PMCID: PMC11974730 DOI: 10.1101/2025.03.22.644616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Intraepithelial lymphocytes expressing the γδ T cell receptor (γδ IEL) provide continuous surveillance of the intestinal epithelium. We report that mice harboring a microbiota-specific hyperproliferative γδ IEL (γδ HYP ) phenotype also upregulate the expression of the ectonucleotidase CD39, a marker of regulatory γδ T cells. Enhanced TCR and IL-15 signaling correlates with a progression from a naïve-like CD39 neg γδ IEL to a more mature, tissue-adapted CD39 hi IEL population. We found that TCRγδ activation drives CD122-mediated CD39 upregulation on γδ HYP IELs and increased mucosal IL-15 further amplifies CD39 expression in these cells. Further investigation revealed that CD39 induction requires sustained exposure to the γδ HYP -associated microbiota. Moreover, CD39 hi γδ IELs exhibit a reduced capacity to produce pro-inflammatory cytokine, which may explain the lack of histopathology in γδ HYP mice. Overall, our study identifies a previously unappreciated mechanism by which an altered microbiota amplifies CD39 expression on γδ HYP IELs, leading to the expansion of γδ IELs with regulatory potential.
Collapse
|
25
|
Dong Y, Shayegan B, Su Y, Neira SV, Tang D. A novel multigene panel (Sig27) robustly predicts poor prognosis of renal cell carcinoma via high-level associations with immunosuppressive features. BJC REPORTS 2025; 3:16. [PMID: 40097553 PMCID: PMC11914224 DOI: 10.1038/s44276-025-00128-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 01/17/2025] [Accepted: 02/08/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND We investigated a 27-gene panel (Sig27), derived from prostate cancer, for risk stratification of RCC (clear cell RCC/ccRCC, papillary RCC/pRCC, and chromophobe RCC/chRCC). METHODS Sig27 gene expressions were examined in 960 RCC and 201 kidney tissues. Sig27 was evaluated for predicting overall survival (OS), association with immune checkpoints (IC), regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSC), and tumor-associated macrophages (TAM) in RCC. RESULTS Sig27 robustly predicts OS of ccRCC, pRCC, and chRCC. Sig27 stratifies high-risk ccRCCs: median survival month (MSM) 19.3 and 80.4% of deaths and high-risk pRCCs (MSM 19.6 and 58.6% of death) compared to low-risk ccRCCs (2.9% of death) and pRCCs (2.7% of fatality). Sig27 contains several novel genes related to the RCC immunosuppressive features. FPR3, NOD2, MCTP1, LAMP3, TFEC, and FAM65B are highly correlated with MDSC, Treg, TAM and multiple (≥12) ICs in RCCs. FPR3 and NOD2 are pattern recognition receptors and initiate proinflammatory responses via sensing pathogen-associated molecular patterns and damage-associated molecular patterns; their upregulations may contribute to chronic inflammation in RCC. The Sig27 metagene is expressed in ccRCC-associated immune cells: exhausted CD8T cells, TAM, Treg, and others. CONCLUSIONS Sig27 is a novel and effective pan-RCC biomarker with high-level associations with RCC immunosuppressive features.
Collapse
Affiliation(s)
- Ying Dong
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada
- Department of Surgery, McMaster University, Hamilton, ON, L8S 4K1, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada
| | - Bobby Shayegan
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada
- Department of Surgery, McMaster University, Hamilton, ON, L8S 4K1, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada
| | - Yingying Su
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada
- Department of Surgery, McMaster University, Hamilton, ON, L8S 4K1, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada
| | - Sandra Vega Neira
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada
- Department of Surgery, McMaster University, Hamilton, ON, L8S 4K1, Canada
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada
| | - Damu Tang
- Urological Cancer Center for Research and Innovation (UCCRI), St Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada.
- Department of Surgery, McMaster University, Hamilton, ON, L8S 4K1, Canada.
- The Research Institute of St Joe's Hamilton, St Joseph's Hospital, Hamilton, ON, L8N 4A6, Canada.
| |
Collapse
|
26
|
Krisna SS, Deagle RC, Ismailova N, Esomojumi A, Roy-Dorval A, Roth F, Berberi G, del Rincon SV, Fritz JH. Immunometabolic analysis of primary murine group 2 innate lymphoid cells: a robust step-by-step approach. Front Immunol 2025; 16:1545790. [PMID: 40181967 PMCID: PMC11966487 DOI: 10.3389/fimmu.2025.1545790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 04/05/2025] Open
Abstract
Group 2 Innate Lymphoid Cells (ILC2s) have recently been shown to exert key regulatory functions in both innate and adaptive immune response networks that drive the establishment and progression of type 2 immunity. Although mainly tissue resident, ILC2s and their crosstalk within tissue microenvironments influence metabolism at both the local and systemic levels. In turn, the energetic demand and metabolic status within these systems shape the diverse phenotypes and effector functions of ILC2s. Deciphering these metabolic networks in ILC2s is therefore essential in understanding their various roles in health as well as their associated pathophysiologies. Here we detail a framework of experimental approaches to study key immunometabolic states of primary murine ILC2s and link them to unique phenotypes and their corresponding functionality. Utilizing flow cytometry, Single Cell ENergetic metabolism by profilIng Translation inHibition (SCENITH), and the Seahorse platform we provide a framework that allows in-depth analysis of cellular bioenergetic states to determine the immunometabolic wiring of ILC2s. Connecting immunometabolic states and networks to ILC2 phenotypes and effector functions with this method will allow future in-depth studies to assess the potential of novel pharmaceutics in altering ILC2 functionality in clinical settings.
Collapse
Affiliation(s)
- Sai Sakktee Krisna
- Department of Physiology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits (MRCCT), Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Rebecca C. Deagle
- McGill University Research Centre on Complex Traits (MRCCT), Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Nailya Ismailova
- McGill University Research Centre on Complex Traits (MRCCT), Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Ademola Esomojumi
- McGill University Research Centre on Complex Traits (MRCCT), Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Audrey Roy-Dorval
- McGill University Research Centre on Complex Traits (MRCCT), Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Frederik Roth
- McGill University Research Centre on Complex Traits (MRCCT), Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Gabriel Berberi
- McGill University Research Centre on Complex Traits (MRCCT), Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Sonia V. del Rincon
- Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- Department of Oncology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Jörg H. Fritz
- Department of Physiology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits (MRCCT), Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| |
Collapse
|
27
|
Wise AD, TenBarge EG, Mendonça JDC, Mennen EC, McDaniel SR, Reber CP, Holder BE, Bunch ML, Belevska E, Marshall MG, Vaccaro NM, Blakely CR, Wellawa DH, Ferris J, Sheldon JR, Bieber JD, Johnson JG, Burcham LR, Monteith AJ. Mitochondria sense bacterial lactate and drive release of neutrophil extracellular traps. Cell Host Microbe 2025; 33:341-357.e9. [PMID: 40020664 PMCID: PMC11955204 DOI: 10.1016/j.chom.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/06/2024] [Accepted: 02/05/2025] [Indexed: 03/03/2025]
Abstract
Neutrophils induce oxidative stress, creating a harsh phagosomal environment. However, Staphylococcus aureus can survive these conditions, requiring neutrophils to deploy mechanisms that sense bacterial persistence. We find that staphylococcal lactate is a metabolic danger signal that triggers neutrophil extracellular trap release (NETosis). Neutrophils coordinate mitochondria in proximity to S. aureus-containing phagosomes, allowing transfer of staphylococcal lactate to mitochondria where it is rapidly converted into pyruvate and causes mitochondrial reactive oxygen species, a precursor to NETosis. Similar results were observed in response to phylogenetically distinct bacteria, implicating lactate accumulation as a broad signal triggering NETosis. Furthermore, patients with systemic lupus erythematosus (SLE) are more susceptible to bacterial infections. We find that SLE neutrophils cannot sense bacterial lactate impairing their capacity to undergo NETosis upon S. aureus infection but initiate aberrant NETosis triggered by apoptotic debris. Thus, neutrophils adapt mitochondria as sensory organelles that detect bacterial metabolic activity and dictate downstream antibacterial processes.
Collapse
Affiliation(s)
- Ashley D Wise
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Eden G TenBarge
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | | | - Ellie C Mennen
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Sarah R McDaniel
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Callista P Reber
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Bailey E Holder
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Madison L Bunch
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Eva Belevska
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | | | - Nicole M Vaccaro
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | | | - Dinesh H Wellawa
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada; Vaccine and Infectious Disease Organization, Saskatoon, SK, Canada
| | - Jennifer Ferris
- Division of Rheumatology, University of Tennessee Medical Center, Knoxville, TN, USA
| | - Jessica R Sheldon
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada; Vaccine and Infectious Disease Organization, Saskatoon, SK, Canada
| | - Jeffry D Bieber
- Division of Rheumatology, University of Tennessee Medical Center, Knoxville, TN, USA
| | - Jeremiah G Johnson
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Lindsey R Burcham
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Andrew J Monteith
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA; Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
28
|
Thiruvalluvan M, Billet S, Liu Z, Lownik J, Waissengrin B, Kim H, Villamejor AL, Milshteyn L, Li X, Gayhart M, Araña M, Sankar K, Posadas EM, Lopategui J, You S, Reckamp KL, Bhowmick NA. CD105 blockade restores osimertinib sensitivity in drug-resistant EGFR-mutant non-small cell lung cancer. Drug Resist Updat 2025; 81:101237. [PMID: 40090182 DOI: 10.1016/j.drup.2025.101237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/27/2025] [Accepted: 03/08/2025] [Indexed: 03/18/2025]
Abstract
AIM To investigate the role of CD105 in mediating drug resistance to EGFR-targeted therapy in non-small cell lung cancer (NSCLC). METHODS Imaging mass cytometry was conducted on 66 NSCLC tumors, 44 of which had EGFR mutations. We correlated clinical variables, including overall survival, with CD105 (endoglin) expression, a co-receptor for bone morphogenetic protein (BMP) signaling. Two osimertinib-resistant EGFR-mutant cell lines were developed to study the effects of EGFR and CD105 disruption. Single cell RNA sequencing of the isogenic parental and osimertinib resistant lines was performed. Additionally, ATAC sequencing and Single Cell ENergetIc metabolism by profiling Translation inHibition analysis (SCENITH) was used to assess promoter chromatin status and glycolytic state. RESULTS We found a negative correlation between CD105 expression and overall survival in patients. Treatment with osimertinib or EGFR knockdown significantly elevated CD105 expression in EGFR-mutant cell lines. Single-cell RNA sequencing identified a subset of cells with heightened endothelial characteristics and altered pyrimidine metabolism, associated with osimertinib resistance. These cells exhibited a slow-cycling behavior, characterized by elevated chromatin condensation and reduced glycolysis. Combining osimertinib with carotuximab, a CD105 neutralizing antibody, significantly reduced the slow-cycling transcriptomic signature, increased chromatin accessibility, and restored glycolysis compared to osimertinib treatment alone. Mass spectrometry confirmed that carotuximab re-engaged EGFR signaling by coupling it with CD105. Consequently, carotuximab re-sensitized resistant tumors to osimertinib by increasing their mitotic index and ERK signaling in mouse models. CONCLUSION Carotuximab effectively reduced the slow-cycling cell population and restored osimertinib sensitivity, offering a promising strategy for managing refractory NSCLC.
Collapse
Affiliation(s)
- Manish Thiruvalluvan
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Cedars-Sinai Cancer Center, Los Angeles, CA, USA
| | - Sandrine Billet
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Cedars-Sinai Cancer Center, Los Angeles, CA, USA
| | - Zhenqiu Liu
- Department of Statistics, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Joseph Lownik
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Barliz Waissengrin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Cedars-Sinai Cancer Center, Los Angeles, CA, USA
| | - Hyoyoung Kim
- Cedars-Sinai Cancer Center, Los Angeles, CA, USA; Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anton L Villamejor
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Xiamo Li
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Matthew Gayhart
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Manuel Araña
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kamya Sankar
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Cedars-Sinai Cancer Center, Los Angeles, CA, USA
| | - Edwin M Posadas
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Cedars-Sinai Cancer Center, Los Angeles, CA, USA
| | - Jean Lopategui
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sungyong You
- Cedars-Sinai Cancer Center, Los Angeles, CA, USA; Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Karen L Reckamp
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Cedars-Sinai Cancer Center, Los Angeles, CA, USA
| | - Neil A Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Cedars-Sinai Cancer Center, Los Angeles, CA, USA.
| |
Collapse
|
29
|
Jo DH, Kaczmarek S, Khan AUH, Pervin J, Clark DM, Gadde S, Wang L, McComb S, Visram A, Lee SH. Entinostat, a histone deacetylase inhibitor, enhances CAR-NK cell anti-tumor activity by sustaining CAR expression. Front Immunol 2025; 16:1533044. [PMID: 40124378 PMCID: PMC11925867 DOI: 10.3389/fimmu.2025.1533044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
Allogeneic natural killer (NK) cell therapy has demonstrated significant potential in cancer immunotherapy by harnessing NK cells to target malignancies. CD138-targeting chimeric antigen receptor (CAR)-engineered NK cells offer a promising therapeutic option for multiple myeloma (MM). However, sustaining CAR expression on CAR-NK cells during ex vivo expansion poses a challenge to developing effective immunotherapies. In this study, primary NK cells were isolated, cryopreserved, and modified to express anti-CD138 CARs through retroviral transduction. Histone deacetylase inhibitors (HDACi), particularly entinostat (ENT), were applied to enhance CAR expression stability in CAR-NK cells. Our findings indicate that ENT treatment significantly improves and maintains CAR expression, thereby enhancing the cytotoxic activity of CAR-NK cells against CD138-positive multiple myeloma cells. ENT-treated CAR-NK cells exhibited prolonged persistence and more significant tumor reduction in an MM tumor-bearing mouse model, highlighting the therapeutic potential of HDACi-treated CAR-NK cells. This study provides the first evidence that HDAC inhibitors can sustain CAR expression in CAR-NK cells in a promoter-dependent manner, potentially enhancing anti-tumor efficacy in multiple myeloma and underscoring the possible need for further clinical evaluation.
Collapse
Affiliation(s)
- Dong-Hyeon Jo
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Shelby Kaczmarek
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Abrar Ul Haq Khan
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jannat Pervin
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Diana M. Clark
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Suresh Gadde
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Scott McComb
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON, Canada
| | - Alissa Visram
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
30
|
Ehlers G, Tödtmann AM, Holsten L, Willers M, Heckmann J, Schöning J, Richter M, Heinemann AS, Pirr S, Heinz A, Dopfer C, Händler K, Becker M, Büchel J, Wöckel A, von Kaisenberg C, Hansen G, Hiller K, Schultze JL, Härtel C, Kastenmüller W, Vaeth M, Ulas T, Viemann D. Oxidative phosphorylation is a key feature of neonatal monocyte immunometabolism promoting myeloid differentiation after birth. Nat Commun 2025; 16:2239. [PMID: 40050264 PMCID: PMC11885822 DOI: 10.1038/s41467-025-57357-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
Neonates primarily rely on innate immune defense, yet their inflammatory responses are usually restricted compared to adults. This is controversially interpreted as a sign of immaturity or essential programming, increasing or decreasing the risk of sepsis, respectively. Here, combined transcriptomic, metabolic, and immunological studies in monocytes of healthy individuals reveal an inverse ontogenetic shift in metabolic pathway activities with increasing age. Neonatal monocytes are characterized by enhanced oxidative phosphorylation supporting ongoing myeloid differentiation. This phenotype is gradually replaced during early childhood by increasing glycolytic activity fueling the inflammatory responsiveness. Microbial stimulation shifts neonatal monocytes to an adult-like metabolism, whereas ketogenic diet in adults mimicking neonatal ketosis cannot revive a neonate-like metabolism. Our findings disclose hallmarks of innate immunometabolism during healthy postnatal immune adaptation and suggest that premature activation of glycolysis in neonates might increase their risk of sepsis by impairing myeloid differentiation and promoting hyperinflammation.
Collapse
Affiliation(s)
- Greta Ehlers
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Annika Marie Tödtmann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Lisa Holsten
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn, Bonn, Germany
| | - Maike Willers
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Julia Heckmann
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Jennifer Schöning
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Maximilian Richter
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Anna Sophie Heinemann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Sabine Pirr
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Alexander Heinz
- Department for Bioinformatics and Biochemistry, BRICS, Technical University Braunschweig, Braunschweig, Germany
| | - Christian Dopfer
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Kristian Händler
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute of Human Genetics, University of Lübeck, Lübeck, Germany
| | - Matthias Becker
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Modular High Performance Computing and Artificial Intelligence, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Johanna Büchel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | | | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Karsten Hiller
- Department for Bioinformatics and Biochemistry, BRICS, Technical University Braunschweig, Braunschweig, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn, Bonn, Germany
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Wolfgang Kastenmüller
- Würzburg Institute of Systems Immunology, Max Planck Research Group, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Martin Vaeth
- Würzburg Institute of Systems Immunology, Max Planck Research Group, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Thomas Ulas
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn, Bonn, Germany
| | - Dorothee Viemann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
- Center for Infection Research, University Würzburg, Würzburg, Germany.
| |
Collapse
|
31
|
Zhu W, Chen G, Xiao Z, Wang M, Li Z, Shi Y, Luo X, Li Z, Huang H, Chen X, Liang L, Liang D. Circadian Rhythm Disruption Exacerbates Autoimmune Uveitis: The Essential Role of PER1 in Treg Cell Metabolic Support for Stability and Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2400004. [PMID: 39823532 PMCID: PMC11904989 DOI: 10.1002/advs.202400004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 12/16/2024] [Indexed: 01/19/2025]
Abstract
Circadian rhythm plays a critical role in the progression of autoimmune diseases. While our previous study demonstrated the therapeutic effects of melatonin in experimental autoimmune uveitis, the involvement of circadian rhythm remained unclear. Using a light-induced circadian rhythm disruption model, we showed that disrupted circadian rhythms exacerbate autoimmune uveitis by impairing the stability and function of Treg cells. Mechanistically, we identified the core clock gene Per1, which is significantly reduced under circadian disruption, is essential for Treg cell metabolism and immunoregulatory function. This study underscores the pivotal role of circadian rhythm-related Treg cells in autoimmune disease progression.
Collapse
Affiliation(s)
- Wenjie Zhu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Guanyu Chen
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Zhiqiang Xiao
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Minzhen Wang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Zhuang Li
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Yuxun Shi
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Xiaohui Luo
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Zuoyi Li
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Haixiang Huang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Xiaoqing Chen
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Lingyi Liang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| | - Dan Liang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhou510060China
| |
Collapse
|
32
|
Zahoor I, Nematullah M, Ahmed ME, Fatma M, Sajad M, Ayasolla K, Cerghet M, Palaniyandi S, Ceci V, Carrera G, Buttari F, Centonze D, Mao-Draayer Y, Rattan R, Chiurchiù V, Giri S. Maresin-1 promotes neuroprotection and modulates metabolic and inflammatory responses in disease-associated cell types in preclinical models of multiple sclerosis. J Biol Chem 2025; 301:108226. [PMID: 39864620 PMCID: PMC11903811 DOI: 10.1016/j.jbc.2025.108226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/19/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025] Open
Abstract
Multiple sclerosis (MS) is a prevalent inflammatory neurodegenerative disease in young people, causing neurological abnormalities and impairment. To investigate a novel therapeutic agent for MS, we observed the impact of maresin 1 (MaR1) on disease progression in a well-known, relapsing-remitting experimental autoimmune encephalomyelitis mouse model. Treatment with MaR1 accelerated inflammation resolution, reduced neurological impairment, and delayed disease development by reducing immune cell infiltration (CD4+IL-17+ and CD4+IFNγ+) into the central nervous system. Furthermore, MaR1 administration enhanced IL-10 production, primarily in macrophages and CD4+ cells. However, neutralizing IL-10 with an anti-IL-10 antibody eliminated the protective impact by MaR1 in relapsing-remitting experimental autoimmune encephalomyelitis model, implying the significance of IL-10 in MaR1 treatment. Metabolism has been recognized as a critical mediator of effector activity in many types of immune cells. In our investigation, MaR1 administration significantly repaired metabolic dysregulation in CD4+ cells, macrophages, and microglia in EAE mice. Furthermore, MaR1 treatment restored defective efferocytosis in treated macrophages and microglia. MaR1 also preserved myelin in EAE mice and regulated O4+ oligodendrocyte metabolism by reversing metabolic dysregulation via increased mitochondrial activity and decreased glycolysis. Overall, in a preclinical MS animal model, MaR1 therapy has anti-inflammatory and neuroprotective properties. It also induced metabolic reprogramming in disease-associated cell types, increased efferocytosis, and maintained myelination. Moreover, our data on patient-derived peripheral blood mononuclear cells substantiated the protective role of MaR1, expanding the therapeutic spectrum of specialized proresolving lipid mediators. Altogether, these findings suggest the potential of MaR1 as a novel therapeutic agent for MS and other autoimmune diseases.
Collapse
MESH Headings
- Animals
- Docosahexaenoic Acids/pharmacology
- Docosahexaenoic Acids/therapeutic use
- Mice
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/pathology
- Disease Models, Animal
- Female
- Inflammation/drug therapy
- Inflammation/metabolism
- Inflammation/pathology
- Macrophages/metabolism
- Macrophages/drug effects
- Macrophages/pathology
- Macrophages/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Humans
- Mice, Inbred C57BL
- Interleukin-10/metabolism
- Neuroprotection/drug effects
- Neuroprotective Agents/pharmacology
Collapse
Affiliation(s)
- Insha Zahoor
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | | | | | - Mena Fatma
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | - Mir Sajad
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | | | - Mirela Cerghet
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA
| | - Suresh Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health, Detroit, Michigan, USA; Department of Physiology, Wayne State University, Detroit, Michigan, USA
| | - Veronica Ceci
- Institute of Translational Pharmacology, National Research Council, Rome, Italy; Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giulia Carrera
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Fabio Buttari
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), Italy
| | - Diego Centonze
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), Italy
| | - Yang Mao-Draayer
- Oklahoma Medical Research Foundation, Oklahoma, Farmington Hills, Michigan, USA
| | - Ramandeep Rattan
- Women's Health Services, Henry Ford Health, Detroit, Michigan, USA
| | - Valerio Chiurchiù
- Institute of Translational Pharmacology, National Research Council, Rome, Italy; Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health, Detroit, Michigan, USA.
| |
Collapse
|
33
|
Vaughn N. Cytometry at the Intersection of Metabolism and Epigenetics in Lymphocyte Dynamics. Cytometry A 2025; 107:165-176. [PMID: 40052492 DOI: 10.1002/cyto.a.24919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 04/11/2025]
Abstract
Landmark studies at the turn of the century revealed metabolic reprogramming as a driving force for lymphocyte differentiation and function. In addition to metabolic changes, differentiating lymphocytes must remodel their epigenetic landscape to properly rewire their gene expression. Recent discoveries have shown that metabolic shifts can shape the fate of lymphocytes by altering their epigenetic state, bringing together these two areas of inquiry. The ongoing evolution of high-dimensional cytometry has enabled increasingly comprehensive analyses of metabolic and epigenetic landscapes in lymphocytes that transcend the technical limitations of the past. Here, we review recent insights into the interplay between metabolism and epigenetics in lymphocytes and how its dysregulation can lead to immunological dysfunction and disease. We also discuss the latest technical advances in cytometry that have enabled these discoveries and that we anticipate will advance future work in this area.
Collapse
Affiliation(s)
- Nicole Vaughn
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
34
|
Viel S, Vivier E, Walzer T, Marçais A. Targeting metabolic dysfunction of CD8 T cells and natural killer cells in cancer. Nat Rev Drug Discov 2025; 24:190-208. [PMID: 39668206 DOI: 10.1038/s41573-024-01098-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 12/14/2024]
Abstract
The importance of metabolic pathways in regulating immune responses is now well established, and a mapping of the bioenergetic metabolism of different immune cell types is under way. CD8 T cells and natural killer (NK) cells contribute to cancer immunosurveillance through their cytotoxic functions and secretion of cytokines and chemokines, complementing each other in target recognition mechanisms. Several immunotherapies leverage these cell types by either stimulating their activity or redirecting their specificity against tumour cells. However, the anticancer activity of CD8 T cells and NK cells is rapidly diminished in the tumour microenvironment, closely linked to a decline in their metabolic capacities. Various strategies have been developed to restore cancer immunosurveillance, including targeting bioenergetic metabolism or genetic engineering. This Review provides an overview of metabolic dysfunction in CD8 T cells and NK cells within the tumour microenvironment, highlighting current therapies aiming to overcome these issues.
Collapse
Affiliation(s)
- Sébastien Viel
- Plateforme de Biothérapie et de Production de Médicaments de Thérapie Innovante, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
- APHM, Hôpital de la Timone, Marseille, France
- Paris Saclay Cancer Cluster, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Inserm, Prédicteurs moléculaires et nouvelles cibles en oncologie, Villejuif, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS UMR5308 ENS de Lyon, Lyon, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS UMR5308 ENS de Lyon, Lyon, France.
| |
Collapse
|
35
|
Ryabova A, Romanishkin I, Markova I, Pominova D. Simultaneous Application of Methylene Blue and Chlorin e6 Photosensitizers: Investigation on a Cell Culture. Sovrem Tekhnologii Med 2025; 17:58-68. [PMID: 40071078 PMCID: PMC11892575 DOI: 10.17691/stm2025.17.1.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Indexed: 03/14/2025] Open
Abstract
The application of photosensitizers for inhibition of oxidative phosphorylation in order to temporally decrease oxygen uptake by tumor cells in the course of photodynamic therapy (PDT) evokes growing interest. The aim of the study is to overcome tumor hypoxia for further photodynamic therapy with simultaneous use of type I photosensitizer methylene blue (MB) and type II photosensitizer chlorin e6. Material and Methods A photodynamic activity of MB and its combined use with chlorin e6 has been studied on the HeLa cell culture, their effect on cell metabolism in their co-accumulation and subsequent irradiation has also been assessed. Results MB generates reactive oxygen species in the cells in contrast to chlorin e6, which produces singlet oxygen. Besides, MB is converted to a colorless leucoform at low concentrations in the process of de-oxygenation. Incubation of cells with MB concurrently with chlorin e6 results in its greater fluorescence as compared to the incubation with MB only. MB concentration in the range of 1-10 mg/kg and the laser radiation dose of 60 J/cm2 do not cause cell death, probably, due to the MB transition to the photodynamically inactive leucoform. Cell death is observed after PDT in all samples with chlorin e6 and with MB at the 0-20 mg/kg concentration ranges and at 60 J/cm2 radiation dose. The phototoxicity of MB together with chlorin e6 is higher than that of chlorin e6 alone. The analysis of metabolic NADH cofactor lifetime after the incubation of the cells with MB and chlorin e6, and after PDT with them has revealed the presence of stress seen as an extension of NADH fluorescence cloud along the metabolic axis. After PDT with low concentrations of MB, the NADH fluorescent cloud on the phasor diagram shifts to the right towards short lifetimes (closer to anaerobic glycolysis along the NADH metabolic trajectory). The PDT with MB and chlorin e6 leads to the shift of the NADH fluorescence cloud on the phasor diagram to the left towards long lifetimes (closer to oxidative phosphorylation along the NADH metabolic trajectory). In this case, the cells die due to necrosis. Conclusion The co-accumulation of MB with chlorin e6 prevents MB reduction to a colorless leucoform, decreasing the oxygen uptake by the cells and making it possible to use simultaneously type I and II photodynamic reactions.
Collapse
Affiliation(s)
- A.V. Ryabova
- Senior Researcher, Laser Biospectroscopy Laboratory, Light-Induced Surface Phenomena Department, Natural Sciences Center; Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., Moscow, 119991, Russia; Associate Professor, Department 87 “Laser Micro-, Nano-, and Biotechnologies, Engineering Physics Institute for Biomedicine”; National Research Nuclear University MEPhI, 31 Kashirskoye Highway, Moscow, 115409, Russia
| | - I.D. Romanishkin
- Junior Researcher, Laser Biospectroscopy Laboratory, Light-Induced Surface Phenomena Department, Natural Sciences Center; Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., Moscow, 119991, Russia
| | - I.V. Markova
- PhD Student; National Research Nuclear University MEPhI, 31 Kashirskoye Highway, Moscow, 115409, Russia; Engineer, Laser Biospectroscopy Laboratory, Light-Induced Surface Phenomena Department, Natural Sciences Center; Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., Moscow, 119991, Russia
| | - D.V. Pominova
- PhD, Senior Researcher, Laser Biospectroscopy Laboratory, Light-Induced Surface Phenomena Department, Natural Sciences Center; Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., Moscow, 119991, Russia; Associate Professor, Department 87 “Laser Micro-, Nano-, and Biotechnologies, Engineering Physics Institute for Biomedicine”; National Research Nuclear University MEPhI, 31 Kashirskoye Highway, Moscow, 115409, Russia
| |
Collapse
|
36
|
Obare LM, Simmons J, Oakes J, Zhang X, Nochowicz C, Priest S, Bailin SS, Warren CM, Mashayekhi M, Beasley HK, Shao J, Meenderink LM, Sheng Q, Stolze J, Gangula R, Absi T, Ru Su Y, Neikirk K, Chopra A, Gabriel CL, Temu T, Pakala S, Wilfong EM, Gianella S, Phillips EJ, Harrison DG, Hinton A, Kalams SA, Kirabo A, Mallal SA, Koethe JR, Wanjalla CN. CD3+ T-cell: CD14+ monocyte complexes are dynamic and increased with HIV and glucose intolerance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae054. [PMID: 40073149 PMCID: PMC11952877 DOI: 10.1093/jimmun/vkae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/07/2024] [Indexed: 03/14/2025]
Abstract
Persistent systemic inflammation is associated with an elevated risk of cardiometabolic diseases. However, the characteristics of the innate and adaptive immune systems in individuals who develop these conditions remain poorly defined. Doublets, or cell-cell complexes, are routinely eliminated from flow cytometric and other immune phenotyping analyses, which limits our understanding of their relationship to disease states. Using well-characterized clinical cohorts, including participants with controlled human immunodeficiency virus (HIV) as a model for chronic inflammation and increased immune cell interactions, we show that circulating CD14+ monocytes complexed to CD3+ T cells are dynamic, biologically relevant, and increased in individuals with diabetes after adjusting for confounding factors. The complexes form functional immune synapses with increased expression of proinflammatory cytokines and greater glucose utilization. Furthermore, in persons with HIV, the CD3+ T cell: CD14+ monocyte complexes had more HIV copies compared to matched CD14+ monocytes or CD4+ T cells alone. Our results demonstrate that circulating CD3+ T-cell: CD14+ monocyte pairs represent dynamic cellular interactions that may contribute to inflammation and cardiometabolic disease pathogenesis. CD3+ T-cell: CD14+ monocyte complexes may originate or be maintained, in part, by chronic viral infections. These findings provide a foundation for future studies investigating mechanisms linking T cell-monocyte cell-cell complexes to developing immune-mediated diseases, including HIV and diabetes.
Collapse
Affiliation(s)
- Laventa M Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Joshua Simmons
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jared Oakes
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Xiuqi Zhang
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Cindy Nochowicz
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Stephen Priest
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Samuel S Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Christian M Warren
- Veterans Affairs Flow Cytometry Core, Veterans AffairsTennessee Valley Healthcare System, Nashville, TN, United States
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, United States
| | - Leslie M Meenderink
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, United States
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States
| | - Joey Stolze
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States
| | - Rama Gangula
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Yan Ru Su
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Curtis L Gabriel
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tecla Temu
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Suman Pakala
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Erin M Wilfong
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sara Gianella
- Division of Infectious Diseases, University of California, San Diego, CA, United States
| | - Elizabeth J Phillips
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - David G Harrison
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Spyros A Kalams
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Simon A Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, United States
| | - John R Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, United States
| | - Celestine N Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
37
|
Woods PS, Mutlu GM. Differences in glycolytic metabolism between tissue-resident alveolar macrophages and recruited lung macrophages. Front Immunol 2025; 16:1535796. [PMID: 40092977 PMCID: PMC11906440 DOI: 10.3389/fimmu.2025.1535796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Immunometabolism has emerged as a key area of focus in immunology and has the potential to lead to new treatments for immune-related diseases. It is well-established that glycolytic metabolism is essential for adaptation to hypoxia and for macrophage inflammatory function. Macrophages have been shown to upregulate their glycolytic metabolism in response to pathogens and pathogen-associated molecular patterns such as LPS. As a direct link to the external environment, the lungs' distinctive nutrient composition and multiple macrophage subtypes provide a unique opportunity to study macrophage metabolism. This review aims to highlight how the steady-state airway and severely inflamed airway offer divergent environments for macrophage glycolytic metabolism. We describe the differences in glycolytic metabolism between tissue-resident alveolar macrophages, and other lung macrophages at steady-state and during inflammation/injury. We also provide an overview of experimental guidelines on how to assess metabolism at the cellular level using Seahorse-based bioenergetic analysis including a review of pharmacologic agents used to inhibit or activate glycolysis.
Collapse
Affiliation(s)
| | - Gökhan M. Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University
of Chicago, Chicago, IL, United States
| |
Collapse
|
38
|
Nagatani A, Minamihata K, Adachi M, Wakabayashi R, Goto M, Kamiya N. Dual-functional co-crystal of streptavidin and ssDNA: electrostatic assembly with positively charged peptide tags. RSC Adv 2025; 15:6817-6822. [PMID: 40035016 PMCID: PMC11873783 DOI: 10.1039/d4ra08326a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
We have achieved a novel co-crystal in which the dual functions of the protein and single-stranded DNA are maintained by introducing a charged peptide tag at the C-terminus of the protein. The functionalities allowed the co-crystals to be modified with high selectivity. Additionally, we have confirmed that energy transfer occurs between the two molecules modified within the co-crystal. Therefore, this co-crystal has the potential as a novel biomaterial applicable to biosensors.
Collapse
Affiliation(s)
- Ayasa Nagatani
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University 744 Motooka Fukuoka 819-0395 Japan
| | - Kosuke Minamihata
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University 744 Motooka Fukuoka 819-0395 Japan
| | - Motoyasu Adachi
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology 4-9-1 Anagawa, Inage Chiba 263-8555 Japan
- Department of Quantum Life Science, Graduate School of Science, Chiba University Chiba 263-8522 Japan
| | - Rie Wakabayashi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University 744 Motooka Fukuoka 819-0395 Japan
| | - Masahiro Goto
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University 744 Motooka Fukuoka 819-0395 Japan
- Division of Biotechnology, Center for Future Chemistry, Kyushu University 744 Motooka Fukuoka 819-0395 Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University 744 Motooka Fukuoka 819-0395 Japan
- Division of Biotechnology, Center for Future Chemistry, Kyushu University 744 Motooka Fukuoka 819-0395 Japan
| |
Collapse
|
39
|
Brown AC, Carroll OR, Mayall JR, Zounemat-Kermani N, Vinzenz SLE, Gomez HM, Mills EF, Kim RY, Donovan C, Baines KJ, Williams EJ, Berthon BS, Wynne K, Scott HA, Pinkerton JW, Guo Y, Hansbro PM, Foster PS, Wark PAB, Dahlen SE, Adcock IM, Wood LG, Horvat JC. Female sex hormones and the oral contraceptive pill modulate asthma severity through GLUT-1. Mucosal Immunol 2025:S1933-0219(25)00024-8. [PMID: 40021011 DOI: 10.1016/j.mucimm.2025.02.006] [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: 09/27/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Females are disproportionately affected by asthma. An increased understanding of how female sex hormones influence key pathophysiological processes that underpin asthma may identify new, more effective asthma therapies, particularly for females with severe, poorly controlled asthma. We assessed the effects of oral ethinylestradiol/levonorgestrel (representing OCP use) and depot-medroxyprogesterone acetate (DMPA) and estradiol injections on key features of experimental asthma, and determined their effects on glucose transporter-1 (GLUT-1). The effects of OCP use on clinical asthma outcomes, and the relationships between estrogen receptors and type 2 (T2), non-T2, and GLUT-1 responses, in clinical asthma were also determined. OCP and DMPA reduce T2 responses, disease features, and lung expression of GLUT-1, whereas estradiol increases lung expression of GLUT-1, and results in severe, corticosteroid-insensitive, neutrophil-enriched disease, in experimental asthma. OCP use is associated with reduced T2 cytokine and GLUT-1 responses in clinical asthma. GLUT-1 expression is increased in sputum of severe asthmatics, and positively correlates with estrogen receptor expression and both T2 and non-T2 inflammatory responses. Significantly, OCP or GLUT-1 inhibition protects against obesity-associated or estradiol-induced, severe, experimental asthma, respectively. Together, these data show how female sex hormones and the OCP likely modulate asthma severity by modifying GLUT-1 responses in the airways.
Collapse
Affiliation(s)
- Alexandra C Brown
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Olivia R Carroll
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Jemma R Mayall
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | | | - Samantha L E Vinzenz
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Henry M Gomez
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Ed F Mills
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Richard Y Kim
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia; Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Chantal Donovan
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia; Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Katherine J Baines
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Evan J Williams
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Bronwyn S Berthon
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Katie Wynne
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Hayley A Scott
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - James W Pinkerton
- Respiratory Pharmacology & Toxicology Group, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Yike Guo
- Hong Kong University of Science and Technology, Hong Kong
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Paul S Foster
- Woolcock Institute of Medical Research and Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Peter A B Wark
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sven-Erik Dahlen
- Clinical Lung and Allergy Research Unit, Department of Medicine Huddinge, Karolinska Institutet, and, Department of Respiratory Medicine and Allergy, Karolinska University Hospital Huddinge, and Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ian M Adcock
- The Airway Disease Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Lisa G Wood
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- The University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.
| |
Collapse
|
40
|
Ferreira BH, Mazeda C, Dourado E, Simões JL, Prata AR, Argüello RJ, Duarte IF, Pierre P, Almeida CR. Distinct metabolic profiles of circulating plasmacytoid dendritic cells in systemic sclerosis patients stratified by clinical phenotypes. Arthritis Res Ther 2025; 27:35. [PMID: 39972361 PMCID: PMC11837673 DOI: 10.1186/s13075-025-03500-3] [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: 12/06/2024] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Plasmacytoid dendritic cells (pDCs) play a key role in systemic sclerosis (SSc) pathophysiology. However, despite the recognised importance of metabolic reprogramming for pDC function, their metabolic profile in SSc remains to be elucidated. Thus, our study aimed to explore the metabolic profile of pDCs in SSc and their potential contribution to the disease. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated from the blood of healthy donors and SSc patients. SCENITH™, a single-cell flow cytometry-based method, was applied to infer the metabolic profile of circulating pDCs from patients with SSc. pDCs (CD304+ Lin-) at steady-state or stimulated with CpG A were analysed. Toll-like receptor (TLR)9 activation was confirmed by ribosomal protein S6 phosphorylation. RESULTS Circulating pDCs from ten healthy donors and fourteen SSc patients were analysed. pDCs from anti-centromere antibody-positive (ACA+) patients displayed higher mitochondrial dependence and lower glycolytic capacity than those from anti-topoisomerase I antibody-positive (ATA+) patients. Furthermore, cells from both ACA+ patients and limited cutaneous SSc (lcSSc) patients showed a stronger response towards TLR9 activation than cells from ATA+, anti-RNA polymerase III antibody-positive (ARA+) or diffuse cutaneous SSc (dcSSc) patients. CONCLUSIONS An innovative single cell flow cytometry-based methodology was applied to analyse the metabolic profile of pDCs from SSc patients. Our results suggest that pDCs from ACA+ patients rely more on oxidative phosphorylation (OXPHOS) and are more responsive to external stimuli, whereas pDCs from ATA+ patients may exhibit a more activated or exhausted profile.
Collapse
Affiliation(s)
- Beatriz H Ferreira
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Carolina Mazeda
- Rheumatology Department, Unidade Local de Saúde da Região de Aveiro, Aveiro, Portugal
- Aveiro Rheumatology Research Centre, Egas Moniz Health Alliance, Aveiro, Portugal
- EpiDoc Unit, Nova Medical School, NOVA University Lisbon, Lisboa, Portugal
| | - Eduardo Dourado
- Rheumatology Department, Unidade Local de Saúde da Região de Aveiro, Aveiro, Portugal
- Aveiro Rheumatology Research Centre, Egas Moniz Health Alliance, Aveiro, Portugal
- Rheumatology Research Unit, Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - João L Simões
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
- School of Health Sciences (ESSUA), University of Aveiro, Aveiro, Portugal
| | - Ana Rita Prata
- Rheumatology Department, Unidade Local de Saúde da Região de Aveiro, Aveiro, Portugal
- Aveiro Rheumatology Research Centre, Egas Moniz Health Alliance, Aveiro, Portugal
| | - Rafael J Argüello
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, CNRS, INSERM, Marseille, France
- GammaOmics, Marseille, France
| | - Iola F Duarte
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
- Department of Chemistry, LAQV-REQUIMTE, University of Aveiro, Aveiro, Portugal
| | - Philippe Pierre
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, CNRS, INSERM, Marseille, France
- Shanghai Institute of Immunology, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Catarina R Almeida
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
41
|
Widhalm R, Granitzer S, Natha B, Zoboli O, Derx J, Zeisler H, Salzer H, Weiss S, Schmitner N, Kimmel RA, Österreicher T, Oberle R, Hengstschläger M, Distel M, Gundacker C. Perfluorodecanoic acid (PFDA) increases oxidative stress through inhibition of mitochondrial β-oxidation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125595. [PMID: 39734044 DOI: 10.1016/j.envpol.2024.125595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/04/2024] [Accepted: 12/25/2024] [Indexed: 12/31/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are a large group of synthetic organic chemicals that are ubiquitous environmental pollutants. Among PFAS, perfluorodecanoic acid (PFDA) is one of the most toxic compounds, but the molecular basis behind its toxicity is not fully understood. In an interspecies comparison with placental cells (HTR-8/SVneo) and zebrafish embryos, we demonstrate that PFDA induces mitochondrial dysfunction and impairs fatty acid β-oxidation. Reduced β-oxidation leads to less TCA cycle activity, resulting in less NADH and consequently NADPH production. Thereby NADPH-dependent glutathione recycling is impaired, increasing cellular oxidative stress that can only be partially compensated by NRF2 activation.
Collapse
Affiliation(s)
- Raimund Widhalm
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria; Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria.
| | - Sebastian Granitzer
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria; Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria
| | - Benjamin Natha
- Zebrafish Platform Austria for Preclinical Drug Screening (ZANDR), St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Ottavia Zoboli
- Institute for Water Quality and Resource Management, TU Wien, Vienna, Austria
| | - Julia Derx
- Institute of Hydraulic Engineering and Water Resources Management, TU Wien, Vienna, Austria; Interuniversity Cooperation Centre Water and Health, Vienna, Austria
| | - Harald Zeisler
- Department of Obstetrics and Gynecology, Medical University Vienna, Austria
| | - Hans Salzer
- Clinic for Pediatrics and Adolescent Medicine, University Clinic Tulln, Tulln, Austria
| | | | - Nicole Schmitner
- Institute of Molecular Biology, Center for Molecular Biosciences Innsbruck (CMBI), Leopold Franzens University Innsbruck, Innsbruck, Austria
| | - Robin A Kimmel
- Institute of Molecular Biology, Center for Molecular Biosciences Innsbruck (CMBI), Leopold Franzens University Innsbruck, Innsbruck, Austria
| | - Tamina Österreicher
- Center for Pathobiochemistry and Genetics, Institute of Medical Chemistry and Pathobiochemistry, Medical University of Vienna, Vienna, Austria
| | - Raimund Oberle
- Center for Pathobiochemistry and Genetics, Institute of Medical Chemistry and Pathobiochemistry, Medical University of Vienna, Vienna, Austria
| | - Markus Hengstschläger
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Martin Distel
- Zebrafish Platform Austria for Preclinical Drug Screening (ZANDR), St. Anna Children's Cancer Research Institute, Vienna, Austria; Innovative Cancer Models, St. Anna Children's Cancer Research Institute, Vienna, Austria; Division of Pediatric Hematology and Oncology, Intermountain Primary Children's Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, USA
| | - Claudia Gundacker
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria; Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria
| |
Collapse
|
42
|
Battram AM, Mañé-Pujol J, Moreno DF, Oliver-Caldés A, Carpio J, Cardus O, Rodríguez-Lobato LG, Urbano-Ispizua Á, Fernández de Larrea C. Genetic disruption of Blimp-1 drastically augments the antitumor efficacy of BCMA-targeting CAR T cells. Blood Adv 2025; 9:627-641. [PMID: 39642314 PMCID: PMC11847098 DOI: 10.1182/bloodadvances.2024013209] [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: 03/19/2024] [Revised: 10/15/2024] [Accepted: 11/11/2024] [Indexed: 12/08/2024] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T cells directed against B-cell maturation antigen (BCMA) are an effective treatment for multiple myeloma (MM), but short persistence and frequent relapses are challenges for this immunotherapy. This lack of durability has been attributed to the premature terminal differentiation of CAR T cells, which prevents the formation of long-lived memory cells that maintain antitumor responses. To improve long-term efficacy, we used CRISPR/CRISPR-associated protein 9-mediated gene editing to ablate the expression of the transcription factor Blimp-1. Blimp-1 knockout (KO) CAR T cells displayed a memory-like phenotype compared with control (Mock) CAR T cells, but had reduced effector function, with a striking loss of granzyme B. However, in a murine model of advanced MM, Blimp-1 KO CAR T cells effectively slowed or even prevented disease progression, significantly outperforming Mock CAR T cells in improving survival (P = .006). To understand this enhanced in vivo effectiveness, Blimp-1 KO CAR T cells were characterized after being repeatedly challenged with tumor cells in vitro. In this setting, Blimp-1 KO CAR T cells maintained a highly active state with high expression of memory markers, but, crucially, demonstrated enhanced effector function and increased energetic capacity. RNA-sequencing analysis of tumor-exposed Blimp-1 KO CAR T cells confirmed the presence of a memory-like transcriptomic signature and, additionally, revealed enhanced ribosome biogenesis and repressed CAR T-cell dysfunction as mechanisms that could contribute to improved antitumor activity. Put together, our findings show that dampening Blimp-1 expression altered the phenotype and function of anti-BCMA CAR T cells, leading to augmented therapeutic efficacy in MM.
Collapse
Affiliation(s)
- Anthony M. Battram
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Joan Mañé-Pujol
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
| | - David F. Moreno
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Aina Oliver-Caldés
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Judit Carpio
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Oriol Cardus
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
| | - Luis Gerardo Rodríguez-Lobato
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Álvaro Urbano-Ispizua
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Department of Hematology, University of Barcelona, Barcelona, Spain
| | - Carlos Fernández de Larrea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic of Barcelona, Barcelona, Spain
- Department of Hematology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
43
|
Woodworth KE, Froom ZSCS, Osborne ND, Rempe CN, Wheeler B, Medd K, Callaghan NI, Qian H, Acharya AP, Charron C, Huyer LD. Development of itaconate polymer microparticles for intracellular regulation of pro-inflammatory macrophage activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635692. [PMID: 39974988 PMCID: PMC11838496 DOI: 10.1101/2025.01.30.635692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Itaconate (IA) is an endogenous metabolite and a potent regulator of the innate immune system. Its use in immunomodulatory therapies has faced limitations due to inherent challenges in achieving controlled delivery and requirements for high extracellular concentrations to achieve internalization of the highly polar small molecule to achieve its intracellular therapeutic activity. Microparticle (MP)-based delivery strategies are a promising approach for intracellular delivery of small molecule metabolites through macrophage phagocytosis and subsequent intracellular polymer degradation-based delivery. Toward the goal of intracellular delivery of IA, degradable polyester polymer-(poly(itaconate-co-dodecanediol)) based IA polymer microparticles (IA-MPs) were generated using an emulsion method, forming micron-scale (∼ 1.5 µm) degradable microspheres. IA-MPs were characterized with respect to their material properties and IA release kinetics to inform particle fabrication. Treatment of murine bone marrow-derived macrophages with an optimized particle concentration of 0.1 mg/million cells enabled phagocytosis-mediated internalization and low levels of cytotoxicity. Flow cytometry demonstrated IA-MP-specific regulation of IA-sensitive inflammatory targets. Metabolic analyses demonstrated that IA-MP internalization inhibited oxidative metabolism and induced glycolytic reliance, consistent with the established mechanism of IA-associated inhibition of succinate dehydrogenase. This development of IA-based polymer microparticles provides a basis for additional innovative metabolite-based microparticle drug delivery systems for the treatment of inflammatory disease.
Collapse
|
44
|
Søndergaard JN, Tulyeu J, Priest D, Sakaguchi S, Wing JB. Single cell suppression profiling of human regulatory T cells. Nat Commun 2025; 16:1325. [PMID: 39900891 PMCID: PMC11791207 DOI: 10.1038/s41467-024-55746-1] [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: 05/15/2024] [Accepted: 12/23/2024] [Indexed: 02/05/2025] Open
Abstract
Regulatory T cells (Treg) play an important role in regulating immune homeostasis in health and disease. Traditionally their suppressive function has been assayed by mixing purified cell populations, which does not provide an accurate picture of a physiologically relevant response. To overcome this limitation, we here develop 'single cell suppression profiling of human Tregs' (scSPOT). scSPOT uses a 52-marker CyTOF panel, a cell division detection algorithm, and a whole PBMC system to assess the effect of Tregs on all other cell types simultaneously. In this head-to-head comparison, we find Tregs having the clearest suppressive effects on effector memory CD8 T cells through partial division arrest, cell cycle inhibition, and effector molecule downregulation. Additionally, scSPOT identifies a Treg phenotypic split previously observed in viral infection and propose modes of action by the FDA-approved drugs Ipilimumab and Tazemetostat. scSPOT is thus scalable, robust, widely applicable, and may be used to better understand Treg immunobiology and screen for therapeutic compounds.
Collapse
Affiliation(s)
- Jonas Nørskov Søndergaard
- Human Immunology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan.
| | - Janyerkye Tulyeu
- Human Immunology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - David Priest
- Laboratory of Human Single Cell Immunology, WPI-IFReC, Osaka University, Suita, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, WPI-IFReC, Osaka University, Suita, Japan
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - James B Wing
- Human Immunology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan.
- Laboratory of Human Single Cell Immunology, WPI-IFReC, Osaka University, Suita, Japan.
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka, Japan.
| |
Collapse
|
45
|
Zhang J, Hao L, Li S, He Y, Zhang Y, Li N, Hu X. mTOR/HIF-1α pathway-mediated glucose reprogramming and macrophage polarization by Sini decoction plus ginseng soup in ALF. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156374. [PMID: 39798342 DOI: 10.1016/j.phymed.2025.156374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/22/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Acute liver failure (ALF) has a high mortality rate, and despite treatment advancements, long-term outcomes remain poor. PURPOSE This study explores the therapeutic targets and pathways of Sini Decoction plus Ginseng Soup (SNRS) in ALF using bioinformatics and network pharmacology, focusing on its impact on macrophage polarization through glucose metabolism reprogramming. The efficacy of SNRS was validated in an LPS/D-GalN-induced ALF model, and its optimal concentration was determined for in vitro macrophage intervention. STUDY DESIGN AND METHODS Differentially expressed genes (DEGs) in HBV-induced and acetaminophen-induced ALF were identified from GEO datasets. The correlation between target gene expression and immune cell infiltration in ALF liver tissue was analyzed. AST, ALT, TNF-α, HMGB1, IL-1β, IL-6, and IL-10 levels were measured, and liver histopathology was assessed. Macrophage polarization was analyzed via immunofluorescence, flow cytometry, and Western blot. Glycolysis-related enzymes and metabolites, including HK2, PFK-1, PKM2, and LDHA, were quantified. Cellular ultrastructure was examined by transmission electron microscopy. RESULTS Five key glycolysis-regulating genes (HK2, CDK1, SOD1, VEGFA, GOT1) were identified, with significant involvement in the HIF-1 signaling pathway. Immune infiltration was markedly higher in ALF liver tissue. SNRS improved survival, reduced ALT/AST levels, alleviated liver injury, and modulated macrophage polarization by decreasing CD86 and increasing CD163 expression. In vitro, SNRS inhibited LPS-induced inflammatory cytokine release, lactate production, p-mTOR/mTOR ratio, and HIF-1α expression. CONCLUSION SNRS modulates macrophage polarization and glucose metabolism reprogramming via the mTOR/HIF-1α pathway, showing promise as a treatment for ALF.
Collapse
Affiliation(s)
- Junli Zhang
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu 610075, Sichuan Province, PR China; Department of Infectious Diseases, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Qinghuai District, Nanjing, Jiangsu 210029, PR China
| | - Liyuan Hao
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu 610075, Sichuan Province, PR China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China
| | - Shenghao Li
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu 610075, Sichuan Province, PR China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China
| | - Ying He
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu 610075, Sichuan Province, PR China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China
| | - Yang Zhang
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China
| | - Na Li
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu 610075, Sichuan Province, PR China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, PR China.
| |
Collapse
|
46
|
Sola‐Leyva A, Pathare ADS, Apostolov A, Aleksejeva E, Kask K, Tammiste T, Ruiz‐Durán S, Risal S, Acharya G, Salumets A. The hidden impact of GLP-1 receptor agonists on endometrial receptivity and implantation. Acta Obstet Gynecol Scand 2025; 104:258-266. [PMID: 39696822 PMCID: PMC11782050 DOI: 10.1111/aogs.15010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/27/2024] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
Increasing infertility rates represent a growing medical challenge in modern societies resulting from a complex interplay of sociocultural trends, lifestyle factors, exposure to environmental toxins, and underlying health problems. Women's fertility is particularly vulnerable to these shifts. The obesogenic lifestyle not only accelerates weight gain, but also disrupts ovulation driving the rise in infertility. Among several medications used for treating obesity and type 2 diabetes, glucagon-like peptide-1 receptor agonists (GLP-1RAs) show promising improvement in female fertility most likely by stimulating ovulation. However, the effects of GLP-1RAs on the endometrium remain unclear. Further studies are needed to investigate the impact of GLP-1RAs on endometrial receptivity and embryo implantation and early development. The aim of this study is to address the knowledge gap regarding the effects of GLP-1RAs on human reproduction, with special focus on the endometrium. Understanding these mechanisms may help to develop new strategies for improving fertility treatment, reduce implantation failure and address potential safety concerns regarding teratogenicity and adverse developmental outcomes for children born to women conceiving during or soon after GLP-1RA treatment.
Collapse
Affiliation(s)
- Alberto Sola‐Leyva
- Department of Gynecology and Reproductive MedicineKarolinska University HospitalStockholmSweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and TechnologyKarolinska InstitutetStockholmSweden
- Celvia CC, Competence Centre on Health TechnologiesTartuEstonia
| | | | - Apostol Apostolov
- Department of Gynecology and Reproductive MedicineKarolinska University HospitalStockholmSweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and TechnologyKarolinska InstitutetStockholmSweden
- Celvia CC, Competence Centre on Health TechnologiesTartuEstonia
- Department of Biotechnology, Institute of Molecular and Cell BiologyUniversity of TartuTartuEstonia
| | | | - Keiu Kask
- Celvia CC, Competence Centre on Health TechnologiesTartuEstonia
- Department of Obstetrics and Gynecology, Institute of Clinical MedicineUniversity of TartuTartuEstonia
| | - Triin Tammiste
- Department of Obstetrics and Gynecology, Institute of Clinical MedicineUniversity of TartuTartuEstonia
- West Tallinn Central HospitalWomen's ClinicTallinnEstonia
| | - Susana Ruiz‐Durán
- Department of Obstetrics and GynecologyVirgen de las Nieves University HospitalGranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | - Sanjiv Risal
- Department of Gynecology and Reproductive MedicineKarolinska University HospitalStockholmSweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and TechnologyKarolinska InstitutetStockholmSweden
| | - Ganesh Acharya
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and TechnologyKarolinska InstitutetStockholmSweden
- Center for Fetal Medicine KarolinskaUniversity HospitalStockholmSweden
- Women's Health and Perinatology Research Group, Department of Clinical MedicineUiT‐The Arctic University of NorwayTromsøNorway
| | - Andres Salumets
- Department of Gynecology and Reproductive MedicineKarolinska University HospitalStockholmSweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and TechnologyKarolinska InstitutetStockholmSweden
- Celvia CC, Competence Centre on Health TechnologiesTartuEstonia
- Department of Obstetrics and Gynecology, Institute of Clinical MedicineUniversity of TartuTartuEstonia
| |
Collapse
|
47
|
Dumolard L, Gerster T, Chuffart F, Decaens T, Hilleret MN, Larrat S, Saas P, Jouvin-Marche E, Durantel D, Marche PN, Macek Jilkova Z, Aspord C. HBV and HBsAg strongly reshape the phenotype, function, and metabolism of DCs according to patients' clinical stage. Hepatol Commun 2025; 9:e0625. [PMID: 39878655 PMCID: PMC11781764 DOI: 10.1097/hc9.0000000000000625] [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: 03/25/2024] [Accepted: 09/25/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Hepatitis B is a liver infection caused by HBV. Infected individuals who fail to control the viral infection develop chronic hepatitis B and are at risk of developing life-threatening liver diseases, such as cirrhosis or liver cancer. Dendritic cells (DCs) play important roles in the immune response against HBV but are functionally impaired in patients with chronic hepatitis B. The underlying mechanisms involved in HBV-induced DC dysfunctions remain to be elucidated. METHODS We explored DC modulations by HBV and HBsAg by exposing blood-derived cDC1s, cDC2s, and plasmacytoid DCs from healthy donors to HBV or HBsAg and stimulating them with toll-like receptor ligand. Their phenotypic and functional features, as well as their metabolic profile, were analyzed through multiparametric flow cytometry and multiplex assays and further explored on patients' samples. RESULTS We found that HBV deeply reshaped the DC secretome in response to toll-like receptor ligand. Strikingly, we observed that HBV-exposed DCs secrete high levels of CX3CL1 (fractalkine), a chemokine responsible for attracting antiviral effectors to the site of infection. HBsAg exposure favored DC activation while drastically altering TRAIL expression in response to toll-like receptor ligand and increasing the secretion of cytokines/chemokines involved in immune tolerance. HBsAg further dampened the metabolism of DC subsets while driving metabolic switches. Notably, the relevance of the CX3CL1/CX3CR1 axis, TGF-β, and metabolic disturbances was demonstrated within intrahepatic DC subsets in patients according to disease stage. CONCLUSIONS Our work brings new insights into the immunomodulation induced by HBV on DCs, which contribute to impaired antiviral responses and progression toward chronicity.
Collapse
Affiliation(s)
- Lucile Dumolard
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Theophile Gerster
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
- Etablissement Français du Sang Auvergne-Rhone-Alpes, R&D Laboratory, Grenoble, France
| | - Florent Chuffart
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Thomas Decaens
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
| | - Marie-Noelle Hilleret
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
| | - Sylvie Larrat
- University Grenoble Alpes, Laboratoire de Virologie, CHU Grenoble Alpes, Grenoble, France
| | - Philippe Saas
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- Etablissement Français du Sang Auvergne-Rhone-Alpes, R&D Laboratory, Grenoble, France
| | - Evelyne Jouvin-Marche
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
| | - David Durantel
- INSERM, U1111, Centre International de Recherche en Infectiologie (CIRI), University of Lyon (UCBL1), CNRS UMR_5308, ENS de Lyon, Lyon, France
| | - Patrice N. Marche
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Zuzana Macek Jilkova
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
| | - Caroline Aspord
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- Etablissement Français du Sang Auvergne-Rhone-Alpes, R&D Laboratory, Grenoble, France
| |
Collapse
|
48
|
Abrams ED, Basu A, Zavorka Thomas ME, Henrickson SE, Abraham RS. Expanding the diagnostic toolbox for complex genetic immune disorders. J Allergy Clin Immunol 2025; 155:255-274. [PMID: 39581295 DOI: 10.1016/j.jaci.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/29/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024]
Abstract
Laboratory-based immunology evaluation is essential to the diagnostic workup of patients with complex immune disorders, and is as essential, if not more so, depending on the context, as genetic testing, because it enables identification of aberrant pathways amenable to therapeutic intervention and clarifies variants of uncertain significance. There have been considerable advances in techniques and instrumentation in the clinical laboratory in the past 2 decades, although there are still "miles to go." One of the goals of the clinical laboratory is to ensure advanced diagnostic testing is widely accessible to physicians and thus patients, through reference laboratories, particularly in the context of academic medical centers. This ensures a greater likelihood of translating research discoveries into the diagnostic laboratory, on the basis of patient care needs rather than a sole emphasis on commercial utility. However, these advances are under threat from burdensome regulatory oversight that can compromise, at best, and curtail, at worst, the ability to rapidly diagnose rare immune disorders and ensure delivery of precision medicine. This review discusses the clinical utility of diagnostic immunology tools, beyond cellular immunophenotyping of lymphocyte subsets, which can be used in conjunction with clinical and other laboratory data for diagnosis as well as monitoring of therapeutic response in patients with genetic immunologic diseases.
Collapse
Affiliation(s)
- Eric D Abrams
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Amrita Basu
- Diagnostic Immunology Laboratory, Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Megan E Zavorka Thomas
- Diagnostic Immunology Laboratory, Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Sarah E Henrickson
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa; Institute for Immunology and Immune Health, University of Pennsylvania, Philadelphia, Pa; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Roshini S Abraham
- Diagnostic Immunology Laboratory, Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio.
| |
Collapse
|
49
|
Stier MT, Sewell AE, Mwizerwa EL, Sim CY, Tanner SM, Nichols CM, Durai HH, Jennings EQ, Lindau P, Wilfong EM, Newcomb DC, Bastarache JA, Ware LB, Rathmell JC. Metabolic Adaptations Rewire CD4 T Cells in a Subset-Specific Manner in Human Critical Illness with and without Sepsis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635146. [PMID: 39975258 PMCID: PMC11838299 DOI: 10.1101/2025.01.27.635146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Host immunity in sepsis has features of hyperinflammation together with progressive immunosuppression, particularly among CD4 T cells, that can predispose to secondary infections and ineffectual organ recovery. Metabolic and immunologic dysfunction are archetypal findings in critically ill patients with sepsis, but whether these factors are mechanistically linked remains incompletely defined. We characterized functional metabolic properties of human CD4 T cells from critically ill patients with and without sepsis and healthy adults. CD4 T cells in critical illness showed increased subset-specific metabolic plasticity, with regulatory T cells (Tregs) acquiring glycolytic capacity that stabilized suppressive markers FOXP3 and TIGIT and correlated with clinical illness severity. Single-cell transcriptomics identified differential kynurenine metabolism in Tregs, which was validated ex vivo as a mechanism of Treg glycolytic adaptation and suppressive rewiring. These findings underscore immunometabolic dysfunction as a driver of CD4 T cell remodeling in sepsis and suggest therapeutic avenues to restore an effective immune response.
Collapse
Affiliation(s)
- Matthew T. Stier
- Division of Allergy, Pulmonary & Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Allison E. Sewell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Erin L. Mwizerwa
- Division of Allergy, Pulmonary & Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Chooi Ying Sim
- Division of Allergy, Pulmonary & Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Samantha M. Tanner
- Division of Allergy, Pulmonary & Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Casey M. Nichols
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Heather H. Durai
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Erin Q. Jennings
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Paul Lindau
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Erin M. Wilfong
- Division of Allergy, Pulmonary & Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Dawn C. Newcomb
- Division of Allergy, Pulmonary & Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Julie A. Bastarache
- Division of Allergy, Pulmonary & Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary & Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeffrey C. Rathmell
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
50
|
Venkateswaran SV, Kreuzaler P, Maclachlan C, McMahon G, Greenidge G, Collinson L, Bunch J, Yuneva M. A multimodal imaging pipeline to decipher cell-specific metabolic functions and tissue microenvironment dynamics. Nat Protoc 2025:10.1038/s41596-024-01118-4. [PMID: 39880930 PMCID: PMC7617660 DOI: 10.1038/s41596-024-01118-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/15/2024] [Indexed: 01/31/2025]
Abstract
Tissue microenvironments are extremely complex and heterogeneous. It is challenging to study metabolic interaction between the different cell types in a tissue with the techniques that are currently available. Here we describe a multimodal imaging pipeline that allows cell type identification and nanoscale tracing of stable isotope-labeled compounds. This pipeline extends upon the principles of correlative light, electron and ion microscopy, by combining confocal microscopy reporter or probe-based fluorescence, electron microscopy, stable isotope labeling and nanoscale secondary ion mass spectrometry. We apply this method to murine models of hepatocellular and mammary gland carcinomas to study uptake of glucose derived carbon (13C) and glutamine derived nitrogen (15N) by tumor-associated immune cells. In vivo labeling with fluorescent-tagged antibodies (B220, CD3, CD8a, CD68) in tandem with confocal microscopy allows for the identification of specific cell types (B cells, T cells and macrophages) in the tumor microenvironment. Subsequent image correlation with electron microscopy offers the contrast and resolution to image membranes and organelles. Nanoscale secondary ion mass spectrometry tracks the enrichment of stable isotopes within these intracellular compartments. The whole protocol described here would take ~6 weeks to perform from start to finish. Our pipeline caters to a broad spectrum of applications as it can easily be adapted to trace the uptake and utilization of any stable isotope-labeled nutrient, drug or a probe by defined cellular populations in any tissue in situ.
Collapse
Affiliation(s)
| | - Peter Kreuzaler
- The Francis Crick Institute, London, UK.
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany.
| | | | - Greg McMahon
- The National Physical Laboratory, Teddington, UK
| | | | | | | | | |
Collapse
|