1
|
Gu J, Luo Y, Liang M, Fan Y, Zhang X, Ji G, Jin X. A novel framework for industrial pesticide effluent assessment: Integrating chemical screening, multi-endpoint responses and literature-based validation. JOURNAL OF HAZARDOUS MATERIALS 2025; 490:137830. [PMID: 40058200 DOI: 10.1016/j.jhazmat.2025.137830] [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: 12/01/2024] [Revised: 02/12/2025] [Accepted: 03/01/2025] [Indexed: 04/16/2025]
Abstract
Industrial pesticide effluents pose substantial risks to aquatic ecosystems, yet comprehensive understanding of their toxicological impacts remains limited. This study presents an integrated approach to evaluate the ecological risks of pesticide manufacturing effluents through chemical screening and multi-endpoints biological responses. Using zebrafish embryos as a model organism, we demonstrated that effluent discharge point (EDP) sample induced 100 % mortality, while diluted samples exhibited significant developmental toxicity, cardiovascular injury, immunosuppression, and behavioral alterations. Non-targeted metabolomics analysis revealed the molecular mechanisms underlying these toxic responses. Through chemical screening and targeted quantification, we identified three predominant azole fungicides - propiconazole (2.11 μg/L), hexaconazole (13.3 μg/L), and tebuconazole (18.66 μg/L) - that exhibited synergistic toxicity. Notably, our innovative meta-analysis framework based on literature data validated the toxicological profiles of detected compounds, providing an efficient alternative to conventional bioassays. This study establishes a comprehensive framework for assessing industrial effluent toxicity and demonstrates the value of integrating chemical analysis with biological responses for environmental risk assessment.
Collapse
Affiliation(s)
- Jie Gu
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Yiwen Luo
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Mengyuan Liang
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Yue Fan
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, China
| | - Xinyu Zhang
- School of Environmental Science and Engineering, Changzhou University, Changzhou 213164, China
| | - Guixiang Ji
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China.
| | - Xiaowei Jin
- China National Environmental Monitoring Centre, Beijing 100012, China.
| |
Collapse
|
2
|
Zhang Y, Tang M, Deng Q, Zhang Y, Zhao J, Zhu Y, Meng Y, Wang S, Liu Z, Guan Y, Li J, Du L. Estrogen-Driven Maintenance of GLUT1/GLUT4/SGLT1 under glucose starvation drives energy homeostasis in bovine PMNs. J Steroid Biochem Mol Biol 2025; 250:106716. [PMID: 40043818 DOI: 10.1016/j.jsbmb.2025.106716] [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: 11/25/2024] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/10/2025]
Abstract
The negative energy balance (NEB) and fluctuations in estrogen (17β-estradiol, E2) during the perinatal period alter glucose metabolism in bovine polymorphonuclear neutrophils (PMN) by affecting the activity of glucose transporters. In the peripheral blood, glucose uptake by PMNs is primarily dependent on the Glucose transporter type1 (GLUT1), Glucose transporter type4 (GLUT4), and Sodium-glucose cotransporter1 (SGLT1). However, the mechanisms through which E2 regulates energy metabolism in these cells, particularly through the modulation of glucose transporter activity, are currently unclear. This study aimed to explore the regulatory mechanisms underlying the effect of E2 on the homeostasis of glucose metabolism in PMNs. The results revealed that E2 enhances the expression of GLUT1, GLUT4, and SGLT1 (P < 0.05) and increases hexokinase (HK) activity (P < 0.05) in PMNs. Additionally, E2 was found to inhibit Glycogen synthase kinase-3β (GSK-3β) activity (P < 0.05), increase glycogen and ATP levels (P < 0.05), and reduce apoptosis in PMNs. When PMNs were treated with 5 μM STF-31 (GLUT1 inhibitor) or 50 μM Phlorizin (SGLT2 inhibitor), their GSK-3β activity was significantly increased (P < 0.05). Further analysis indicated that E2 helps maintain cellular glycogen and ATP homeostasis in PMNs by regulating the competitive interactions among GLUT1, GLUT4, and SGLT1. Additionally, when cells were treated with 100 μM AF-1890 (HK inhibitor), the expression of GLUT1, GLUT4, and SGLT1 was significantly reduced (P < 0.05). However, E2 mitigated the inhibitory effect of AF-1890 on HK activity and reduced its influence on intracellular energy levels by promoting the expression of GLUT1, GLUT4, and SGLT1. This study demonstrates that E2 positively regulates the expression of GLUT1, GLUT4 and SGLT1 in PMNs, facilitating glucose uptake under low-glucose conditions. E2 also negatively regulates GSK-3β activity increasing cellular glycogen and ATP levels and thus maintaining energy homeostasis in these cells.
Collapse
Affiliation(s)
- Yue Zhang
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China; Inner Mongolia Engineering Technology Research Center of Prevention and Control the Beef Cattle Disease, Tongliao, Inner Mongolia Autonomous Region 028000, China; Beef Cattle Industry School of Inner Mongolia Autonomous Region, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Mingyu Tang
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Qinghua Deng
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China; Inner Mongolia Engineering Technology Research Center of Prevention and Control the Beef Cattle Disease, Tongliao, Inner Mongolia Autonomous Region 028000, China; Beef Cattle Industry School of Inner Mongolia Autonomous Region, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Yuming Zhang
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China; Inner Mongolia Engineering Technology Research Center of Prevention and Control the Beef Cattle Disease, Tongliao, Inner Mongolia Autonomous Region 028000, China; Beef Cattle Industry School of Inner Mongolia Autonomous Region, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Junkang Zhao
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Yuli Zhu
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Yao Meng
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Shuang Wang
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Zhenhua Liu
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Yinxiang Guan
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Jinyu Li
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China
| | - Liyin Du
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, Inner Mongolia Autonomous Region 028000, China; Inner Mongolia Engineering Technology Research Center of Prevention and Control the Beef Cattle Disease, Tongliao, Inner Mongolia Autonomous Region 028000, China; Beef Cattle Industry School of Inner Mongolia Autonomous Region, Tongliao, Inner Mongolia Autonomous Region 028000, China.
| |
Collapse
|
3
|
Li X, Zhou W, Zhou L, Li Y, Wu X, Chen J. Neutrophil-derived exosomal S100A8 aggravates lung injury in sepsis by inducing pyroptosis. Mol Immunol 2025; 181:29-39. [PMID: 40056630 DOI: 10.1016/j.molimm.2025.03.003] [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: 11/26/2024] [Revised: 02/11/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025]
Abstract
Acute lung injury (ALI) is a common and life-threatening complication in patients with sepsis, with pro-inflammatory cell pyroptosis playing a crucial role in the associated organ damage. In this study, we aimed to identify potential therapeutic targets. Utilizing the GEO database (GSE232753), we analyzed the differentially expressed genes in the peripheral blood of healthy individuals and sepsis patients, identifying the significantly upregulated gene S100A8. Subsequently, we constructed a septic ALI model using lipopolysaccharide (LPS). Notably, S100A8 was highly expressed not only in serum and bronchoalveolar lavage fluid (BALF) but also in neutrophil exosomes. We then co-incubated BEAS-2B cells with neutrophil exosomes that were either treated or untreated with LPS. Cell proliferation activity was assessed using the CCK-8 assay, cell death was evaluated through propidium iodide (PI) staining, and the changes in pyroptosis indicators were detected via Western blot and ELISA. To further validate that LPS-induced neutrophil exosomes promote BEAS-2B cell pyroptosis through the delivery of S100A8, we conducted additional experiments involving the addition of S100A8 protein alone or S100A8 antibody in conjunction with neutrophil exosome treatment, followed by relevant assessments. Moreover, in vivo validation was also performed. Mechanistically, we revealed that S100A8 induces pyroptosis in BEAS-2B cells through the TLR4 signaling pathway. In conclusion, our findings provide new promising targets for the treatment of septic ALI.
Collapse
Affiliation(s)
- Xinxin Li
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China
| | - Wei Zhou
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China
| | - Liangliang Zhou
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China
| | - Yingbin Li
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China
| | - Xufeng Wu
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China
| | - Jianjun Chen
- Department of Emergency Intensive Care Medicine & Emergency Medicine, The First People's Hospital of Yancheng, Yancheng First Hospital Affiliated Nanjing University Medical College, Yancheng, Jiangsu 224000, China.
| |
Collapse
|
4
|
Lika J, Votava JA, Datta R, Mellado Fritz CA, Kralovec AM, Smith FM, Huttenlocher A, Skala MC, Fan J. Mitochondrial metabolism is rapidly re-activated in mature neutrophils to support stimulation-induced response. Front Immunol 2025; 16:1572927. [PMID: 40356902 PMCID: PMC12066771 DOI: 10.3389/fimmu.2025.1572927] [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: 02/07/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Neutrophils are highly abundant innate immune cells that are constantly produced from myeloid progenitors in the bone marrow. Differentiated neutrophils can perform an arsenal of effector functions critical for host defense. This study aims to quantitatively understand neutrophil mitochondrial metabolism throughout differentiation and activation, and to elucidate the impact of mitochondrial metabolism on neutrophil functions. Methods To study metabolic remodeling throughout neutrophil differentiation, murine ER-Hoxb8 myeloid progenitor-derived neutrophils and human induced pluripotent stem cell-derived neutrophils were assessed as models. To study the metabolic remodeling upon neutrophil activation, differentiated ER-Hoxb8 neutrophils and primary human neutrophils were activated with various stimuli, including ionomycin, monosodium urate crystals, and phorbol 12-myristate 13-acetate. Characterization of cellular metabolism by isotopic tracing, extracellular flux analysis, metabolomics, and fluorescence-lifetime imaging microscopy revealed dynamic changes in mitochondrial metabolism. Results As neutrophils mature, mitochondrial metabolism decreases drastically, energy production is offloaded from oxidative phosphorylation, and glucose oxidation through the TCA cycle is substantially reduced. Nonetheless, mature neutrophils retain the capacity for mitochondrial metabolism. Upon stimulation with certain stimuli, TCA cycle is rapidly activated. Mitochondrial pyruvate carrier inhibitors reduce this re-activation of the TCA cycle and inhibit the release of neutrophil extracellular traps. Treatment with these inhibitors also impacts neutrophil redox status, migration, and apoptosis without significantly changing overall bioenergetics. Conclusions Together, these results demonstrate that mitochondrial metabolism is dynamically remodeled and plays a significant role in neutrophils. Furthermore, these findings point to the therapeutic potential of mitochondrial pyruvate carrier inhibitors in a range of conditions where dysregulated neutrophil response drives inflammation and contributes to pathology.
Collapse
Affiliation(s)
- Jorgo Lika
- Morgridge Institute for Research, Madison, WI, United States
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, WI, United States
| | - James A. Votava
- Morgridge Institute for Research, Madison, WI, United States
| | - Rupsa Datta
- Morgridge Institute for Research, Madison, WI, United States
| | - Carlos A. Mellado Fritz
- Morgridge Institute for Research, Madison, WI, United States
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI, United States
| | - Aleksandr M. Kralovec
- Morgridge Institute for Research, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States
| | - Frances M. Smith
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, WI, United States
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, WI, United States
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI, United States
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, WI, United States
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI, United States
| |
Collapse
|
5
|
Bowers E, Entrup GP, Islam M, Mohan R, Lerner A, Mancuso P, Moore BB, Singer K. High fat diet feeding impairs neutrophil phagocytosis, bacterial killing, and neutrophil-induced hematopoietic regeneration. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:680-693. [PMID: 40094316 PMCID: PMC12041776 DOI: 10.1093/jimmun/vkaf024] [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: 06/03/2024] [Revised: 01/08/2025] [Accepted: 02/01/2025] [Indexed: 03/19/2025]
Abstract
The prevalence of obesity and metabolic diseases have risen significantly over the past decades. Chronic inflammation in obesity is a link between obesity and secondary disease. While macrophages and monocytes are known to contribute to metabolic disease risk during diet exposure, little is known about the contribution of neutrophils. We assessed the impact of obesity on neutrophils using a 16-week model of diet-induced obesity. Bone marrow (BM) neutrophils significantly expanded with chronic high-fat diet (HFD), significantly decreased TNFɑ protein release, and impaired neutrophil regenerative function compared to normal diet (ND) neutrophils. scRNAseq and flow cytometry demonstrated HFD neutrophil heterogeneity and validated that these cells do not have elevated expression of proinflammatory genes without secondary stimulation. HFD neutrophils showed elevated expression of genes associated with lipid metabolism-acyl-CoA thioesterase 1 (Acot1), carnitine palmitoyltransferase 1a (Cpt1a), and perilipin 2 (Plin2). Consistent with the importance of lipid metabolism in driving dysfunction, neutrophils from HFD-fed animals and neutrophils treated with palmitate had impaired bacterial phagocytosis and killing responses. These data shed light on the complex regulation of intracellular lipids and the role of metabolism on neutrophil function during homeostasis and disease.
Collapse
Affiliation(s)
- Emily Bowers
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Gabrielle P Entrup
- Immunology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Mohammed Islam
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Ramkumar Mohan
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Arianna Lerner
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Peter Mancuso
- Immunology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Bethany B Moore
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kanakadurga Singer
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
- Department Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
6
|
Bai HX, Gao YX, Wang S, Ma GY, Zhao W, Li XQ, Wang YF, Nong QN, Wang YB, Tan J, Duan Q, Cao W. Structure characteristics of a novel pectic polysaccharide from Fructus Corni and its protective effect on alcoholic fatty liver. Carbohydr Polym 2025; 352:123153. [PMID: 39843058 DOI: 10.1016/j.carbpol.2024.123153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/09/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025]
Abstract
Alcoholic fatty liver disease (AFLD) is characterized by the accumulation of hepatic lipid and has no effective treatment yet. Fructus Corni is a traditional Chinese medicinal herb, and its extractions have demonstrated hepatoprotective properties. We hypothesize that the polysaccharides in Fructus Corni might have therapeutic effects on AFLD. In this study, we isolated a novel homogeneous polysaccharide, APFC-2 (Mw= 63.0 kDa), from the Fructus Corni, and its structure was elucidated by monosaccharide composition, methylation analysis, partial acid hydrolysis, and NMR spectra. APFC-2 is a pectic polysaccharide characterized by a backbone of T-β-Galp-(1 → 6)-β-Galp-(1 → 3,6)-β-Galp-(1 → [4)-α-GalpA-OMe-(1 → 4)-α-GalpA-(1→]m → [2,4)-α-Rhap-(1 → 4)-α-GalpA-(1→]n, with branches comprising T-Araf-(1→, →3)-α-Araf-(1→, →3,5)-α-Araf-(1→, and →5)-α-Araf-(1→. In vivo experiments indicated that APFC-2 could significantly reduce hepatic steatosis, fasting triglyceride, and cholesterol levels in AFLD mice. Cell proliferation and Oil Red O staining results showed that APFC-2 concentration-dependently increased cell viability and significantly improved lipid metabolism in vitro. Mechanistically, APFC-2 markedly inhibited the formation of lipid both in vitro and in vivo through activating liver kinase B1 (LKB1) and then regulating adenosine 5'-monophosphate-activated protein kinase (AMPK)-SREBP-1 and AMPK-PPAR-α pathways. This research provides a theoretical basis for the potential application of Fructus Corni pectic polysaccharide as a specific activator of LKB1 for treating AFLD.
Collapse
Affiliation(s)
- Hong-Xin Bai
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Yu-Xuan Gao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Shuyao Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Guang-Yuan Ma
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Wenjing Zhao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Xiao-Qiang Li
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, Xi'an 710032, China
| | - Yu-Fan Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Qiu-Na Nong
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Yu-Bo Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Jin Tan
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Qimei Duan
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Wei Cao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, Xi'an 710032, China.
| |
Collapse
|
7
|
Wang Y, Lin F, Zhu G, Zhou X, Hu Y, Liu J. Diazonium-based derivatization for enhanced detection of phosphorylated metabolites by LC-MS in cells. J Pharm Biomed Anal 2025; 255:116642. [PMID: 39700864 DOI: 10.1016/j.jpba.2024.116642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/03/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
Phosphorylated small molecule metabolites play crucial roles in physiological processes such as glycogen metabolism and inflammation regulation. However, their high polarity, structural similarity, poor chromatographic separation, and weak mass spectrometric signals make their accurate quantification challenging, thereby hindering the study of related metabolic mechanisms and diseases. To address these challenges, we developed a novel derivatization reagent, DMQX (5-diazomethane quinoxaline), and combined it with liquid chromatography-mass spectrometry (LC-MS). This approach achieved baseline separation of five groups of isomers and enabled the quantification of 24 phosphorylated metabolites, providing comprehensive coverage of these metabolites in biological pathways. We applied this method to quantify 21 endogenous phosphorylated metabolites in HepG2 cells with and without vesicular stomatitis virus infection, demonstrating the potential of this analytical approach for advancing the study of metabolic mechanisms through quantitative analysis of phosphorylated metabolites in biological samples.
Collapse
Affiliation(s)
- Yikang Wang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310058, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Feifei Lin
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guozheng Zhu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310058, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaoxue Zhou
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310058, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Youhong Hu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310058, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jia Liu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310058, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
8
|
Hellenthal KEM, Thomas K, Ludwig N, Cappenberg A, Schemmelmann L, Tekath T, Margraf A, Mersmann S, Henke K, Rossaint J, Zarbock A, Amini W. Glutamine modulates neutrophil recruitment and effector functions during sterile inflammation. J Leukoc Biol 2025; 117:qiae243. [PMID: 39504570 DOI: 10.1093/jleuko/qiae243] [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: 07/18/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 11/08/2024] Open
Abstract
During sterile inflammation, tissue damage induces excessive activation and infiltration of neutrophils into tissues, where they critically contribute to organ dysfunction. Tight regulation of neutrophil migration and their effector functions is crucial to prevent overshooting immune responses. Neutrophils utilize more glutamine, the most abundant free α-amino acid in the human blood, than other leukocytes. However, under inflammatory conditions, the body's requirements exceed its ability to produce sufficient amounts of glutamine. This study investigates the impact of glutamine on neutrophil recruitment and their key effector functions. Glutamine treatment effectively reduced neutrophil activation by modulating β2-integrin activity and chemotaxis in vitro. In a murine in vivo model of sterile inflammation induced by renal ischemia-reperfusion injury, glutamine administration significantly attenuated neutrophil recruitment into injured kidneys. Transcriptomic analysis revealed, glutamine induces transcriptomic reprograming in murine neutrophils, thus improving mitochondrial functionality and glutathione metabolism. Further, glutamine influenced key neutrophil effector functions, leading to decreased production of reactive oxygen species and formation of neutrophil extracellular traps. Mechanistically, we used a transglutaminase 2 inhibitor to identify transglutaminase 2 as a downstream mediator of glutamine effects on neutrophils. In conclusion, our findings suggest that glutamine diminishes activation and recruitment of neutrophils and thus identify glutamine as a potent means to curb overshooting neutrophil responses during sterile inflammation.
Collapse
Affiliation(s)
- Katharina E M Hellenthal
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Katharina Thomas
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Nadine Ludwig
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
- Department of Cardiothoracic Surgery, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Anika Cappenberg
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Lena Schemmelmann
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Tobias Tekath
- Institute of Medical Informatics, University of Muenster, Albert-Schweitzer-Campus 1, Building A11, 48149 Muenster, Germany
| | - Andreas Margraf
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Sina Mersmann
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Katharina Henke
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Wida Amini
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| |
Collapse
|
9
|
Llibre A, Kucuk S, Gope A, Certo M, Mauro C. Lactate: A key regulator of the immune response. Immunity 2025; 58:535-554. [PMID: 40073846 DOI: 10.1016/j.immuni.2025.02.008] [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: 11/22/2024] [Revised: 01/22/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025]
Abstract
Lactate, the end product of both anaerobic and aerobic glycolysis in proliferating and growing cells-with the latter process known as the Warburg effect-is historically considered a mere waste product of cell and tissue metabolism. However, research over the past ten years has unveiled multifaceted functions of lactate that critically shape and impact cellular biology. Beyond serving as a fuel source, lactate is now known to influence gene expression through histone modification and to function as a signaling molecule that impacts a wide range of cellular activities. These properties have been particularly studied in the context of both adaptive and innate immune responses. Here, we review the diverse roles of lactate in the regulation of the immune system during homeostasis and disease pathogenesis (including cancer, infection, cardiovascular diseases, and autoimmunity). Furthermore, we describe recently proposed therapeutic interventions for manipulating lactate metabolism in human diseases.
Collapse
Affiliation(s)
- Alba Llibre
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Salih Kucuk
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Atrayee Gope
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Michelangelo Certo
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Claudio Mauro
- College of Medicine and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
10
|
Yang R, Zhang G, Meng Z, Wang L, Li Y, Li H, Yan S, Wei X, Wang S, Cui H. Glutamate dehydrogenase 1-catalytic glutaminolysis feedback activates EGFR/PI3K/AKT pathway and reprograms glioblastoma metabolism. Neuro Oncol 2025; 27:668-681. [PMID: 39446525 PMCID: PMC11889723 DOI: 10.1093/neuonc/noae222] [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: 03/13/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Glutamine is an important nutrient for cancer cell growth that provides biological sources for nucleic acid and fatty acid synthesis, but the role of glutaminolysis in signal transduction and glioblastoma (GBM) progression remains little known. METHODS Knockdown and overexpression cells were obtained to explore the functional roles of glutamate dehydrogenase 1 (GDH1) in cell proliferation, tumor formation, and aerobic glycolysis. RNA-seq, Chromatin immunoprecipitation, luciferase assay, and western blot were performed to verify the regulation of the EGFR-AKT pathway by the GDH1 (also known as GLUD1) and KDM6A. Metabolite-level measurements and Seahorse Assay were performed to assess the functional role of GHD1 in reprogramming glycolysis. RESULTS Here, we report that GDH1 catalytic glutaminolysis is essential for GBM cell line proliferation and brain tumorigenesis even in high-glucose conditions. Glutamine is metabolized through glutaminolysis to produce α-ketoglutarate (α-KG). We demonstrate that glutamine in combination with leucine activates mammalian TORC1 by enhancing glutaminolysis and α-KG production. α-KG increases the transcription of PDPK1 by reducing the suppressive histone modification H3K27me3 and then promotes the activation of the PI3K/AKT/mTOR pathway. This transcriptional activation induced by α-KG requires histone demethylase KDM6A, which is a 2-oxoglutarate oxygenase that plays an important role in converting α-KG to succinate. Furthermore, we show that GDH1-catalytic glutaminolysis also increases the expression of HK2 and promotes glycolysis in high-glucose conditions dependent on KDM6A-mediated demethylation of H3K27. CONCLUSIONS These findings suggest a novel function of glutaminolysis in the regulation of signal transduction and metabolism reprogramming and provide further evidence for the unique role of glutaminolysis in GBM progression.
Collapse
Affiliation(s)
- Rui Yang
- Biomedical Laboratory, School of Medicine, Liaocheng University, Liaocheng, China
| | - Guanghui Zhang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhen Meng
- Biomedical Laboratory, School of Medicine, Liaocheng University, Liaocheng, China
| | - Li Wang
- Biomedical Laboratory, School of Medicine, Liaocheng University, Liaocheng, China
| | - Yanping Li
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Haibin Li
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Siyuan Yan
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Xiaonan Wei
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Shanshan Wang
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Hongjuan Cui
- Jinfeng Laboratory, Chongqing, China
- Medical Research Institute, State Key Laboratory of Resources Insects, Southwest University, Chongqing, China
| |
Collapse
|
11
|
Mirchandani AS, Sanchez-Garcia MA, Walmsley SR. How oxygenation shapes immune responses: emerging roles for physioxia and pathological hypoxia. Nat Rev Immunol 2025; 25:161-177. [PMID: 39349943 DOI: 10.1038/s41577-024-01087-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 03/04/2025]
Abstract
Most eukaryotes require oxygen for their survival and, with increasing multicellular complexity, oxygen availability and delivery rates vary across the tissues of complex organisms. In humans, healthy tissues have markedly different oxygen gradients, ranging from the hypoxic environment of the bone marrow (where our haematopoietic stem cells reside) to the lungs and their alveoli, which are among the most oxygenated areas of the body. Immune cells are therefore required to adapt to varying oxygen availability as they move from the bone marrow to peripheral organs to mediate their effector functions. These changing oxygen gradients are exaggerated during inflammation, where oxygenation is often depleted owing to alterations in tissue perfusion and increased cellular activity. As such, it is important to consider the effects of oxygenation on shaping the immune response during tissue homeostasis and disease conditions. In this Review, we address the relevance of both physiological oxygenation (physioxia) and disease-associated hypoxia (where cellular oxygen demand outstrips supply) for immune cell functions, discussing the relevance of hypoxia for immune responses in the settings of tissue homeostasis, inflammation, infection, cancer and disease immunotherapy.
Collapse
Affiliation(s)
- Ananda Shanti Mirchandani
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | | | - Sarah Ruth Walmsley
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
12
|
Huang S, Shi J, Shen J, Fan X. Metabolic reprogramming of neutrophils in the tumor microenvironment: Emerging therapeutic targets. Cancer Lett 2025; 612:217466. [PMID: 39862916 DOI: 10.1016/j.canlet.2025.217466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Neutrophils are pivotal in the immune system and have been recognized as significant contributors to cancer development and progression. These cells undergo metabolic reprogramming in response to various stimulus, including infections, diseases, and the tumor microenvironment (TME). Under normal conditions, neutrophils primarily rely on aerobic glucose metabolism for energy production. However, within the TME featured by hypoxic and nutrient-deprived conditions, they shift to altered anaerobic glycolysis, lipid metabolism, mitochondrial metabolism and amino acid metabolism to perform their immunosuppressive functions and facilitate tumor progression. Targeting neutrophils within the TME is a promising therapeutic approach. Yet, focusing on their metabolic pathways presents a novel strategy to enhance cancer immunotherapy. This review synthesizes the current understanding of neutrophil metabolic reprogramming in the TME, with an emphasis on the underlying molecular mechanisms and signaling pathways. Studying neutrophil metabolism in the TME poses challenges, such as their short lifespan and the metabolic complexity of the environment, necessitating the development of advanced research methodologies. This review also discusses emerging solutions to these challenges. In conclusion, given their integral role in the TME, targeting the metabolic pathways of neutrophils could offer a promising avenue for cancer therapy.
Collapse
Affiliation(s)
- Shiyun Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| | - Jiahao Shi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| | - Jianfeng Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| |
Collapse
|
13
|
Lika J, Votava JA, Datta R, Kralovec AM, Smith FM, Huttenlocher A, Skala MC, Fan J. Mitochondrial metabolism is rapidly re-activated in mature neutrophils to support stimulation-induced response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636312. [PMID: 39975244 PMCID: PMC11838513 DOI: 10.1101/2025.02.03.636312] [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
Neutrophils are highly abundant innate immune cells that are constantly produced from myeloid progenitors in the bone marrow. Differentiated neutrophils can perform an arsenal of effector functions critical for host defense. This study aims to quantitatively understand neutrophil mitochondrial metabolism throughout differentiation and activation, and to elucidate the impact of mitochondrial metabolism on neutrophil functions. To study metabolic remodeling throughout neutrophil differentiation, murine ER-Hoxb8 myeloid progenitor-derived neutrophils and human induced pluripotent stem cell-derived neutrophils were assessed as models. To study the metabolic remodeling upon neutrophil activation, differentiated ER-Hoxb8 neutrophils and primary human neutrophils were activated with various stimuli, including ionomycin, MSU crystals, and PMA. Characterization of cellular metabolism by isotopic tracing, extracellular flux analysis, metabolomics, and fluorescence-lifetime imaging microscopy revealed dynamic changes in mitochondrial metabolism. As neutrophils mature, mitochondrial metabolism decreases drastically, energy production is fully offloaded from oxidative phosphorylation, and glucose oxidation through TCA cycle is substantially reduced. Nonetheless, mature neutrophils retain the capacity for mitochondrial metabolism. Upon stimulation with certain stimuli, TCA cycle is rapidly activated. Mitochondrial pyruvate carrier inhibitors reduce this re-activation of the TCA cycle and inhibit the release of neutrophil extracellular traps. Mitochondrial metabolism also impacts neutrophil redox status, migration, and apoptosis without significantly changing overall bioenergetics. Together, these results demonstrate that mitochondrial metabolism is dynamically remodeled and plays a significant role in neutrophil function and fate. Furthermore, these findings point to the therapeutic potential of mitochondrial pyruvate carrier inhibitors in a range of conditions where dysregulated neutrophil response drives inflammation and contributes to pathology.
Collapse
Affiliation(s)
- Jorgo Lika
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | - Rupsa Datta
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Aleksandr M. Kralovec
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Frances M. Smith
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Jing Fan
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| |
Collapse
|
14
|
Pan J, Lin Y, Liu X, Zhang X, Liang T, Bai X. Harnessing amino acid pathways to influence myeloid cell function in tumor immunity. Mol Med 2025; 31:44. [PMID: 39905317 PMCID: PMC11796060 DOI: 10.1186/s10020-025-01099-4] [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: 12/03/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
Amino acids are pivotal regulators of immune cell metabolism, signaling pathways, and gene expression. In myeloid cells, these processes underlie their functional plasticity, enabling shifts between pro-inflammatory, anti-inflammatory, pro-tumor, and anti-tumor activities. Within the tumor microenvironment, amino acid metabolism plays a crucial role in mediating the immunosuppressive functions of myeloid cells, contributing to tumor progression. This review delves into the mechanisms by which specific amino acids-glutamine, serine, arginine, and tryptophan-regulate myeloid cell function and polarization. Furthermore, we explore the therapeutic potential of targeting amino acid metabolism to enhance anti-tumor immunity, offering insights into novel strategies for cancer treatment.
Collapse
Affiliation(s)
- Jiongli Pan
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Lin
- Health Science Center, Ningbo University, Ningbo, China
| | - Xinyuan Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaozhen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Kim MJ, Akula HK, Marden J, Li K, Hu B, Vaska P, Qu W. The Potential Utility of (2S,4R)-4-[ 18F]fluoroglutamine as a Novel Metabolic Imaging Marker for Inflammation Explored by Rat Models of Arthritis and Paw Edema. Mol Imaging Biol 2025; 27:10-16. [PMID: 39572469 DOI: 10.1007/s11307-024-01967-1] [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: 05/31/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE (2S,4R)-4-[18F]fluoroglutamine ([18F]FGln) is a promising metabolic imaging marker in cancer. Based on the fact that major inflammatory cells are heavily dependent on glutamine metabolism like cancer cells, we explored the potential utility of [18F]FGln as a metabolic imaging marker for inflammation in two rat models: carrageenan-induced paw edema (CIPE) and collagen-induced arthritis (CIA). PROCEDURES The CIPE model (n = 4) was generated by injecting 200 µL of 3% carrageenan solution into the left hind paw three hours before the PET. The CIA model (n = 4) was generated by injecting 200 µg of collagen emulsion subcutaneously at the tail base 3-4 weeks before the PET. A qualitative scoring system was used to assess the severity of paw inflammation. After a CT scan, 15.7 ± 4.9 MBq of [18F]FGln was injected via the tail vein, followed by a dynamic micro-PET scan for 90 min under anesthesia with isoflurane. The standard uptake value of [18F]FGln was measured by placing a volume of interest in each paw. The non-injected right hind paws of the CIPE model rats served as controls for both models. The paws with CIA were pathologically examined after PET. RESULTS The CIPE models showed a trend toward higher uptake in the injected paw compared to the non-injected paw (P = 0.068). In CIA models, uptake in the paws with severe inflammation was significantly higher than the controls (P = 0.011), while that with mild and no inflammation was slightly higher (33%) and lower (-7%), respectively. Combined overall, the [18F]FGln uptake in CIA showed a significant positive correlation with inflammation severity (r = 0.88, P = 0.009). The pathological findings confirmed profound inflammation in CIA. CONCLUSIONS [18F]FGln uptake was increased in both acute and chronic inflammation, and the uptake level was significantly correlated with the severity, suggesting its potential utility as a novel metabolic imaging marker for inflammation.
Collapse
Affiliation(s)
- Min-Jeong Kim
- Department of Psychiatry and Behavioral Health, Stony Brook University School of Medicine, 101 Nicolls Rd, HSC T10-041L, Stony Brook, NY, 11794, USA.
- Department of Neurology, Stony Brook University School of Medicine, Stony Brook, NY, USA.
| | - Hari K Akula
- Department of Psychiatry and Behavioral Health, Stony Brook University School of Medicine, 101 Nicolls Rd, HSC T10-041L, Stony Brook, NY, 11794, USA
- PET Research Core, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Jocelyn Marden
- Department of Psychiatry and Behavioral Health, Stony Brook University School of Medicine, 101 Nicolls Rd, HSC T10-041L, Stony Brook, NY, 11794, USA
- PET Research Core, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Kaixuan Li
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
| | - Bao Hu
- Department of Psychiatry and Behavioral Health, Stony Brook University School of Medicine, 101 Nicolls Rd, HSC T10-041L, Stony Brook, NY, 11794, USA
- PET Research Core, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Paul Vaska
- Department of Radiology, Stony Brook University School of Medicine, Stony Brook, NY, USA
- Department of Biomedical Imaging, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Wenchao Qu
- Department of Psychiatry and Behavioral Health, Stony Brook University School of Medicine, 101 Nicolls Rd, HSC T10-041L, Stony Brook, NY, 11794, USA.
- PET Research Core, Stony Brook University School of Medicine, Stony Brook, NY, USA.
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
16
|
Fresneda Alarcon M, Abdullah GA, Nolan A, Linford C, Meschis MM, Cross AL, Sellin A, Phelan MM, Wright HL. The small molecule inhibitor 3PO is a modulator of neutrophil metabolism, ROS production, and NET release. Clin Exp Immunol 2025; 219:uxaf012. [PMID: 39969221 PMCID: PMC12060009 DOI: 10.1093/cei/uxaf012] [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: 10/15/2024] [Revised: 01/24/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025] Open
Abstract
Neutrophils are key effector leukocytes of the innate immune system and play a pivotal role in defending the host against microbial infections. Recent studies have identified a crucial link between glycolysis and neutrophil cellular functions. Using human neutrophils, we have investigated the intricate relationship between glycolysis, extracellular glucose availability, and the enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), in the regulation of reactive oxygen species (ROS) and neutrophil extracellular trap (NET) production. We have identified that PFKFB3 is elevated in rheumatoid arthritis (RA) neutrophils and that the small molecule PFKFB3 inhibitor 3PO is a key regulator of neutrophil ROS and NET production. 3PO blocked the production of ROS and NETs in a dose-dependent manner in both RA and healthy neutrophils (P < 0.01), and RA neutrophils were more sensitive to lower concentrations of 3PO. Bacterial killing was only partially inhibited by 3PO, and the proportion of live neutrophils after 24 h incubation was unchanged. Using NMR metabolomics, we identified that 3PO increases the concentration of lactate, phenylalanine, and L-glutamine in neutrophils, as well as significantly decreasing intracellular glutathione (adj. P-value < 0.05). We also demonstrated that RA neutrophils produce ROS and NETs in culture conditions which mimic the low glucose environments encountered in RA synovial joints. Our results also suggest that 3PO may have molecular targets beyond PFKFB3. By dissecting the intricate interplay between metabolism and neutrophil effector functions, this study advances the understanding of the molecular mechanisms governing pro-inflammatory neutrophil responses and identifies 3PO as a potential therapeutic for conditions characterized by dysregulated neutrophil activation.
Collapse
Affiliation(s)
| | - Genna Ali Abdullah
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Andy Nolan
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Christina Linford
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Maria Martina Meschis
- Institute of Systems, Molecular and Integrative Biology (ISMIB), Liverpool Head and Neck Centre, University of Liverpool, Liverpool, UK
| | - Andrew L Cross
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Andrew Sellin
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Marie M Phelan
- Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- High Field NMR Facility, Liverpool Shared Research Facilities, University of Liverpool, Liverpool, UK
| | - Helen L Wright
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
17
|
Grudzinska F, Faniyi AA, Belchamber KBR, Chen C, Stockley R, Jasper A, Parekh D, Sapey E, Scott A, Thickett DR. Hospitalised older adults with community-acquired pneumonia and sepsis have dysregulated neutrophil function but preserved glycolysis. Thorax 2025; 80:97-104. [PMID: 39689942 PMCID: PMC11877105 DOI: 10.1136/thorax-2024-222215] [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: 07/17/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024]
Abstract
OBJECTIVE Community-acquired pneumonia (CAP) is a leading cause of hospitalisation in older adults and is associated with a high likelihood of adverse outcomes. Given the ageing population and lack of therapeutic advances in CAP, new strategies to manage the burden of this disease are needed. Neutrophil dysfunction has been widely demonstrated in CAP and is associated with poor outcomes. We hypothesised that impaired glycolytic metabolism was driving neutrophil dysfunction in older adults with CAP. METHODS To investigate the mechanism underlying neutrophil dysfunction in CAP, we recruited older adults with CAP and sepsis, age-matched controls and healthy young adults to assess neutrophil function and glycolytic metabolism in peripheral blood neutrophils. RESULTS We demonstrate that neutrophils from older donors with CAP display a broad range of functional defects, including inaccurate migration to interleukin 8, impaired respiratory burst in response to phorbol 12-myristate 13-acetate and increased spontaneous degranulation compared with age-matched controls. Glycolysis (assessed by extracellular flux and RNA-sequencing) was not significantly altered between age-matched groups; however, basal rates of neutrophil glycolysis were significantly higher in patients with CAP and older adult controls compared with healthy young adults, and stimulated glycolysis was significantly higher in young adults compared with older adults with and without CAP. CONCLUSIONS Our findings suggest that neutrophil dysfunction in older adults with CAP may be implicated in poor outcomes, irrespective of glycolytic metabolism.
Collapse
Affiliation(s)
- Frances Grudzinska
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- School of Translational Medicine, University of Nottingham, Nottingham, UK
| | - Aduragbemi A Faniyi
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Kylie B R Belchamber
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, Birmingham, UK
- The University of Manchester Maternal and Fetal Health Research Centre, Manchester, UK
| | - Celine Chen
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Robert Stockley
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Alice Jasper
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Dhruv Parekh
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, Birmingham, UK
- NIHR Birmingham Clinical Research Facility, Birmingham, UK
| | - Elizabeth Sapey
- NIHR Birmingham Biomedical Research Centre, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- PIONEER HDR-UK Hub in Acute Care, Birmingham, UK
| | - Aaron Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, Birmingham, UK
| | - David R Thickett
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, Birmingham, UK
| |
Collapse
|
18
|
Li S, Zhu Q, Huang A, Lan Y, Wei X, He H, Meng X, Li W, Lin Y, Yang S. A machine learning model and identification of immune infiltration for chronic obstructive pulmonary disease based on disulfidptosis-related genes. BMC Med Genomics 2025; 18:7. [PMID: 39780155 PMCID: PMC11715737 DOI: 10.1186/s12920-024-02076-2] [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: 02/06/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a chronic and progressive lung disease. Disulfidptosis-related genes (DRGs) may be involved in the pathogenesis of COPD. From the perspective of predictive, preventive, and personalized medicine (PPPM), clarifying the role of disulfidptosis in the development of COPD could provide a opportunity for primary prediction, targeted prevention, and personalized treatment of the disease. METHODS We analyzed the expression profiles of DRGs and immune cell infiltration in COPD patients by using the GSE38974 dataset. According to the DRGs, molecular clusters and related immune cell infiltration levels were explored in individuals with COPD. Next, co-expression modules and cluster-specific differentially expressed genes were identified by the Weighted Gene Co-expression Network Analysis (WGCNA). Comparing the performance of the random forest (RF), support vector machine (SVM), generalized linear model (GLM), and eXtreme Gradient Boosting (XGB), we constructed the ptimal machine learning model. RESULTS DE-DRGs, differential immune cells and two clusters were identified. Notable difference in DRGs, immune cell populations, biological processes, and pathway behaviors were noted among the two clusters. Besides, significant differences in DRGs, immune cells, biological functions, and pathway activities were observed between the two clusters.A nomogram was created to aid in the practical application of clinical procedures. The SVM model achieved the best results in differentiating COPD patients across various clusters. Following that, we identified the top five genes as predictor genes via SVM model. These five genes related to the model were strongly linked to traits of the individuals with COPD. CONCLUSION Our study demonstrated the relationship between disulfidptosis and COPD and established an optimal machine-learning model to evaluate the subtypes and traits of COPD. DRGs serve as a target for future predictive diagnostics, targeted prevention, and individualized therapy in COPD, facilitating the transition from reactive medical services to PPPM in the management of the disease.
Collapse
Affiliation(s)
- Sijun Li
- Infectious Disease Laboratory, The Fourth People's Hospital of Nanning, Nanning, China
| | - Qingdong Zhu
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Aichun Huang
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Yanqun Lan
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Xiaoying Wei
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Huawei He
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Xiayan Meng
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Weiwen Li
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Yanrong Lin
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China.
| | - Shixiong Yang
- Administrative Office, The Fourth People's Hospital of Nanning, Nanning, China.
| |
Collapse
|
19
|
Mierzejewski K, Stryiński R, Bogacka I, Golubska M, Carrera M, Kurzynska A. Lipopolysaccharide affects metabolic processes and energy homeostasis in the corpus luteum. Front Mol Biosci 2025; 11:1523098. [PMID: 39845899 PMCID: PMC11753227 DOI: 10.3389/fmolb.2024.1523098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Chronic inflammation caused by Escherichia coli infections has a significant negative impact on the reproductive system and impairs fertility. The corpus luteum (CL) plays a central role not only in regulating the ovary cycle, but also in implantation of the embryo and maintenance of early pregnancy through the secretion of progesterone. Understanding the intricate interplay between inflammatory processes and reproductive organ's function is crucial for the development of effective therapeutic strategies to alleviate reproductive disorders and improve fertility. Methods The aim of this study was to determine the in vitro effects of lipopolysaccharide (LPS) on the proteomic profile of the porcine CL in the mid-luteal phase of the estrous cycle using LC-MS/MS analysis. The CL slices were incubated in the presence of LPS for 24 h. Results We identified 12 differentially regulated proteins after treatment with LPS (7 of them were upregulated, while 5 were downregulated). The analysis showed that these proteins are involved in processes such as glucose metabolism, the tricarboxylic acid cycle (TCA), detoxification processes as well as steroid biosynthesis in the CL. Moreover, we demonstrated that LPS decreases glucose levels and increases progesterone levels in the CL. Conclusion These findings suggest that LPS modulates key metabolic pathways in the CL, potentially impacting its functional activity.
Collapse
Affiliation(s)
- Karol Mierzejewski
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Robert Stryiński
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Iwona Bogacka
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Monika Golubska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Mónica Carrera
- Department of Food Technology, Institute of Marine Research (IIM), Spanish National Research Council (CSIC), Vigo, Spain
| | - Aleksandra Kurzynska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
20
|
Jobe T, Stephan J, Wells CK, De Silva M, Lorkiewicz PK, Hill BG, Wysoczynski M. Phase partitioning of the neutrophil oxidative burst is coordinated by accessory pathways of glucose metabolism and mitochondrial activity. J Biol Chem 2025; 301:108091. [PMID: 39675714 PMCID: PMC11760813 DOI: 10.1016/j.jbc.2024.108091] [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: 10/22/2024] [Revised: 11/26/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024] Open
Abstract
Neutrophils are a part of the innate immune system and produce reactive oxygen species (ROS) to extinguish pathogens. The major source of ROS in neutrophils is NADPH oxidase, which is fueled by NADPH generated via the pentose phosphate pathway; however, it is unclear how other accessory glucose metabolism pathways and mitochondrial activity influence the respiratory burst. We examined the temporal dynamics of the respiratory burst and delineated how metabolism changes over time after neutrophil activation. Bone marrow-derived neutrophils were stimulated with phorbol 12-myristate 13-acetate, and the respiratory burst was measured via extracellular flux analysis. Metabolomics experiments utilizing 13C6-glucose highlighted the activation of glycolysis as well as ancillary pathways of glucose metabolism in activated neutrophils. Phorbol 12-myristate 13-acetate stimulation acutely increased 13C enrichment into glycerol 3-phosphate (G3P) and citrate, whereas increases in 13C enrichment in the glycogen intermediate, UDP-hexose, and end products of the hexosamine and serine biosynthetic pathways occurred only during the late phase of the oxidative burst. Targeted inhibition of the G3P shuttle, glycogenolysis, serine biosynthesis, and mitochondrial respiration demonstrated that the G3P shuttle contributes to the general magnitude of ROS production; that glycogen contributes solely to the early respiratory burst; and that the serine biosynthetic pathway activity and complex III-driven mitochondrial activity influence respiratory burst duration. Collectively, these results show that the neutrophil oxidative burst is highly dynamic, with coordinated changes in metabolism that control the initiation, magnitude, and duration of ROS production.
Collapse
Affiliation(s)
- Tyler Jobe
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, Kentucky; Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Jonah Stephan
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, Kentucky; Department of Biochemistry, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Collin K Wells
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, Kentucky; Department of Biochemistry, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Maleesha De Silva
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, Kentucky
| | - Pawel K Lorkiewicz
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, Kentucky
| | - Bradford G Hill
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, Kentucky.
| | - Marcin Wysoczynski
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, Kentucky.
| |
Collapse
|
21
|
Xiang KF, Wan JJ, Wang PY, Liu X. Role of glycogen in cardiac metabolic stress. Metabolism 2025; 162:156059. [PMID: 39500406 DOI: 10.1016/j.metabol.2024.156059] [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: 08/04/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/11/2024]
Abstract
Metabolic stress in the myocardium arises from a diverse array of acute and chronic pathophysiological contexts. Glycogen mishandling is a key feature of metabolic stress, while maladaptation in energy-stress situations confers functional deficits. Cardiac glycogen serves as a pivotal reserve for myocardial energy, which is classically described as an energy source and contributes to glucose homeostasis during hypoxia or ischemia. Despite extensive research activity, how glycogen metabolism affects cardiovascular disease remains unclear. In this review, we focus on its regulation across myocardial energy metabolism in response to stress, and its role in metabolism, immunity, and autophagy. We further summarize the cardiovascular-related drugs regulating glycogen metabolism. In this way, we provide current knowledge for the understanding of glycogen metabolism in the myocardium.
Collapse
Affiliation(s)
- Ke-Fa Xiang
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China; Department of Cardiology, The 72nd Group Army Hospital, Huzhou University, Huzhou, Zhejiang 313000, China
| | - Jing-Jing Wan
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Peng-Yuan Wang
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China.
| |
Collapse
|
22
|
Yipeng Z, Chao C, Ranran L, Tingting P, Hongping Q. Metabolism: a potential regulator of neutrophil fate. Front Immunol 2024; 15:1500676. [PMID: 39697327 PMCID: PMC11652355 DOI: 10.3389/fimmu.2024.1500676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Neutrophils are essential components of the innate immune system that defend against the invading pathogens, such as bacteria, viruses, and fungi, as well as having regulatory roles in various conditions, including tissue repair, cancer immunity, and inflammation modulation. The function of neutrophils is strongly related to their mode of cell death, as different types of cell death involve various cellular and molecular alterations. Apoptosis, a non-inflammatory and programmed type of cell death, is the most common in neutrophils, while other modes of cell death, including NETOsis, necrosis, necroptosis, autophagy, pyroptosis, and ferroptosis, have specific roles in neutrophil function regulation. Immunometabolism refers to energy and substance metabolism in immune cells, and profoundly influences immune cell fate and immune system function. Intercellular and intracellular signal transduction modulate neutrophil metabolism, which can, in turn, alter their activities by influencing various cell signaling pathways. In this review, we compile an extensive body of evidence demonstrating the role of neutrophil metabolism in their various forms of cell death. The review highlights the intricate metabolic characteristics of neutrophils and their interplay with various types of cell death.
Collapse
Affiliation(s)
| | | | | | - Pan Tingting
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai, China
| | - Qu Hongping
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai, China
| |
Collapse
|
23
|
Jeroundi N, Roy C, Basset L, Pignon P, Preisser L, Blanchard S, Bocca C, Abadie C, Lalande J, Gueguen N, Mabilleau G, Lenaers G, Moreau A, Copin MC, Tcherkez G, Delneste Y, Couez D, Jeannin P. Glycogenesis and glyconeogenesis from glutamine, lactate and glycerol support human macrophage functions. EMBO Rep 2024; 25:5383-5407. [PMID: 39424955 PMCID: PMC11624281 DOI: 10.1038/s44319-024-00278-4] [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: 10/06/2023] [Revised: 09/03/2024] [Accepted: 09/15/2024] [Indexed: 10/21/2024] Open
Abstract
Macrophages fight infection and ensure tissue repair, often operating at nutrient-poor wound sites. We investigated the ability of human macrophages to metabolize glycogen. We observed that the cytokines GM-CSF and M-CSF plus IL-4 induced glycogenesis and the accumulation of glycogen by monocyte-derived macrophages. Glyconeogenesis occurs in cells cultured in the presence of the inflammatory cytokines GM-CSF and IFNγ (M1 cells), via phosphoenolpyruvate carboxykinase 2 (PCK2) and fructose-1,6-bisphosphatase 1 (FBP1). Enzyme inhibition with drugs or gene silencing techniques and 13C-tracing demonstrate that glutamine (metabolized by the TCA cycle), lactic acid, and glycerol were substrates of glyconeogenesis only in M1 cells. Tumor-associated macrophages (TAMs) also store glycogen and can perform glyconeogenesis. Finally, macrophage glycogenolysis and the pentose phosphate pathway (PPP) support cytokine secretion and phagocytosis regardless of the availability of extracellular glucose. Thus, glycogen metabolism supports the functions of human M1 and M2 cells, with inflammatory M1 cells displaying a possible dependence on glyconeogenesis.
Collapse
Affiliation(s)
- Najia Jeroundi
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, LabEx IGO, F-49000, Angers, France
| | - Charlotte Roy
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, LabEx IGO, F-49000, Angers, France
| | - Laetitia Basset
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, LabEx IGO, F-49000, Angers, France
| | - Pascale Pignon
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, LabEx IGO, F-49000, Angers, France
| | - Laurence Preisser
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, LabEx IGO, F-49000, Angers, France
| | - Simon Blanchard
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, LabEx IGO, F-49000, Angers, France
- Immunology and Allergology laboratory, University Hospital, Angers, France
| | - Cinzia Bocca
- Univ Angers, Inserm, CNRS, MitoVasc, SFR ICAT, F-49000, Angers, France
- Department of Genetics and Biochemistry, University Hospital, Angers, France
| | - Cyril Abadie
- Univ Angers, INRAe, IRHS, SFR QUASAV, F-49000, Angers, France
| | - Julie Lalande
- Univ Angers, INRAe, IRHS, SFR QUASAV, F-49000, Angers, France
| | - Naïg Gueguen
- Univ Angers, Inserm, CNRS, MitoVasc, SFR ICAT, F-49000, Angers, France
- Department of Genetics and Biochemistry, University Hospital, Angers, France
| | - Guillaume Mabilleau
- Univ Angers, Nantes Université, Inserm, Oniris, RMeS, SFR ICAT, F-49000, Angers, France
- Department of Cell and Tissue Pathology, University Hospital, Angers, France
| | - Guy Lenaers
- Univ Angers, Inserm, CNRS, MitoVasc, SFR ICAT, F-49000, Angers, France
- Department of Genetics and Biochemistry, University Hospital, Angers, France
| | - Aurélie Moreau
- Inserm, Nantes Université, University Hospital of Nantes, Centre de Recherche Translationnelle en Transplantation et Immunologie, Nantes, France
| | - Marie-Christine Copin
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, LabEx IGO, F-49000, Angers, France
- Department of Cell and Tissue Pathology, University Hospital, Angers, France
| | - Guillaume Tcherkez
- Univ Angers, INRAe, IRHS, SFR QUASAV, F-49000, Angers, France
- Research School of Biology, ANU College of Science, Australian National University, Canberra, ACT, 2601, Australia
| | - Yves Delneste
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, LabEx IGO, F-49000, Angers, France
- Immunology and Allergology laboratory, University Hospital, Angers, France
| | - Dominique Couez
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, LabEx IGO, F-49000, Angers, France
| | - Pascale Jeannin
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, LabEx IGO, F-49000, Angers, France.
- Immunology and Allergology laboratory, University Hospital, Angers, France.
| |
Collapse
|
24
|
Lika J, Fan J. Carbohydrate metabolism in supporting and regulating neutrophil effector functions. Curr Opin Immunol 2024; 91:102497. [PMID: 39366310 PMCID: PMC11609006 DOI: 10.1016/j.coi.2024.102497] [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: 07/01/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/06/2024]
Abstract
Neutrophils, the first responders of the innate immune system, can turn on a range of effector functions upon activation. Emerging research shows activated neutrophils undergo highly dynamic metabolic rewiring. This metabolic rewiring provides energy and reducing power to fuel effector functions and modulate signaling molecules to regulate neutrophil functions. Here, we review the current understanding of the specific metabolic requirements and regulators of neutrophil migration, neutrophil extracellular traps release, and pathogen killing. Particularly, we discuss how major carbohydrate metabolic pathways, including glycolysis, glycogen cycling, pentose phosphate pathway, and TCA cycle, are rewired upon neutrophil activation to support these functions. Continued investigation into the metabolic regulators of neutrophil functions can lead to therapeutic opportunities in various diseases.
Collapse
Affiliation(s)
- Jorgo Lika
- Morgridge Institute for Research, Madison, WI, USA; Medical Scientist Training Program, University of Wisconsin-Madison, Madison, WI, USA; Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI, USA; Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
25
|
Liu N, Zhu XR, Wu CY, Liu YY, Chen MB, Gu JH. PCK1 as a target for cancer therapy: from metabolic reprogramming to immune microenvironment remodeling. Cell Death Discov 2024; 10:478. [PMID: 39578429 PMCID: PMC11584723 DOI: 10.1038/s41420-024-02240-8] [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: 07/12/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
Recently, changes in metabolites and metabolism-related enzymes related to tumor cell proliferation, metastasis, drug resistance, and immunosuppression have become a research hotspot, and researchers have attempted to determine the clinical correlation between specific molecular lesions and metabolic phenotypes. Convincing evidence shows that metabolic reprogramming is closely related to the proliferation, invasion, metastasis, and poor prognosis of malignant tumors. Therefore, targeting metabolic reprogramming is a new direction for cancer treatment. However, how molecular alterations in tumors contribute to metabolic diversity and unique targeting dependencies remains unclear. A full understanding of the underlying mechanisms of metabolic reprogramming in cancer may lead to better identification of therapeutic targets and the development of therapeutic strategies. Evidence for the importance of PCK1, a phosphoenolpyruvate carboxykinase 1, in tumorigenesis and development is accumulating. PCK1 can regulate cell proliferation and metastasis by remodeling cell metabolism. Additionally, PCK1 has "nonclassical" nonmetabolic functions, involving the regulation of gene expression, angiogenesis, epigenetic modification, and other processes, and has an impact on cell survival, apoptosis, and other biological activities, as well as the remodeling of the tumor immune microenvironment. Herein, we provide a comprehensive overview of the functions of PCK1 under physiological and pathological conditions and suggest that PCK1 is a potential target for cancer therapy. We also propose a future exploration direction for targeting PCK1 for cancer therapy from a clinical perspective. Finally, in view of the collective data, the results of our discussion suggest the potential clinical application of targeted PCK1 therapy in combination with chemotherapy and immunotherapy for cancer treatment.
Collapse
Affiliation(s)
- Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Xiao-Ren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Chang-Ying Wu
- Department of Intensive Care Medicine, Chongqing People's Hospital, Chongqing, China
| | - Yuan-Yuan Liu
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Min-Bin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Jin-Hua Gu
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University Kunshan, Kunshan, China.
| |
Collapse
|
26
|
Datta R, Miskolci V, Gallego-López GM, Britt E, Gillette A, Kralovec A, Giese MA, Qian T, Votava J, Fan J, Huttenlocher A, Skala M. Single cell autofluorescence imaging reveals immediate metabolic shifts of neutrophils with activation across biological systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605362. [PMID: 39211087 PMCID: PMC11360992 DOI: 10.1101/2024.07.26.605362] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neutrophils, the most abundant leukocytes in human peripheral circulation, are crucial for the innate immune response. They are typically quiescent but rapidly activate in response to infection and inflammation, performing diverse functions such as oxidative burst, phagocytosis, and NETosis, which require significant metabolic adaptation. Deeper insights into such metabolic changes will help identify regulation of neutrophil functions in health and diseases. Due to their short lifespan and associated technical challenges, the metabolic processes of neutrophils are not completely understood. This study uses optical metabolic imaging (OMI), which entails optical redox ratio and fluorescence lifetime imaging microscopy of intrinsic metabolic coenzymes NAD(P)H and FAD to assess the metabolic state of single neutrophils. Primary human neutrophils were imaged in vitro under a variety of activation conditions and metabolic pathway inhibitors, while metabolic and functional changes were confirmed with mass spectrometry, oxidative burst, and NETosis measurements. Our findings show that neutrophils undergo rapid metabolic remodeling to a reduced redox state indicated by changes in NAD(P)H lifetime and optical redox ratio, with a shift to an oxidized redox state during activation. Additionally, single cell OMI analysis reveals a heterogeneous metabolic response across neutrophils and human donors to live pathogen infection ( Pseudomonas aeruginosa and Toxoplasma gondii ). Finally, consistent OMI changes with activation were confirmed between in vitro human and in vivo zebrafish larvae neutrophils. This study demonstrates the potential of OMI as a versatile tool for studying neutrophil metabolism and underscores its use across different biological systems, offering insights into neutrophil metabolic activity and function at a single cell level.
Collapse
|
27
|
Huo R, Sun Q, Lv Q, Wang Y, Qi W, Zhang M, Li L, Wang X. Simvastatin ameliorates adverse pregnancy by inhibiting glycolysis-related NETs in obstetrical antiphospholipid syndrome. Life Sci 2024; 359:123215. [PMID: 39505298 DOI: 10.1016/j.lfs.2024.123215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/08/2024]
Abstract
AIMS Some patients with Obstetric Antiphospholipid Syndrome (OAPS) still experience miscarriage and placental dysfunction after routine treatment, which is related to an abnormal increase in neutrophil extracellular traps (NETs). The labeling of statins has been revised to remove the contraindication for use during pregnancy. Our aim is to investigate the effect of Simvastatin on pregnancy outcomes in OAPS and its correlation mechanisms with NETs. MAIN METHODS The effect of Simvastatin on pregnancy outcomes was observed. The effect of simvastatin on the function and apoptosis of neutrophils has evaluated. The effect of Simvastatin to NETs and the changes in oxidative stress levels were observed. Different groups of NETs were extracted to intervene the HTR8-Svneo.RNA-seq analysis of the mechanism of which Simvastatin reduces NETs. Seahorse experiment detected the effect of Simvastatin on neutrophil glycolysis levels. Fluorescence co-localization and flow cytometry and Co-IP were used to verify relevant mechanisms. KEY FINDINGS In the OAPS mice, Simvastatin can reduce embryo absorption rate, reshape placental blood flow perfusion. Simultaneously reducing the production of NETs both in vivo and vitro, remolding oxidative stress. Simvastatin can improve neutrophil dysfunction caused by aPL-IgG. The reduction of NETs improved HTR8-Svneo's dysfunction. The intervention of Simvastatin on neutrophils under the stimulation of aPL-IgG showed a signature in glycolytic. The key rate limiting enzyme PKM2 in glycolysis interacts with Cit-H2b and PI3K/AKT signaling pathway. SIGNIFICANCE Our study providing basic theoretical support for the treatment of OAPS with Simvastatin.
Collapse
Affiliation(s)
- Ruiheng Huo
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China; Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China
| | - Qipeng Sun
- School of Clinical and Basic Medicine (Institute of Basic Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China
| | - Qingfeng Lv
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China; Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China
| | - Yuan Wang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Weiyi Qi
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, People's Republic of China
| | - Lei Li
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China; Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China.
| | - Xietong Wang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China; Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China; School of Clinical and Basic Medicine (Institute of Basic Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People's Republic of China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, People's Republic of China.
| |
Collapse
|
28
|
Kling L, Eulenberg-Gustavus C, Jerke U, Rousselle A, Eckardt KU, Schreiber A, Kettritz R. β 2-integrins control HIF1α activation in human neutrophils. Front Immunol 2024; 15:1406967. [PMID: 39469705 PMCID: PMC11513320 DOI: 10.3389/fimmu.2024.1406967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/12/2024] [Indexed: 10/30/2024] Open
Abstract
During inflammation, human neutrophils engage β2-integrins to migrate from the blood circulation to inflammatory sites with high cytokine but low oxygen concentrations. We tested the hypothesis that the inhibition of prolyl hydroxylase domain-containing enzymes (PHDs), cytokines, and β2-integrins cooperates in HIF pathway activation in neutrophils. Using either the PHD inhibitor roxadustat (ROX) (pseudohypoxia) or normobaric hypoxia to stabilize HIF, we observed HIF1α protein accumulation in adherent neutrophils. Several inflammatory mediators did not induce HIF1α protein but provided additive or even synergistic signals (e.g., GM-CSF) under pseudohypoxic and hypoxic conditions. Importantly, and in contrast to adherent neutrophils, HIF1α protein expression was not detected in strictly suspended neutrophils despite PHD enzyme inhibition and the presence of inflammatory mediators. Blocking β2-integrins in adherent and activating β2-integrins in suspension neutrophils established the indispensability of β2-integrins for increasing HIF1α protein. Using GM-CSF as an example, increased HIF1α mRNA transcription via JAK2-STAT3 was necessary but not sufficient for HIF1α protein upregulation. Importantly, we found that β2-integrins led to HIF1α mRNA translation through the phosphorylation of the essential translation initiation factors eIF4E and 4EBP1. Finally, pseudohypoxic and hypoxic conditions inducing HIF1α consistently delayed apoptosis in adherent neutrophils on fibronectin under low serum concentrations. Pharmacological HIF1α inhibition reversed delayed apoptosis, supporting the importance of this pathway for neutrophil survival under conditions mimicking extravascular sites. We describe a novel β2-integrin-controlled mechanism of HIF1α stabilization in human neutrophils. Conceivably, this mechanism restricts HIF1α activation in response to hypoxia and pharmacological PHD enzyme inhibitors to neutrophils migrating toward inflammatory sites.
Collapse
Affiliation(s)
- Lovis Kling
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Claudia Eulenberg-Gustavus
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Jerke
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Anthony Rousselle
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Adrian Schreiber
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ralph Kettritz
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
29
|
Qiu S, Jiang Q, Li Y. The association between pan-immune-inflammation value and chronic obstructive pulmonary disease: data from NHANES 1999-2018. Front Physiol 2024; 15:1440264. [PMID: 39434724 PMCID: PMC11491374 DOI: 10.3389/fphys.2024.1440264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Background The pan-immune-inflammation value (PIV) is an emerging biomarker quantitatively reflecting the systemic immune-inflammatory status. The predictive value of PIV has been well-established across various clinical settings. However, its role in chronic obstructive pulmonary disease (COPD) remains unclear and necessitates further investigation. Methods Data from NHANES 1999-2018 were filtered. Logistic regression analyses were used to assess the correlation between COPD prevalence and PIV in all participants. COX regression analyses and Kaplan-Meier survival curves were used to investigate the relationship between COPD all-cause mortality and PIV in COPD patients. Restricted cubic spline (RCS) analyses and piecewise linear regression analyses were additionally employed to explore the correlation between PIV and COPD. Subgroup analyses were performed to further clarify the effects of other covariates on the associations. Sensitivity analyses were employed to assess the robustness of the results. Results A total of 28,485 participants aged 40 years and older were recruited for this study. After fully adjusting for covariates, higher PIV levels were independently associated with increased COPD prevalence (OR = 1.67; 95% CI: 1.39-2.01) and all-cause mortality (HR = 2.04; 95% CI: 1.41-2.95). The COPD prevalence curve exhibited an inflection point at Log10-PIV of 2.24, showing no significant correlation on the left side (OR = 0.86; 95% CI: 0.45-1.64) but a positive correlation on the right side (OR = 2.00; 95% CI: 1.57-2.55). The COPD all-cause mortality curve displayed an inflection point at Log10-PIV of 2.38, indicating a negative correlation on the left side (HR = 0.23; 95% CI: 0.12-0.44) and a positive correlation on the right side (HR = 4.12; 95% CI: 2.62-6.48). Subgroup analyses with interaction tests showed that the strength of the correlation between PIV and COPD prevalence was influenced by race, smoking status, and BMI (all p for interaction <0.05). The relationship between PIV and COPD all-cause mortality was unaffected by any covariates (all p for interaction >0.05). Conclusion Elevated PIV levels are associated with increased COPD prevalence. COPD patients with either elevated or reduced PIV levels experience higher all-cause mortality. Further large-scale, longitudinal studies are required to corroborate these findings.
Collapse
Affiliation(s)
| | | | - Yang Li
- Department of Respiratory and Critical Care Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
30
|
Churchhouse AMD, Billard CV, Suzuki T, Pohl SÖG, Doleschall NJ, Donnelly K, Nixon C, Arends MJ, Din S, Kirkwood K, Marques Junior J, Von Kriegsheim A, Coffelt SB, Myant KB. Loss of DOCK2 potentiates Inflammatory Bowel Disease-associated colorectal cancer via immune dysfunction and IFNγ induction of IDO1 expression. Oncogene 2024; 43:3094-3107. [PMID: 39242821 PMCID: PMC11473400 DOI: 10.1038/s41388-024-03135-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 09/09/2024]
Abstract
Inflammatory Bowel Disease-associated colorectal cancer (IBD-CRC) is a known and serious complication of Inflammatory Bowel Disease (IBD) affecting the colon. However, relatively little is known about the pathogenesis of IBD-associated colorectal cancer in comparison with its sporadic cancer counterpart. Here, we investigated the function of Dock2, a gene mutated in ~10% of IBD-associated colorectal cancers that encodes a guanine nucleotide exchange factor (GEF). Using a genetically engineered mouse model of IBD-CRC, we found that whole body loss of Dock2 increases tumourigenesis via immune dysregulation. Dock2-deficient tumours displayed increased levels of IFNγ-associated genes, including the tryptophan metabolising, immune modulatory enzyme, IDO1, when compared to Dock2-proficient tumours. This phenotype was driven by increased IFNγ-production in T cell populations, which infiltrated Dock2-deficient tumours, promoting IDO1 expression in tumour epithelial cells. We show that IDO1 inhibition delays tumourigenesis in Dock2 knockout mice, and we confirm that this pathway is conserved across species as IDO1 expression is elevated in human IBD-CRC and in sporadic CRC cases with mutated DOCK2. Together, these data demonstrate a previously unidentified tumour suppressive role of DOCK2 that limits IFNγ-induced IDO1 expression and cancer progression, opening potential new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Antonia M D Churchhouse
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital Campus, Edinburgh, UK
| | - Caroline V Billard
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital Campus, Edinburgh, UK
| | - Toshiyasu Suzuki
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Sebastian Ö G Pohl
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital Campus, Edinburgh, UK
| | - Nora J Doleschall
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital Campus, Edinburgh, UK
| | - Kevin Donnelly
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital Campus, Edinburgh, UK
| | - Colin Nixon
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, UK
| | - Mark J Arends
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital Campus, Edinburgh, UK
| | - Shahida Din
- Edinburgh IBD Unit, Western General Hospital, Edinburgh, UK
| | - Kathryn Kirkwood
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - Jair Marques Junior
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital Campus, Edinburgh, UK
| | - Alex Von Kriegsheim
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital Campus, Edinburgh, UK
| | - Seth B Coffelt
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Kevin B Myant
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital Campus, Edinburgh, UK.
| |
Collapse
|
31
|
Uchańska A, Morytko A, Kwiecień K, Oleszycka E, Grygier B, Cichy J, Kwiecińska P. Lazy neutrophils - a lack of DGAT1 reduces the chemotactic activity of mouse neutrophils. Inflamm Res 2024; 73:1631-1643. [PMID: 39043892 PMCID: PMC11445369 DOI: 10.1007/s00011-024-01920-6] [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: 07/25/2023] [Revised: 05/07/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Neutrophils are key players in the innate immune system, actively migrating to sites of inflammation in the highly energetic process of chemotaxis. In this study, we focus on the role of acyl-CoA: diacylglycerol acyltransferase 1 (DGAT1), an enzyme that catalyzes the synthesis of triglycerides, the major form of stored energy, in neutrophil chemotaxis. METHODS AND RESULTS Using a mouse model of psoriasis, we show that DGAT1-deficiency reduces energy-demanding neutrophil infiltration to the site of inflammation, but this inhibition is not caused by decreased glycolysis and reduced ATP production by neutrophils lacking DGAT1. Flow cytometry and immunohistochemistry analysis demonstrate that DGAT1 also does not influence lipid accumulation in lipid droplets during inflammation. Interestingly, as has been shown previously, a lack of DGAT1 leads to an increase in the concentration of retinoic acid, and here, using real-time PCR and publicly-available next-generation RNA sequencing datasets, we show the upregulation of retinoic acid-responsive genes in Dgat1KO neutrophils. Furthermore, supplementation of WT neutrophils with exogenous retinoic acid mimics DGAT1-deficiency in the inhibition of neutrophil chemotaxis in in vitro transwell assay. CONCLUSIONS These results suggest that impaired skin infiltration by neutrophils in Dgat1KO mice is a result of the inhibitory action of an increased concentration of retinoic acid, rather than impaired lipid metabolism in DGAT1-deficient mice.
Collapse
Affiliation(s)
- Alicja Uchańska
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
- Selvita S.A, Cracow, Poland
| | - Agnieszka Morytko
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Kamila Kwiecień
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Ewa Oleszycka
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Beata Grygier
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Science, Cracow, Poland
| | - Joanna Cichy
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Patrycja Kwiecińska
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland.
- Laboratory of Stem Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland.
| |
Collapse
|
32
|
Cui D, Yu T. Unveiling the glycolysis in sepsis: Integrated bioinformatics and machine learning analysis identifies crucial roles for IER3, DSC2, and PPARG in disease pathogenesis. Medicine (Baltimore) 2024; 103:e39867. [PMID: 39331858 PMCID: PMC11441936 DOI: 10.1097/md.0000000000039867] [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: 11/23/2023] [Accepted: 08/26/2024] [Indexed: 09/29/2024] Open
Abstract
Sepsis, a multifaceted syndrome driven by an imbalanced host response to infection, remains a significant medical challenge. At its core lies the pivotal role of glycolysis, orchestrating immune responses especially in severe sepsis. The intertwined dynamics between glycolysis, sepsis, and immunity, however, have gaps in knowledge with several Crucial genes still shrouded in ambiguity. We harvested transcriptomic profiles from the peripheral blood of 107 septic patients juxtaposed against 29 healthy controls. Delving into this dataset, differential expression analysis shed light on genes distinctly linked to glycolysis in both cohorts. Harnessing the prowess of LASSO regression and SVM-RFE, we isolated Crucial genes, paving the way for a sepsis risk prediction model, subsequently vetted via Calibration and decision curve analysis. Using the CIBERSORT algorithm, we further mapped 22 immune cell subtypes within the septic samples, establishing potential interactions with the delineated Crucial genes. Our efforts unveiled 21 genes intricately tied to glycolysis that exhibited differential expression patterns. Gene set enrichment analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses offered insights, spotlighting pathways predominantly associated with oxidative phosphorylation, PPAR signaling pathway, Glycolysis/Gluconeogenesis and HIF-1 signaling pathway. Among the myriad genes, IER3, DSC2, and PPARG emerged as linchpins, their prominence in sepsis further validated through ROC analytics. These sentinel genes demonstrated profound affiliations with various immune cell facets, bridging the complex terrain of glycolysis, sepsis, and immune responses. In line with our endeavor to "unveil the glycolysis in sepsis," the discovery of IER3, DSC2, and PPARG reinforces their cardinal roles in sepsis pathogenesis. These revelations accentuate the intricate dance between glycolysis and immunological shifts in septic conditions, offering novel avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Dongqing Cui
- Emergency Department, Beijing Sixth Hospital, Beijing, China
| | - Tian Yu
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
33
|
Bobrovskikh AV, Zubairova US, Naumenko LG, Doroshkov AV. Catching the Big Fish in Big Data: A Meta-Analysis of Zebrafish Kidney scRNA-Seq Datasets Highlights Conserved Molecular Profiles of Macrophages and Neutrophils in Vertebrates. BIOLOGY 2024; 13:773. [PMID: 39452082 PMCID: PMC11505477 DOI: 10.3390/biology13100773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
The innate immune system (IIS) is an ancient and essential defense mechanism that protects animals against a wide range of pathogens and diseases. Although extensively studied in mammals, our understanding of the IIS in other taxa remains limited. The zebrafish (Danio rerio) serves as a promising model organism for investigating IIS-related processes, yet the immunogenetics of fish are not fully elucidated. To address this gap, we conducted a meta-analysis of single-cell RNA sequencing (scRNA-seq) datasets from zebrafish kidney marrow, encompassing approximately 250,000 immune cells. Our analysis confirms the presence of key genetic pathways in zebrafish innate immune cells that are similar to those identified in mammals. Zebrafish macrophages specifically express genes encoding cathepsins, major histocompatibility complex class II proteins, integral membrane proteins, and the V-ATPase complex and demonstrate the enrichment of oxidative phosphorylation ferroptosis processes. Neutrophils are characterized by the significant expression of genes encoding actins, cytoskeleton organizing proteins, the Arp2/3 complex, and glycolysis enzymes and have demonstrated their involvement in GnRH and CLR signaling pathways, adherents, and tight junctions. Both macrophages and neutrophils highly express genes of NOD-like receptors, phagosomes, and lysosome pathways and genes involved in apoptosis. Our findings reinforce the idea about the existence of a wide spectrum of immune cell phenotypes in fish since we found only a small number of cells with clear pro- or anti-inflammatory signatures.
Collapse
Affiliation(s)
- Aleksandr V. Bobrovskikh
- Department of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia;
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
| | - Ulyana S. Zubairova
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
- Department of Information Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Ludmila G. Naumenko
- Department of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia;
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
| | - Alexey V. Doroshkov
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
- Department of Genomics and Bioinformatics, Institute of Fundamental Biology and Biotechnology, Siberian Federal University, 660036 Krasnoyarsk, Russia
| |
Collapse
|
34
|
Britt EC, Qing X, Votava JA, Lika J, Wagner AS, Shen S, Arp NL, Khan H, Schieke SM, Fletcher CD, Huttenlocher A, Fan J. Activation induces shift in nutrient utilization that differentially impacts cell functions in human neutrophils. Proc Natl Acad Sci U S A 2024; 121:e2321212121. [PMID: 39284072 PMCID: PMC11441510 DOI: 10.1073/pnas.2321212121] [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: 12/07/2023] [Accepted: 07/29/2024] [Indexed: 09/25/2024] Open
Abstract
Neutrophils utilize a variety of metabolic sources to support their crucial functions as the first responders in innate immunity. Here, through in vivo and ex vivo isotopic tracing, we examined the contributions of different nutrients to neutrophil metabolism under specific conditions. Human peripheral blood neutrophils, in contrast to a neutrophil-like cell line, rely on glycogen storage as a major metabolic source under resting state but rapidly switch to primarily using extracellular glucose upon activation with various stimuli. This shift is driven by a substantial increase in glucose uptake, enabled by rapidly increased GLUT1 on cell membrane, that dominates the simultaneous increase in gross glycogen cycling capacity. Shifts in nutrient utilization impact neutrophil functions in a function-specific manner: oxidative burst depends on glucose utilization, whereas NETosis and phagocytosis can be flexibly supported by either glucose or glycogen, and neutrophil migration and fungal control are enhanced by the shift from glycogen utilization to glucose utilization. This work provides a quantitative and dynamic understanding of fundamental features in neutrophil metabolism and elucidates how metabolic remodeling shapes neutrophil functions, which has broad health relevance.
Collapse
Affiliation(s)
- Emily C. Britt
- Morgridge Institute for Research, Madison, WI53715
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI53706
| | - Xin Qing
- Morgridge Institute for Research, Madison, WI53715
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI53706
| | | | - Jorgo Lika
- Morgridge Institute for Research, Madison, WI53715
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI53706
- University of Wisconsin Medical Scientist Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI53792
| | - Andrew S. Wagner
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI53706
| | - Simone Shen
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI53706
| | - Nicholas L. Arp
- Morgridge Institute for Research, Madison, WI53715
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI53706
- University of Wisconsin Medical Scientist Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI53792
| | - Hamidullah Khan
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI53715
- Department of Dermatology, Georgetown University Medical Center Washington DC VA Medical Center, Washington, DC20036
| | - Stefan M. Schieke
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI53715
- Department of Dermatology, Georgetown University Medical Center Washington DC VA Medical Center, Washington, DC20036
| | | | - Anna Huttenlocher
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI53706
- University of Wisconsin Medical Scientist Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI53792
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI53706
- University of Wisconsin Carbone Cancer Center, Madison, WI53792
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI53792
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI53715
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI53706
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI53706
- University of Wisconsin Medical Scientist Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI53792
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI53706
- University of Wisconsin Carbone Cancer Center, Madison, WI53792
| |
Collapse
|
35
|
Ettel P, Weichhart T. Not just sugar: metabolic control of neutrophil development and effector functions. J Leukoc Biol 2024; 116:487-510. [PMID: 38450755 PMCID: PMC7617515 DOI: 10.1093/jleuko/qiae057] [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: 11/30/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024] Open
Abstract
The mammalian immune system is constantly surveying our tissues to clear pathogens and maintain tissue homeostasis. In order to fulfill these tasks, immune cells take up nutrients to supply energy for survival and for directly regulating effector functions via their cellular metabolism, a process now known as immunometabolism. Neutrophilic granulocytes, the most abundant leukocytes in the human body, have a short half-life and are permanently needed in the defense against pathogens. According to a long-standing view, neutrophils were thought to primarily fuel their metabolic demands via glycolysis. Yet, this view has been challenged, as other metabolic pathways recently emerged to contribute to neutrophil homeostasis and effector functions. In particular during neutrophilic development, the pentose phosphate pathway, glycogen synthesis, oxidative phosphorylation, and fatty acid oxidation crucially promote neutrophil maturation. At steady state, both glucose and lipid metabolism sustain neutrophil survival and maintain the intracellular redox balance. This review aims to comprehensively discuss how neutrophilic metabolism adapts during development, which metabolic pathways fuel their functionality, and how these processes are reconfigured in case of various diseases. We provide several examples of hereditary diseases, in which mutations in metabolic enzymes validate their critical role for neutrophil function.
Collapse
Affiliation(s)
- Paul Ettel
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090Vienna, Austria
| | - Thomas Weichhart
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090Vienna, Austria
| |
Collapse
|
36
|
Watts E, Willison J, Arienti S, Sadiku P, Coelho P, Sanchez-Garcia M, Zhang A, Murphy F, Dickinson R, Mirchandani A, Morrison T, Lewis A, Vermaelen W, Ghesquiere B, Carmeliet P, Mazzone M, Maxwell P, Pugh C, Dockrell D, Whyte M, Walmsley S. Differential roles for the oxygen sensing enzymes PHD1 and PHD3 in the regulation of neutrophil metabolism and function. Wellcome Open Res 2024; 8:569. [PMID: 39257914 PMCID: PMC11384204 DOI: 10.12688/wellcomeopenres.19915.2] [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] [Accepted: 08/06/2024] [Indexed: 09/12/2024] Open
Abstract
Background Neutrophils are essential in the early innate immune response to pathogens. Harnessing their antimicrobial powers, without driving excessive and damaging inflammatory responses, represents an attractive therapeutic possibility. The neutrophil population is increasingly recognised to be more diverse and malleable than was previously appreciated. Hypoxic signalling pathways are known to regulate important neutrophil behaviours and, as such, are potential therapeutic targets for regulating neutrophil antimicrobial and inflammatory responses. Methods We used a combination of in vivo and ex vivo models, utilising neutrophil and myeloid specific PHD1 or PHD3 deficient mouse lines to investigate the roles of oxygen sensing prolyl hydroxylase enzymes in the regulation of neutrophilic inflammation and immunity. Mass spectrometry and Seahorse metabolic flux assays were used to analyse the role of metabolic shifts in driving the downstream phenotypes. Results We found that PHD1 deficiency drives alterations in neutrophil metabolism and recruitment, in an oxygen dependent fashion. Despite this, PHD1 deficiency did not significantly alter ex vivo neutrophil phenotypes or in vivo outcomes in mouse models of inflammation. Conversely, PHD3 deficiency was found to enhance neutrophil antibacterial properties without excessive inflammatory responses. This was not linked to changes in the abundance of core metabolites but was associated with increased oxygen consumption and increased mitochondrial reactive oxygen species (mROS) production. Conclusions PHD3 deficiency drives a favourable neutrophil phenotype in infection and, as such, is an important potential therapeutic target.
Collapse
Affiliation(s)
- Emily Watts
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Joseph Willison
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Simone Arienti
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Pranvera Sadiku
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Patricia Coelho
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Manuel Sanchez-Garcia
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Ailiang Zhang
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Fiona Murphy
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, G4 0RE, UK
| | - Rebecca Dickinson
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Ananda Mirchandani
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Tyler Morrison
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Amy Lewis
- The Bateson Centre, Department of Infection and Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, England, S10 2TN, UK
| | - Wesley Vermaelen
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Flanders, Belgium
- Metabolomics Core Facility, Vlaams Instituut voor Biotechnologie KU Leuven Center for Cancer Biology, Leuven, Flanders, Belgium
| | - Bart Ghesquiere
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Flanders, Belgium
- Metabolomics Core Facility, Vlaams Instituut voor Biotechnologie KU Leuven Center for Cancer Biology, Leuven, Flanders, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Vlaams Instituut voor Biotechnologie KU Leuven Center for Cancer Biology, Leuven, Flanders, Belgium
| | - Massimilliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis (VIB-KU Leuven), KU Leuven, Leuven, Flanders, Belgium
| | - Patrick Maxwell
- School of Clinical Medicine, University of Cambridge, Cambridge, England, UK
| | - Christopher Pugh
- Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - David Dockrell
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Moira Whyte
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| | - Sarah Walmsley
- Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, EH16 4UU, UK
| |
Collapse
|
37
|
Faherty L, Zhang WZ, Salih MM, Robinson EK, Perez E, Kim K, Carpenter S, Cloonan SM. Transcriptomic analysis reveals distinct effects of cigarette smoke on murine airspace and bone-marrow derived macrophages. Respir Res 2024; 25:322. [PMID: 39182076 PMCID: PMC11344945 DOI: 10.1186/s12931-024-02939-3] [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: 05/10/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is an inflammatory airway disease characterized by emphysema and chronic bronchitis and a leading cause of mortality worldwide. COPD is commonly associated with several comorbid diseases which contribute to exacerbated patient outcomes. Cigarette smoke (CS) is the most prominent risk factor for COPD development and progression and is known to be detrimental to numerous effector functions of lung resident immune cells, including phagocytosis and cytokine production. However, how CS mediates the various pathologies distant from the lung in COPD, and whether CS has a similar biological effect on systemic immune cells remains unknown. METHODS C57BL/6 mice were exposed to 8 weeks of CS as an experimental model of COPD. Bone marrow cells were isolated from both CS-exposed and room air (RA) control mice and differentiated to bone marrow-derived macrophages (BMDMs). Airspace macrophages (AMs) were isolated from the same CS-exposed and RA mice and bulk RNA-Seq performed. The functional role of differentially expressed genes was assessed through gene ontology analyses. Ingenuity Pathway Analysis was used to determine the activation states of canonical pathways and upstream regulators enriched in differentially expressed genes in both cell types, and to compare the differences between the two cell types. RESULTS CS induced transcriptomic changes in BMDMs, including an upregulation of genes in sirtuin signalling and oxidative phosphorylation pathways and a downregulation of genes involved in histone and lysine methylation. In contrast, CS induced decreased expression of genes involved in pathogen response, phagosome formation, and immune cell trafficking in AMs. Little overlap was observed in differentially expressed protein-coding genes in BMDMs compared to AMs and their associated pathways, highlighting the distinct effects of CS on immune cells in different compartments. CONCLUSIONS CS exposure can induce transcriptomic remodelling in BMDMs which is distinct to that of AMs. Our study highlights the ability of CS exposure to affect immune cell populations distal to the lung and warrants further investigation into the functional effects of these changes and the ensuing role in driving multimorbid disease.
Collapse
Affiliation(s)
- Lynne Faherty
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Tallaght University Hospital, Dublin, Ireland
| | - William Z Zhang
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Mays M Salih
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Elektra K Robinson
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Elizabeth Perez
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Kihwan Kim
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Susan Carpenter
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Suzanne M Cloonan
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
- Tallaght University Hospital, Dublin, Ireland.
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA.
| |
Collapse
|
38
|
Liao F, Scozzi D, Zhou D, Maksimos M, Diedrich C, Cano M, Tague LK, Liu Z, Haspel JA, Leonard JM, Li W, Krupnick AS, Wong BW, Kreisel D, Azab AK, Gelman AE. Nanoparticle targeting of neutrophil glycolysis prevents lung ischemia-reperfusion injury. Am J Transplant 2024; 24:1382-1394. [PMID: 38522826 PMCID: PMC11305958 DOI: 10.1016/j.ajt.2024.03.028] [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: 09/21/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
Neutrophils exacerbate pulmonary ischemia-reperfusion injury (IRI) resulting in poor short and long-term outcomes for lung transplant recipients. Glycolysis powers neutrophil activation, but it remains unclear if neutrophil-specific targeting of this pathway will inhibit IRI. Lipid nanoparticles containing the glycolysis flux inhibitor 2-deoxyglucose (2-DG) were conjugated to neutrophil-specific Ly6G antibodies (NP-Ly6G[2-DG]). Intravenously administered NP-Ly6G(2-DG) to mice exhibited high specificity for circulating neutrophils. NP-Ly6G(2-DG)-treated neutrophils were unable to adapt to hypoglycemic conditions of the lung airspace environment as evident by the loss of demand-induced glycolysis, reductions in glycogen and ATP content, and an increased vulnerability to apoptosis. NP-Ly6G(2-DG) treatment inhibited pulmonary IRI following hilar occlusion and orthotopic lung transplantation. IRI protection was associated with less airspace neutrophil extracellular trap generation, reduced intragraft neutrophilia, and enhanced alveolar macrophage efferocytotic clearance of neutrophils. Collectively, our data show that pharmacologically targeting glycolysis in neutrophils inhibits their activation and survival leading to reduced pulmonary IRI.
Collapse
Affiliation(s)
- Fuyi Liao
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Davide Scozzi
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dequan Zhou
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mina Maksimos
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Camila Diedrich
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Marlene Cano
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laneshia K Tague
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zhyi Liu
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jeffrey A Haspel
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jennifer M Leonard
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Wenjun Li
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alexander S Krupnick
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Brian W Wong
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Abdel Kareem Azab
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, Texas, USA.
| | - Andrew E Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
39
|
de Sena-Tomás C, Rebola Lameira L, Rebocho da Costa M, Naique Taborda P, Laborde A, Orger M, de Oliveira S, Saúde L. Neutrophil immune profile guides spinal cord regeneration in zebrafish. Brain Behav Immun 2024; 120:514-531. [PMID: 38925414 DOI: 10.1016/j.bbi.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/15/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024] Open
Abstract
Spinal cord injury triggers a strong innate inflammatory response in both non-regenerative mammals and regenerative zebrafish. Neutrophils are the first immune population to be recruited to the injury site. Yet, their role in the repair process, particularly in a regenerative context, remains largely unknown. Here, we show that, following rapid recruitment to the injured spinal cord, neutrophils mostly reverse migrate throughout the zebrafish body. In addition, promoting neutrophil inflammation resolution by inhibiting Cxcr4 boosts cellular and functional regeneration. Neutrophil-specific RNA-seq analysis reveals an enhanced activation state that correlates with a transient increase in tnf-α expression in macrophage/microglia populations. Conversely, blocking neutrophil recruitment through Cxcr1/2 inhibition diminishes the presence of macrophage/microglia at the injury site and impairs spinal cord regeneration. Altogether, these findings provide new insights into the role of neutrophils in spinal cord regeneration, emphasizing the significant impact of their immune profile on the outcome of the repair process.
Collapse
Affiliation(s)
- Carmen de Sena-Tomás
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Leonor Rebola Lameira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Mariana Rebocho da Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Patrícia Naique Taborda
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Alexandre Laborde
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal
| | - Michael Orger
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal
| | - Sofia de Oliveira
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine (Hepatology), Albert Einstein College of Medicine, Bronx, NY 10461, USA; Harold and Muriel Block Institute for Clinical and Translational Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Montefiore-Einstein Comprehensive Cancer Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Leonor Saúde
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Histologia e Biologia de Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| |
Collapse
|
40
|
Gammariello CS, Hanson J, Relling AE, Oliveira MXS, Sipka AS, Enger KM, Enger BD. Localized mammary gland changes in milk composition and venous blood metabolite concentrations result from sterile subclinical mastitis. J Dairy Sci 2024; 107:6148-6160. [PMID: 38608954 DOI: 10.3168/jds.2023-24044] [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: 08/03/2023] [Accepted: 03/06/2024] [Indexed: 04/14/2024]
Abstract
Subclinical mastitis reduces milk yield and elicits undesirable changes in milk composition, but the mechanisms resulting in reduced milk production in affected mammary glands are incompletely understood. This study investigated the effects of sterile inflammation on mammary gland metabolism by assessing changes in milk and venous blood composition. Mid-lactation primiparous Holstein cows (n = 4) had udder halves randomly allocated to treatments; quarters of 1 udder half were infused with 2 billion cfu of formalin-fixed Staphylococcus aureus (FX-STAPH) and quarters of the opposite udder half were infused with saline (SAL). Blood samples were collected from the right and left subcutaneous abdominal veins in 2.6 h intervals until 40 h postchallenge and analyzed for blood gas and metabolite concentrations. Milk from FX-STAPH udder halves had significantly increased SCS by the first milking at 8 h postchallenge. By 16 h postchallenge, FX-STAPH udder halves had increased concentrations of protein and lactate and lower lactose concentrations than SAL udder halves. Milk fat concentrations, milk yields, ECM yields, and the ferric reducing antioxidant power of milk were not significantly different between SAL and FX-STAPH udder halves. Venous blood of FX-STAPH halves had marginally greater concentrations of saturated O2, partial pressures of O2, and glucose concentrations than SAL halves. Conversely, total and partial pressures of CO2 did not differ between udder half treatments, suggesting a shift in local metabolite utilization in FX-STAPH udder halves. These results indicate that changes in milk composition resulting from mastitis are accompanied by changes in some key blood metabolite concentrations. The shift in venous blood metabolite concentrations, along with the marked increase in milk lactate, suggests that local mammary tissue or recruited immune cells, or both, alter metabolite usage in mammary tissues. Future studies are needed to quantify the uptake of key milk precursors during mastitis.
Collapse
Affiliation(s)
- C S Gammariello
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691
| | - J Hanson
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691
| | - A E Relling
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691
| | - M X S Oliveira
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691
| | - A S Sipka
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - K M Enger
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691
| | - B D Enger
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691.
| |
Collapse
|
41
|
Min-Jeong K, Akula HK, Marden J, Li K, Hu B, Vaska P, Qu W. The potential utility of (2S,4R)-4-[18F] fluoroglutamine as a novel metabolic imaging marker for inflammation explored by rat models of arthritis and paw edema. RESEARCH SQUARE 2024:rs.3.rs-4493375. [PMID: 38947024 PMCID: PMC11213212 DOI: 10.21203/rs.3.rs-4493375/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Purpose (2S,4R)-4-[18F]fluoroglutamine ([18F]FGln) is a promising metabolic imaging marker in cancer. Based on the fact that major inflammatory cells are heavily dependent on glutamine metabolism like cancer cells, we explored the potential utility of [18F]FGln as a metabolic imaging marker for inflammation in two rat models: carrageenan-induced paw edema (CIPE) and collagen-induced arthritis (CIA). Procedures The CIPE model (n = 4) was generated by injecting 200 μL of 3% carrageenan solution into the left hind paw three hours before the PET. The CIA model (n = 4) was generated by injecting 200 μg of collagen emulsion subcutaneously at the tail base 3-4 weeks before the PET. A qualitative scoring system was used to assess the severity of paw inflammation. After a CT scan, 15.7 ± 4.9 MBq of [18F]FGln was injected via the tail vein, followed by a dynamic micro-PET scan for 90 minutes under anesthesia with isoflurane. The standard uptake value of [18F]FGln was measured by placing a volume of interest in each paw. The non-injected right hind paws of the CIPE model rats served as controls for both models. The paws with CIA were pathologically examined after PET. Results In CIPE models, uptake in the injected paw was higher compared to the non-injected paw by 52-83%. In CIA models, uptake in the paws with severe inflammation was higher than the averaged controls by 54-173%, while that with mild and no inflammation was slightly higher (33%) and lower (-7%), respectively. Combined overall, the [18F]FGln uptake in CIA showed a significant positive correlation with inflammation severity (r = 0.88, P = 0.009). The pathological findings confirmed profound inflammation in CIA. Conclusions [18F]FGln uptake was increased in both acute and chronic inflammation, and the uptake level was significantly correlated with the severity, suggesting its potential utility as a novel metabolic imaging marker for inflammation.
Collapse
Affiliation(s)
- Kim Min-Jeong
- Stony Brook University Health Sciences Center School of Medicine: Stony Brook University Renaissance School of Medicine
| | - Hari K Akula
- Stony Brook University Health Sciences Center School of Medicine: Stony Brook University Renaissance School of Medicine
| | - Jocelyn Marden
- Stony Brook University Health Sciences Center School of Medicine: Stony Brook University Renaissance School of Medicine
| | | | - Bao Hu
- Stony Brook University Health Sciences Center School of Medicine: Stony Brook University Renaissance School of Medicine
| | - Paul Vaska
- Stony Brook University Health Sciences Center School of Medicine: Stony Brook University Renaissance School of Medicine
| | - Wenchao Qu
- Stony Brook University Health Sciences Center School of Medicine: Stony Brook University Renaissance School of Medicine
| |
Collapse
|
42
|
Tambralli A, Harbaugh A, NaveenKumar SK, Radyk MD, Rysenga CE, Sabb K, Hurley JM, Sule GJ, Yalavarthi S, Estes SK, Hoy CK, Smith T, Sarosh C, Madison JA, Schaefer JK, Sood SL, Zuo Y, Sawalha AH, Lyssiotis CA, Knight JS. Neutrophil glucose flux as a therapeutic target in antiphospholipid syndrome. J Clin Invest 2024; 134:e169893. [PMID: 38869951 PMCID: PMC11290966 DOI: 10.1172/jci169893] [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: 02/20/2023] [Accepted: 06/11/2024] [Indexed: 06/15/2024] Open
Abstract
Neutrophil hyperactivity and neutrophil extracellular trap release (NETosis) appear to play important roles in the pathogenesis of the thromboinflammatory autoimmune disease known as antiphospholipid syndrome (APS). The understanding of neutrophil metabolism has advanced tremendously in the past decade, and accumulating evidence suggests that a variety of metabolic pathways guide neutrophil activities in health and disease. Our previous work characterizing the transcriptome of APS neutrophils revealed that genes related to glycolysis, glycogenolysis, and the pentose phosphate pathway (PPP) were significantly upregulated. Here, we found that neutrophils from patients with APS used glycolysis more avidly than neutrophils from people in the healthy control group, especially when the neutrophils were from patients with APS with a history of microvascular disease. In vitro, inhibiting either glycolysis or the PPP tempered phorbol myristate acetate- and APS IgG-induced NETosis, but not NETosis triggered by a calcium ionophore. In mice, inhibiting either glycolysis or the PPP reduced neutrophil reactive oxygen species production and suppressed APS IgG-induced NETosis ex vivo. When APS-associated thrombosis was evaluated in mice, inhibiting either glycolysis or the PPP markedly suppressed thrombosis and circulating NET remnants. In summary, these data identify a potential role for restraining neutrophil glucose flux in the treatment of APS.
Collapse
Affiliation(s)
- Ajay Tambralli
- Division of Rheumatology, Department of Internal Medicine
- Division of Pediatric Rheumatology, Department of Pediatrics
| | | | | | | | | | - Kaitlyn Sabb
- Division of Rheumatology, Department of Internal Medicine
| | | | - Gautam J. Sule
- Division of Rheumatology, Department of Internal Medicine
| | | | | | - Claire K. Hoy
- Division of Rheumatology, Department of Internal Medicine
| | - Tristin Smith
- Division of Rheumatology, Department of Internal Medicine
| | - Cyrus Sarosh
- Division of Rheumatology, Department of Internal Medicine
| | - Jacqueline A. Madison
- Division of Rheumatology, Department of Internal Medicine
- Division of Pediatric Rheumatology, Department of Pediatrics
| | - Jordan K. Schaefer
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Suman L. Sood
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yu Zuo
- Division of Rheumatology, Department of Internal Medicine
| | - Amr H. Sawalha
- Departments of Pediatrics, Medicine, and Immunology, and Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
43
|
Taniguchi T, Okahashi N, Matsuda F. 13C-metabolic flux analysis reveals metabolic rewiring in HL-60 neutrophil-like cells through differentiation and immune stimulation. Metab Eng Commun 2024; 18:e00239. [PMID: 38883865 PMCID: PMC11176794 DOI: 10.1016/j.mec.2024.e00239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/18/2024] Open
Abstract
Neutrophils are innate immune cells and the first line of defense for the maintenance of homeostasis. However, our knowledge of the metabolic rewiring associated with their differentiation and immune stimulation is limited. Here, quantitative 13C-metabolic flux analysis was performed using HL-60 cells as the neutrophil model. A metabolic model for 13C-metabolic flux analysis of neutrophils was developed based on the accumulation of 13C in intracellular metabolites derived from 13C-labeled extracellular carbon sources and intracellular macromolecules. Aspartate and glutamate in the medium were identified as carbon sources that enter central carbon metabolism. Furthermore, the breakdown of macromolecules, estimated to be fatty acids and nucleic acids, was observed. Based on these results, a modified metabolic model was used for 13C-metabolic flux analysis of undifferentiated, differentiated, and lipopolysaccharide (LPS)-activated HL-60 cells. The glucose uptake rate and glycolytic flux decreased with differentiation, whereas the tricarboxylic acid (TCA) cycle flux remained constant. The addition of LPS to differentiated HL-60 cells activated the glucose uptake rate and pentose phosphate pathway (PPP) flux levels, resulting in an increased rate of total NADPH regeneration, which could be used to generate reactive oxygen species. The flux levels of fatty acid degradation and synthesis were also increased in LPS-activated HL-60 cells. Overall, this study highlights the quantitative metabolic alterations in multiple pathways via the differentiation and activation of HL-60 cells using 13C-metabolic flux analysis.
Collapse
Affiliation(s)
- Takeo Taniguchi
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Nobuyuki Okahashi
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Biotechnology, Osaka University Shimadzu Analytical Innovation Research Laboratory, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Industrial Biotechnology Initiative Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Fumio Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Biotechnology, Osaka University Shimadzu Analytical Innovation Research Laboratory, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Industrial Biotechnology Initiative Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
44
|
Fang Y, Li Z, Yang L, Li W, Wang Y, Kong Z, Miao J, Chen Y, Bian Y, Zeng L. Emerging roles of lactate in acute and chronic inflammation. Cell Commun Signal 2024; 22:276. [PMID: 38755659 PMCID: PMC11097486 DOI: 10.1186/s12964-024-01624-8] [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: 01/01/2024] [Accepted: 04/20/2024] [Indexed: 05/18/2024] Open
Abstract
Traditionally, lactate has been considered a 'waste product' of cellular metabolism. Recent findings have shown that lactate is a substance that plays an indispensable role in various physiological cellular functions and contributes to energy metabolism and signal transduction during immune and inflammatory responses. The discovery of lactylation further revealed the role of lactate in regulating inflammatory processes. In this review, we comprehensively summarize the paradoxical characteristics of lactate metabolism in the inflammatory microenvironment and highlight the pivotal roles of lactate homeostasis, the lactate shuttle, and lactylation ('lactate clock') in acute and chronic inflammatory responses from a molecular perspective. We especially focused on lactate and lactate receptors with either proinflammatory or anti-inflammatory effects on complex molecular biological signalling pathways and investigated the dynamic changes in inflammatory immune cells in the lactate-related inflammatory microenvironment. Moreover, we reviewed progress on the use of lactate as a therapeutic target for regulating the inflammatory response, which may provide a new perspective for treating inflammation-related diseases.
Collapse
Affiliation(s)
- Yunda Fang
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhengjun Li
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- College of Health Economics Management, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lili Yang
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jingwen Library, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wen Li
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Acupuncture-Moxibustion and Tuina, ·School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yutong Wang
- School of Acupuncture-Moxibustion and Tuina, ·School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ziyang Kong
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Acupuncture-Moxibustion and Tuina, ·School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia Miao
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yanqi Chen
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yaoyao Bian
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- School of Acupuncture-Moxibustion and Tuina, ·School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- TCM Rehabilitation Center, Jiangsu Second Chinese Medicine Hospital, Nanjing, 210023, China.
| | - Li Zeng
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, 999078, China.
| |
Collapse
|
45
|
Wu X, Fan Y, Wang K, Miao Y, Chang Y, Ming J, Wang X, Lu S, Liu R, Zhang F, Zhang Y, Qin H, Shi J. NIR-II imaging-guided precise photodynamic therapy for augmenting tumor-starvation therapy by glucose metabolism reprogramming interference. Sci Bull (Beijing) 2024; 69:1263-1274. [PMID: 38418300 DOI: 10.1016/j.scib.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/31/2023] [Accepted: 02/02/2024] [Indexed: 03/01/2024]
Abstract
Metabolic reprogramming is a mechanism by which cancer cells alter their metabolic patterns to promote cell proliferation and growth, thereby enabling their resistance to external stress. 2-Deoxy-D-glucose (2DG) can eliminate their energy source by inhibiting glucose glycolysis, leading to cancer cell death through starvation. However, a compensatory increase in mitochondrial metabolism inhibits its efficacy. Herein, we propose a synergistic approach that combines photodynamic therapy (PDT) with starvation therapy to address this challenge. To monitor the nanodrugs and determine the optimal triggering time for precise tumor therapy, a multifunctional nano-platform comprising lanthanide-doped nanoparticle (LnNP) cores was constructed and combined with mesoporous silicon shells loaded with 2DG and photosensitizer chlorin e6 (Ce6) in the mesopore channels. Under 980 nm near-infrared light excitation, the downshifted 1550 nm fluorescence signal in the second near-infrared (NIR-II, 1000-1700 nm) window from the LnNPs was used to monitor the accumulation of nanomaterials in tumors. Furthermore, upconverted 650 nm light excited the Ce6 to generate singlet oxygen for PDT, which damaged mitochondrial function and enhanced the efficacy of 2DG by inhibiting hexokinase 2 and lactate dehydrogenase A expressions. As a result, glucose metabolism reprogramming was inhibited and the efficiency of starvation therapy was significantly enhanced. Overall, the proposed NIR-II bioimaging-guided PDT-augmented starvation therapy, which simultaneously inhibited glycolysis and mitochondria, facilitated the effects of a cancer theranostic system.
Collapse
Affiliation(s)
- Xiawei Wu
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yong Fan
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers and iChem, Fudan University, Shanghai 200433, China
| | - Kairuo Wang
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yunqiu Miao
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yongliang Chang
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Jiang Ming
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers and iChem, Fudan University, Shanghai 200433, China
| | - Xinyue Wang
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Shengwei Lu
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Ruichi Liu
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Fan Zhang
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers and iChem, Fudan University, Shanghai 200433, China
| | - Yang Zhang
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| | - Huanlong Qin
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, China
| |
Collapse
|
46
|
Zhang Y, Nie Y, Liu X, Wan X, Shi Y, Zhang K, Wu P, He J. Tumor metabolic crosstalk and immunotherapy. Clin Transl Oncol 2024; 26:797-807. [PMID: 37740892 DOI: 10.1007/s12094-023-03304-4] [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: 04/13/2023] [Accepted: 08/08/2023] [Indexed: 09/25/2023]
Abstract
Tumor cells must resist the host's immune system while maintaining growth under harsh conditions of acidity and hypoxia, which indicates that tumors are more robust than normal tissue. Immunotherapeutic agents have little effect on solid tumors, mostly because of the tumor density and the difficulty of penetrating deeply into the tissue to achieve the theoretical therapeutic effect. Various therapeutic strategies targeting the tumor microenvironment (TME) have been developed. Immunometabolic disorders play a dominant role in treatment resistance at both the TME and host levels. Understanding immunometabolic factors and their treatment potential may be a way forward for tumor immunotherapy. Here, we summarize the metabolism of substances that affect tumor progression, the crosstalk between the TME and immunosuppression, and some potential tumor-site targets. We also summarize the progress and challenges of tumor immunotherapy.
Collapse
Affiliation(s)
- Yiwen Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yueli Nie
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- School of Pharmacy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xitian Wan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yuanyuan Shi
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Keyong Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Pan Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- School of Pharmacy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jian He
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- School of Pharmacy, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
47
|
Long MB, Howden AJM, Keir HR, Rollings CM, Giam YH, Pembridge T, Delgado L, Abo-Leyah H, Lloyd AF, Sollberger G, Hull R, Gilmour A, Hughes C, New BJM, Cassidy D, Shoemark A, Richardson H, Lamond AI, Cantrell DA, Chalmers JD, Brenes AJ. Extensive acute and sustained changes to neutrophil proteomes post-SARS-CoV-2 infection. Eur Respir J 2024; 63:2300787. [PMID: 38097207 PMCID: PMC10918319 DOI: 10.1183/13993003.00787-2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 11/23/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND Neutrophils are important in the pathophysiology of coronavirus disease 2019 (COVID-19), but the molecular changes contributing to altered neutrophil phenotypes following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are not fully understood. We used quantitative mass spectrometry-based proteomics to explore neutrophil phenotypes immediately following acute SARS-CoV-2 infection and during recovery. METHODS Prospective observational study of hospitalised patients with PCR-confirmed SARS-CoV-2 infection (May to December 2020). Patients were enrolled within 96 h of admission, with longitudinal sampling up to 29 days. Control groups comprised non-COVID-19 acute lower respiratory tract infection (LRTI) and age-matched noninfected controls. Neutrophils were isolated from peripheral blood and analysed using mass spectrometry. COVID-19 severity and recovery were defined using the World Health Organization ordinal scale. RESULTS Neutrophil proteomes from 84 COVID-19 patients were compared to those from 91 LRTI and 42 control participants. 5800 neutrophil proteins were identified, with >1700 proteins significantly changed in neutrophils from COVID-19 patients compared to noninfected controls. Neutrophils from COVID-19 patients initially all demonstrated a strong interferon signature, but this signature rapidly declined in patients with severe disease. Severe disease was associated with increased abundance of proteins involved in metabolism, immunosuppression and pattern recognition, while delayed recovery from COVID-19 was associated with decreased granule components and reduced abundance of metabolic proteins, chemokine and leukotriene receptors, integrins and inhibitory receptors. CONCLUSIONS SARS-CoV-2 infection results in the sustained presence of circulating neutrophils with distinct proteomes suggesting altered metabolic and immunosuppressive profiles and altered capacities to respond to migratory signals and cues from other immune cells, pathogens or cytokines.
Collapse
Affiliation(s)
- Merete B Long
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
- Indicates equal contribution
| | - Andrew J M Howden
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
- Indicates equal contribution
| | - Holly R Keir
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
- Indicates equal contribution
| | - Christina M Rollings
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
- Indicates equal contribution
| | - Yan Hui Giam
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Thomas Pembridge
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Lilia Delgado
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Hani Abo-Leyah
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Amy F Lloyd
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Gabriel Sollberger
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Rebecca Hull
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Amy Gilmour
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Chloe Hughes
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Benjamin J M New
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Diane Cassidy
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Amelia Shoemark
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Hollian Richardson
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Angus I Lamond
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Doreen A Cantrell
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
- Indicates joint senior authorship
| | - Alejandro J Brenes
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
- Indicates joint senior authorship
| |
Collapse
|
48
|
Horn CM, Arumugam P, Van Roy Z, Heim CE, Fallet RW, Bertrand BP, Shinde D, Thomas VC, Romanova SG, Bronich TK, Hartman CW, Garvin KL, Kielian T. Granulocytic myeloid-derived suppressor cell activity during biofilm infection is regulated by a glycolysis/HIF1a axis. J Clin Invest 2024; 134:e174051. [PMID: 38421730 PMCID: PMC11014666 DOI: 10.1172/jci174051] [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: 07/18/2023] [Accepted: 02/20/2024] [Indexed: 03/02/2024] Open
Abstract
Staphylococcus aureus is a leading cause of biofilm-associated prosthetic joint infection (PJI). A primary contributor to infection chronicity is an expansion of granulocytic myeloid-derived suppressor cells (G-MDSCs), which are critical for orchestrating the antiinflammatory biofilm milieu. Single-cell sequencing and bioinformatic metabolic algorithms were used to explore the link between G-MDSC metabolism and S. aureus PJI outcome. Glycolysis and the hypoxia response through HIF1a were significantly enriched in G-MDSCs. Interfering with both pathways in vivo, using a 2-deoxyglucose nanopreparation and granulocyte-targeted Hif1a conditional KO mice, respectively, attenuated G-MDSC-mediated immunosuppression and reduced bacterial burden in a mouse model of S. aureus PJI. In addition, single-cell RNA-Seq (scRNA-Seq) analysis of granulocytes from PJI patients also showed an enrichment in glycolysis and hypoxia-response genes. These findings support the importance of a glycolysis/HIF1a axis in promoting G-MDSC antiinflammatory activity and biofilm persistence during PJI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Svetlana G. Romanova
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Tatiana K. Bronich
- Department of Pharmacy, Northeastern University, Boston, Massachusetts, USA
| | - Curtis W. Hartman
- Department of Orthopaedic Surgery and Rehabilitation, UNMC, Omaha, Nebraska, USA
| | - Kevin L. Garvin
- Department of Orthopaedic Surgery and Rehabilitation, UNMC, Omaha, Nebraska, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology and
| |
Collapse
|
49
|
Aymonnier K, Bosetta E, Leborgne NGF, Ullmer A, Le Gall M, De Chiara A, Salnot V, Many S, Scapini P, Wicks I, Chatfield S, Martin KR, Witko-Sarsat V. G-CSF reshapes the cytosolic PCNA scaffold and modulates glycolysis in neutrophils. J Leukoc Biol 2024; 115:205-221. [PMID: 37824822 DOI: 10.1093/jleuko/qiad122] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/22/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023] Open
Abstract
Cytosolic proliferating cell nuclear antigen (PCNA) is involved in neutrophil survival and function, in which it acts as a scaffold and associates with proteins involved in apoptosis, NADPH oxidase activation, cytoskeletal dynamics, and metabolism. While the PCNA interactome has been characterized in neutrophils under homeostatic conditions, less is known about neutrophil PCNA in pathophysiological contexts. Granulocyte colony-stimulating factor (G-CSF) is a cytokine produced in response to inflammatory stimuli that regulates many aspects of neutrophil biology. Here, we used isolated normal-density neutrophils from G-CSF-treated haemopoietic stem cell donors (GDs) as a model to understand the role of PCNA during inflammation. Proteomic analysis of the neutrophil cytosol revealed significant differences between GDs and healthy donors (HDs). PCNA was one of the most upregulated proteins in GDs, and the PCNA interactome was significantly different in GDs compared with HDs. Importantly, while PCNA associated with almost all enzymes involved in glycolysis in HDs, these associations were decreased in GDs. Functionally, neutrophils from GDs had a significant increase in glycolysis compared with HDs. Using p21 competitor peptides, we showed that PCNA negatively regulates neutrophil glycolysis in HDs but had no effect on GD neutrophils. These data demonstrate that G-CSF alters the PCNA scaffold, affecting interactions with key glycolytic enzymes, and thus regulates glycolysis, the main energy pathway utilized by neutrophils. By this selective control of glycolysis, PCNA can organize neutrophils functionality in parallel with other PCNA mechanisms of prolonged survival. PCNA may therefore be instrumental in the reprogramming that neutrophils undergo in inflammatory or tumoral settings.
Collapse
Affiliation(s)
- Karen Aymonnier
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Enzo Bosetta
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Nathan G F Leborgne
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Audrey Ullmer
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Morgane Le Gall
- Proteom'IC facility, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du Faubourg Saint Jacques, Paris F-75014, France
| | - Alessia De Chiara
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Virginie Salnot
- Proteom'IC facility, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du Faubourg Saint Jacques, Paris F-75014, France
| | - Souganya Many
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Patrizia Scapini
- Department of General Pathology, University of Verona, Verona 37134, Italy
| | - Ian Wicks
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Grattan Street, Parkville, Victoria 3010, Australia
- Department of Rheumatology, Royal Melbourne Hospital, Grattan Street, Parkville, Victoria 3050, Australia
| | - Simon Chatfield
- Department of Rheumatology, Royal Melbourne Hospital, Grattan Street, Parkville, Victoria 3050, Australia
| | - Katherine R Martin
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Grattan Street, Parkville, Victoria 3010, Australia
| | - Véronique Witko-Sarsat
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| |
Collapse
|
50
|
Hogg M, Wolfschmitt EM, Wachter U, Zink F, Radermacher P, Vogt JA. Ex Vivo 13C-Metabolic Flux Analysis of Porcine Circulating Immune Cells Reveals Cell Type-Specific Metabolic Patterns and Sex Differences in the Pentose Phosphate Pathway. Biomolecules 2024; 14:98. [PMID: 38254698 PMCID: PMC10813356 DOI: 10.3390/biom14010098] [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: 11/10/2023] [Revised: 12/08/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
In general, females present with stronger immune responses than males, but scarce data are available on sex-specific differences in immunometabolism. In this study, we characterized porcine peripheral blood mononuclear cell (PBMC) and granulocyte energy metabolism using a Bayesian 13C-metabolic flux analysis, which allowed precise determination of the glycolytic, pentose phosphate pathway (PPP), and tricarboxylic acid cycle (TCA) fluxes, together with an assessment of the superoxide anion radical (O2•-) production and mitochondrial O2 consumption. A principal component analysis allowed for identifying the cell type-specific patterns of metabolic plasticity. PBMCs displayed higher TCA cycle activity, especially glutamine-derived aspartate biosynthesis, which was directly related to mitochondrial respiratory activity and inversely related to O2•- production. In contrast, the granulocytes mainly utilized glucose via glycolysis, which was coupled to oxidative PPP utilization and O2•- production rates. The granulocytes of the males had higher oxidative PPP fluxes compared to the females, while the PBMCs of the females displayed higher non-oxidative PPP fluxes compared to the males associated with the T helper cell (CD3+CD4+) subpopulation of PBMCs. The observed sex-specific differences were not directly attributable to sex steroid plasma levels, but we detected an inverse correlation between testosterone and aldosterone plasma levels and showed that aldosterone levels were related with non-oxidative PPP fluxes of both cell types.
Collapse
Affiliation(s)
- Melanie Hogg
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (E.-M.W.); (U.W.); (F.Z.); (P.R.); (J.A.V.)
| | | | | | | | | | | |
Collapse
|