1
|
Yap JQ, Nikouee A, Lau JE, Walsh G, Zang QS. Mitochondria at the Heart of Sepsis: Mechanisms, Metabolism, and Sex Differences. Int J Mol Sci 2025; 26:4211. [PMID: 40362448 PMCID: PMC12071423 DOI: 10.3390/ijms26094211] [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: 02/20/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Sepsis is a life-threatening condition that occurs when the body is unable to effectively combat infection, leading to systemic inflammation and multi-organ failure. Interestingly, females exhibit lower sepsis incidence and improved clinical outcomes compared to males. However, the mechanisms underlying these sex-specific differences remain poorly understood. While sex hormones have been a primary focus, emerging evidence suggests that non-hormonal factors also play contributory roles. Despite sex differences in sepsis, clinical management is the same for both males and females, with treatment focused on combating infection using antibiotics and hemodynamic support through fluid therapy. However, even with these interventions, mortality remains high, highlighting the need for more effective and targeted therapeutic strategies. Sepsis-induced cardiomyopathy (SIC) is a key contributor to multi-organ failure and is characterized by left ventricular dilation and impaired cardiac contractility. In this review, we explore sex-specific differences in sepsis and SIC, with a particular focus on mitochondrial metabolism. Mitochondria generate the ATP required for cardiac function through fatty acid and glucose oxidation, and recent studies have revealed distinct metabolic profiles between males and females, which can further differ in the context of sepsis and SIC. Targeting these metabolic pathways could provide new avenues for sepsis treatment.
Collapse
Affiliation(s)
- John Q. Yap
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Azadeh Nikouee
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Jessie E. Lau
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Gabriella Walsh
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Qun Sophia Zang
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
- Cardiovascular Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| |
Collapse
|
2
|
Sánchez‐García S, Povo‐Retana A, Marin S, Madurga S, Fariñas M, Aleixandre N, Castrillo A, de la Rosa JV, Alvarez‐Lucena C, Landauro‐Vera R, Prieto P, Cascante M, Boscá L. Immunometabolic Effect of Nitric Oxide on Human Macrophages Challenged With the SARS-CoV2-Induced Cytokine Storm. A Fluxomic Approach. Adv Healthc Mater 2025; 14:e2401688. [PMID: 39502019 PMCID: PMC11694080 DOI: 10.1002/adhm.202401688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 10/04/2024] [Indexed: 01/03/2025]
Abstract
The cytokine storm associated with SARS-CoV-2 infection is one of the most distinctive pathological signatures in COVID-19 patients. Macrophages respond to this pro-inflammatory challenge by reprogramming their functional and metabolic phenotypes. Interestingly, human macrophages fail to express the inducible form of the NO synthase (NOS2) in response to pro-inflammatory activation and, therefore, NO is not synthesized by these cells. The contribution of exogenously added NO, via a chemical NO-donor, on the immunometabolic changes associated with the cytokine storm is investigated. By using metabolic, transcriptomic, and functional assays the effect of NO in human macrophages is evaluated and found specific responses. Moreover, through integrative fluxomic analysis, pathways modified by NO that contribute to the expression of a particular phenotype in human macrophages are identified, which includes a decrease in mitochondrial respiration and TCA with a slight increase in the glycolytic flux. A significant ROS increase and preserved cell viability are observed in the presence of NO, which may ease the inflammatory response and host defense. Also, NO reverses the cytokine storm-induced itaconate accumulation. These changes offer additional clues to understanding the potential crosstalk between NO and the COVID-19 cytokine storm-dependent signaling pathways.
Collapse
Affiliation(s)
- Sergio Sánchez‐García
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Adrián Povo‐Retana
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Sergio Madurga
- Department of Material Science and Physical Chemistry & Research Institute of Theoretical and Computational Chemistry (IQTCUB)University of BarcelonaBarcelona08028Spain
| | - Marco Fariñas
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
| | - Nuria Aleixandre
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
- Department of Material Science and Physical Chemistry & Research Institute of Theoretical and Computational Chemistry (IQTCUB)University of BarcelonaBarcelona08028Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| | - Juan V. de la Rosa
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| | - Carlota Alvarez‐Lucena
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Rodrigo Landauro‐Vera
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Patricia Prieto
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
- Departamento de Farmacología, Farmacognosia y BotánicaFacultad de Farmacia, Universidad Complutense de MadridMadrid28040Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| |
Collapse
|
3
|
Byun MJ, Armon R, Souza TFG, Anderson HD, Saleem A, Pauls SD. Omega-3 polyunsaturated fatty acids modify glucose metabolism in THP-1 monocytes. Biochem Cell Biol 2025; 103:1-10. [PMID: 39899814 DOI: 10.1139/bcb-2024-0202] [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] [Indexed: 02/05/2025] Open
Abstract
Chronic inflammation is a driving factor in diseases like obesity and type 2 diabetes. Enhanced cellular glucose metabolism may contribute to heightened immune activation. A human supplementation trial showed that the n-3 PUFA α-linolenic acid (ALA) reduced oxidative phosphorylation in monocytes. Our objective here is to assess the direct effects of ALA and docosahexaenoic acid (DHA) on glucose metabolism in a cell culture model and to explore possible molecular mechanisms. THP-1 monocytes were treated with 10-40 µmol/L of ALA or DHA and compared with vehicle and oleic acid controls. The Seahorse XFe24 and Oroboros O2k Oxygraph systems were used to approximate catabolic rates in the presence of glucose. Both ALA and DHA reduced oxidative phosphorylation. We identified pyruvate dehydrogenase kinase 4 (PDK4) as a possible mechanistic candidate explaining the effect of DHA. Additionally, both n-3 PUFAs reduced lipopolysaccharides-induced IL-1β production, while only DHA increased reactive oxygen species to a small but significant extent. Our data suggest that ALA and DHA trigger a re-wiring of bioenergetic pathways in monocytes, possibly via the upregulation of PDK4. Given the close relationship between cell metabolism and immune cell activation, this may represent a novel mechanism by which n-3 fatty acids modulate immune function and inflammation.
Collapse
Affiliation(s)
- Michael J Byun
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | - Roni Armon
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Tamiris F G Souza
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Hope D Anderson
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Ayesha Saleem
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Samantha D Pauls
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
4
|
Liu J, Zhou G, Tong Z, Wang X, Liu D. The Values of PDK1 and LDH Levels in Patients with Sepsis and Septic Shock: A Prospective Observational Study. J Inflamm Res 2024; 17:6815-6826. [PMID: 39372591 PMCID: PMC11451472 DOI: 10.2147/jir.s477495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Background Metabolic changing is the significant host stress response during sepsis, but there is increasing evidence that uncontrolled metabolic reprogramming is a contributing factor to sepsis. Nevertheless, its association with outcome in patients with sepsis has been poorly investigated. As the key enzyme of metabolic reprogramming, the clinical value of PDK1 and LDH in patients with sepsis will be investigated in this study. Methods We collected serum from 167 ICU patients within 24 hours of admission for a single-center prospective observational study. The levels of PDK1 and LDH were detected by enzyme-linked adsorption method. Pearson or Spearman coefficient for correlation analysis between PDK1, LDH and clinical indicators. Areas under the ROC curves for evaluation of mortality prediction. Kaplan-Meier survival curve analysis was performed, and Cox proportional hazards model was performed to determine the risk factors for 28-day mortality. Results The PDK1/LDH in the septic shock group was statistically different between both the sepsis group and ICU control group, and had good correlation with ScvO2 and lactate. In predicting 28-day mortality in patients with sepsis, the best AUC was observed for PDK1/LDH, and was higher than the AUC for PDK1, lactate, and SOFA. Additionally, patients with lower PDK1/LDH had markerablely higher 28-day mortality. The multivariate Cox proportional hazards model revealed that PDK1/LDH < 0.1808 were the independent risk factors for 28-day mortality in sepsis. Conclusion The level of PDK1/LDH at admission was markedly decreased in patients with septic shock, which can serve as a novel independent prognostic biomarker for predicting mortality.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Gaosheng Zhou
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Zewen Tong
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Xiaoting Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Dawei Liu
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
5
|
Liang G, Hu JY, Liu RJ, Chao YP, Hu YF, Zheng H, Pan XY, Li YJ, Gong YH, Lin C, Lin JH, Wang JD, Li TX, Pan JP, Guo DY. α-Ketoglutarate plays an inflammatory inhibitory role by regulating scavenger receptor class a expression through N6-methyladenine methylation during sepsis. Eur J Immunol 2024; 54:e2350655. [PMID: 38973083 DOI: 10.1002/eji.202350655] [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: 07/09/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/09/2024]
Abstract
Sepsis arises from an uncontrolled inflammatory response triggered by infection or stress, accompanied by alteration in cellular energy metabolism, and a strong correlation exists between these factors. Alpha-ketoglutarate (α-KG), an intermediate product of the TCA cycle, has the potential to modulate the inflammatory response and is considered a crucial link between energy metabolism and inflammation. The scavenger receptor (SR-A5), a significant pattern recognition receptor, assumes a vital function in anti-inflammatory reactions. In the current investigation, we have successfully illustrated the ability of α-KG to mitigate inflammatory factors in the serum of septic mice and ameliorate tissue damage. Additionally, α-KG has been shown to modulate metabolic reprogramming and macrophage polarization. Moreover, our findings indicate that the regulatory influence of α-KG on sepsis is mediated through SR-A5. We also elucidated the mechanism by which α-KG regulates SR-A5 expression and found that α-KG reduced the N6-methyladenosine level of macrophages by up-regulating the m6A demethylase ALKBH5. α-KG plays a crucial role in inhibiting inflammation by regulating SR-A5 expression through m6A demethylation during sepsis. The outcomes of this research provide valuable insights into the relationship between energy metabolism and inflammation regulation, as well as the underlying molecular regulatory mechanism.
Collapse
Affiliation(s)
- Gang Liang
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
- Zhejiang University school of medicine, Hangzhou, P. R. China
| | - Jia-Yan Hu
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Rou-Jun Liu
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Yu-Peng Chao
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Yi-Fan Hu
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Hong Zheng
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Xin-Yu Pan
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Yuan-Jing Li
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Yang-Hui Gong
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Chi Lin
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Jia-Hao Lin
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Jia-Dong Wang
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Tong-Xin Li
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
| | - Jian-Ping Pan
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
- Institute of Translational Medicine, Hangzhou City University, Hangzhou, P.R. China
| | - Dong-Yang Guo
- Department of Clinical Medicine, Hangzhou City University School of Medicine, Hangzhou, P.R. China
- Institute of Translational Medicine, Hangzhou City University, Hangzhou, P.R. China
| |
Collapse
|
6
|
Pedersen BH, Simões FB, Pogrebnyakov I, Welch M, Johansen HK, Molin S, La Rosa R. Metabolic specialization drives reduced pathogenicity in Pseudomonas aeruginosa isolates from cystic fibrosis patients. PLoS Biol 2024; 22:e3002781. [PMID: 39178315 PMCID: PMC11376529 DOI: 10.1371/journal.pbio.3002781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 09/05/2024] [Accepted: 08/01/2024] [Indexed: 08/25/2024] Open
Abstract
Metabolism provides the foundation for all cellular functions. During persistent infections, in adapted pathogenic bacteria metabolism functions radically differently compared with more naïve strains. Whether this is simply a necessary accommodation to the persistence phenotype or if metabolism plays a direct role in achieving persistence in the host is still unclear. Here, we characterize a convergent shift in metabolic function(s) linked with the persistence phenotype during Pseudomonas aeruginosa colonization in the airways of people with cystic fibrosis. We show that clinically relevant mutations in the key metabolic enzyme, pyruvate dehydrogenase, lead to a host-specialized metabolism together with a lower virulence and immune response recruitment. These changes in infection phenotype are mediated by impaired type III secretion system activity and by secretion of the antioxidant metabolite, pyruvate, respectively. Our results show how metabolic adaptations directly impinge on persistence and pathogenicity in this organism.
Collapse
Affiliation(s)
- Bjarke Haldrup Pedersen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Filipa Bica Simões
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| | - Ivan Pogrebnyakov
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Helle Krogh Johansen
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Molin
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ruggero La Rosa
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
7
|
Wu YL, Zhu AQ, Zhou XT, Zhang KW, Yuan XJ, Yuan M, He J, Pineda MA, Li KP. A Novel Ultrafiltrate Extract of Propolis Exerts Anti-inflammatory Activity through Metabolic Rewiring. Chem Biodivers 2024; 21:e202301315. [PMID: 38189169 DOI: 10.1002/cbdv.202301315] [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: 08/29/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 01/09/2024]
Abstract
Thousands of years ago, humans started to use propolis because of its medicinal properties, and modern science has successfully identified several bioactive molecules within this resinous bee product. However, a natural propolis extract which has been removed the adhesive glue and preserved propolis bioactive compounds is urgently needed to maximise the therapeutic opportunities. In this study, a novel ultrafiltrate fraction from Brazilian green propolis, termed P30K, was demonstrated with anti-inflammatory properties, both in vitro and in vivo. Total flavonoids and total phenolic acids content in P30K were 244.6 mg/g and 275.8 mg/g respectively, while the IC50 value of inhibition of cyclooxygenase-2 (COX-2) was 8.30 μg/mL. The anti-inflammatory activity of P30K was furtherly corroborated in experimental models of lipopolysaccharides (LPS)-induced acute liver and lung injury. Mechanistically, integrated GC-MS and LC-MS based serum metabolomics analysis revealed that P30K modulated citrate cycle (TCA), pyruvate, glyoxylate and dicarboxylate metabolism pathways to inhibit secretion of pro-inflammatory cytokines. Results of network pharmacology and molecular docking suggested that P30K targeted catechol-O-methyltransferases (COMT), 11β-hydroxysteroid dehydrogenases (HSD11B1), and monoamine oxidases (MAOA and MAOB) to promote cellular metabolomic rewiring. Collectively, our work reveals P30K as an efficient therapeutic agent against inflammatory conditions and its efficacy is related to metabolic rewiring.
Collapse
Affiliation(s)
- Yong-Lin Wu
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, 280 East Road, Outer Ring, Guangzhou Higher Education Mega Center, Guangzhou, China, 510006
| | - An-Qi Zhu
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, 280 East Road, Outer Ring, Guangzhou Higher Education Mega Center, Guangzhou, China, 510006
| | - Xiao-Ting Zhou
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, 280 East Road, Outer Ring, Guangzhou Higher Education Mega Center, Guangzhou, China, 510006
| | - Ke-Wei Zhang
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, 280 East Road, Outer Ring, Guangzhou Higher Education Mega Center, Guangzhou, China, 510006
| | - Xu-Jiang Yuan
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Min Yuan
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, 280 East Road, Outer Ring, Guangzhou Higher Education Mega Center, Guangzhou, China, 510006
| | - Jian He
- BYHEALTH Institute of Nutrition & Health., Guangzhou, 510000, China
| | - Miguel A Pineda
- Centre for the Cellular Microenvironment, University of Glasgow, University Place, Glasgow, G12 8TA, UK
| | - Kun-Ping Li
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, 280 East Road, Outer Ring, Guangzhou Higher Education Mega Center, Guangzhou, China, 510006
| |
Collapse
|
8
|
Plokhikh KS, Nesterov SV, Chesnokov YM, Rogov AG, Kamyshinsky RA, Vasiliev AL, Yaguzhinsky LS, Vasilov RG. Association of 2-oxoacid dehydrogenase complexes with respirasomes in mitochondria. FEBS J 2024; 291:132-141. [PMID: 37789611 DOI: 10.1111/febs.16965] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/14/2023] [Accepted: 09/29/2023] [Indexed: 10/05/2023]
Abstract
In the present study, cryo-electron tomography was used to investigate the localization of 2-oxoacid dehydrogenase complexes (OADCs) in cardiac mitochondria and mitochondrial inner membrane samples. Two classes of ordered OADC inner cores with different symmetries were distinguished and their quaternary structures modeled. One class corresponds to pyruvate dehydrogenase complexes and the other to dehydrogenase complexes of α-ketoglutarate and branched-chain α-ketoacids. OADCs were shown to be localized in close proximity to membrane-embedded respirasomes, as observed both in densely packed lamellar cristae of cardiac mitochondria and in ruptured mitochondrial samples where the dense packing is absent. This suggests the specificity of the OADC-respirasome interaction, which allows localized NADH/NAD+ exchange between OADCs and complex I of the respiratory chain. The importance of this local coupling is based on OADCs being the link between respiration, glycolysis and amino acid metabolism. The coupling of these basic metabolic processes can vary in different tissues and conditions and may be involved in the development of various pathologies. The present study shows that this important and previously missing parameter of mitochondrial complex coupling can be successfully assessed using cryo-electron tomography.
Collapse
Affiliation(s)
- Konstantin S Plokhikh
- Kurchatov Complex of NBICS-Technologies, National Research Center Kurchatov Institute, Moscow, Russia
- Shubnikov Institute of Crystallography, Federal Scientific Research Centre "Crystallography and Photonics" of Russian Academy of Sciences, Moscow, Russia
| | - Semen V Nesterov
- Kurchatov Complex of NBICS-Technologies, National Research Center Kurchatov Institute, Moscow, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Yuriy M Chesnokov
- Kurchatov Complex of NBICS-Technologies, National Research Center Kurchatov Institute, Moscow, Russia
- Shubnikov Institute of Crystallography, Federal Scientific Research Centre "Crystallography and Photonics" of Russian Academy of Sciences, Moscow, Russia
| | - Anton G Rogov
- Kurchatov Complex of NBICS-Technologies, National Research Center Kurchatov Institute, Moscow, Russia
| | - Roman A Kamyshinsky
- Kurchatov Complex of NBICS-Technologies, National Research Center Kurchatov Institute, Moscow, Russia
- Shubnikov Institute of Crystallography, Federal Scientific Research Centre "Crystallography and Photonics" of Russian Academy of Sciences, Moscow, Russia
| | - Aleksandr L Vasiliev
- Kurchatov Complex of NBICS-Technologies, National Research Center Kurchatov Institute, Moscow, Russia
- Shubnikov Institute of Crystallography, Federal Scientific Research Centre "Crystallography and Photonics" of Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Lev S Yaguzhinsky
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Belozersky Research Institute for Physico-Chemical Biology, Lomonosov Moscow State University, Russia
| | - Raif G Vasilov
- Kurchatov Complex of NBICS-Technologies, National Research Center Kurchatov Institute, Moscow, Russia
| |
Collapse
|
9
|
Li Q, Liu T, Bai C, Ma X, Liu H, Zheng Z, Wan Y, Yu H, Ma Y, Gu X. iTRAQ-based proteomics reveals the mechanism of action of Yinlai decoction in treating pneumonia in mice consuming a high-calorie diet. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2024; 11:21-32. [DOI: 10.1016/j.jtcms.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2025] Open
|
10
|
Li C, Liu C, Zhang J, Lu Y, Jiang B, Xiong H, Li C. Pyruvate dehydrogenase kinase regulates macrophage polarization in metabolic and inflammatory diseases. Front Immunol 2023; 14:1296687. [PMID: 38193078 PMCID: PMC10773690 DOI: 10.3389/fimmu.2023.1296687] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/13/2023] [Indexed: 01/10/2024] Open
Abstract
Macrophages are highly heterogeneous and plastic, and have two main polarized phenotypes that are determined by their microenvironment, namely pro- and anti-inflammatory macrophages. Activation of pro-inflammatory macrophages is closely associated with metabolic reprogramming, especially that of aerobic glycolysis. Mitochondrial pyruvate dehydrogenase kinase (PDK) negatively regulates pyruvate dehydrogenase complex activity through reversible phosphorylation and further links glycolysis to the tricarboxylic acid cycle and ATP production. PDK is commonly associated with the metabolism and polarization of macrophages in metabolic and inflammatory diseases. This review examines the relationship between PDK and macrophage metabolism and discusses the mechanisms by which PDK regulates macrophage polarization, migration, and inflammatory cytokine secretion in metabolic and inflammatory diseases. Elucidating the relationships between the metabolism and polarization of macrophages under physiological and pathological conditions, as well as the regulatory pathways involved, may provide valuable insights into the etiology and treatment of macrophage-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Chenyu Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Chuanbin Liu
- Department of Pediatric Dentistry, Jining Stomatological Hospital, Jining, Shandong, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Yanyu Lu
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Bingtong Jiang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Chunxia Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
11
|
Yang W, Wang Y, Tao K, Li R. Metabolite itaconate in host immunoregulation and defense. Cell Mol Biol Lett 2023; 28:100. [PMID: 38042791 PMCID: PMC10693715 DOI: 10.1186/s11658-023-00503-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/20/2023] [Indexed: 12/04/2023] Open
Abstract
Metabolic states greatly influence functioning and differentiation of immune cells. Regulating the metabolism of immune cells can effectively modulate the host immune response. Itaconate, an intermediate metabolite derived from the tricarboxylic acid (TCA) cycle of immune cells, is produced through the decarboxylation of cis-aconitate by cis-aconitate decarboxylase in the mitochondria. The gene encoding cis-aconitate decarboxylase is known as immune response gene 1 (IRG1). In response to external proinflammatory stimulation, macrophages exhibit high IRG1 expression. IRG1/itaconate inhibits succinate dehydrogenase activity, thus influencing the metabolic status of macrophages. Therefore, itaconate serves as a link between macrophage metabolism, oxidative stress, and immune response, ultimately regulating macrophage function. Studies have demonstrated that itaconate acts on various signaling pathways, including Keap1-nuclear factor E2-related factor 2-ARE pathways, ATF3-IκBζ axis, and the stimulator of interferon genes (STING) pathway to exert antiinflammatory and antioxidant effects. Furthermore, several studies have reported that itaconate affects cancer occurrence and development through diverse signaling pathways. In this paper, we provide a comprehensive review of the role IRG1/itaconate and its derivatives in the regulation of macrophage metabolism and functions. By furthering our understanding of itaconate, we intend to shed light on its potential for treating inflammatory diseases and offer new insights in this field.
Collapse
Affiliation(s)
- Wenchang Yang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaxin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Ruidong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China.
| |
Collapse
|
12
|
Stakišaitis D, Kapočius L, Kilimaitė E, Gečys D, Šlekienė L, Balnytė I, Palubinskienė J, Lesauskaitė V. Preclinical Study in Mouse Thymus and Thymocytes: Effects of Treatment with a Combination of Sodium Dichloroacetate and Sodium Valproate on Infectious Inflammation Pathways. Pharmaceutics 2023; 15:2715. [PMID: 38140056 PMCID: PMC10747708 DOI: 10.3390/pharmaceutics15122715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 11/17/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
The research presents data from a preclinical study on the anti-inflammatory effects of a sodium dichloroacetate and sodium valproate combination (DCA-VPA). The 2-week treatment with a DCA 100 mg/kg/day and VPA 150 mg/kg/day combination solution in drinking water's effects on the thymus weight, its cortex/medulla ratio, Hassall's corpuscles (HCs) number in the thymus medulla, and the expression of inflammatory and immune-response-related genes in thymocytes of male Balb/c mice were studied. Two groups of mice aged 6-7 weeks were investigated: a control (n = 12) and a DCA-VPA-treated group (n = 12). The treatment did not affect the body weight gain (p > 0.05), the thymus weight (p > 0.05), the cortical/medulla ratio (p > 0.05), or the number of HCs (p > 0.05). Treatment significantly increased the Slc5a8 gene expression by 2.1-fold (p < 0.05). Gene sequence analysis revealed a significant effect on the expression of inflammation-related genes in thymocytes by significantly altering the expression of several genes related to the cytokine activity pathway, the inflammatory response pathway, and the Il17 signaling pathway in thymocytes. Data suggest that DCA-VPA exerts an anti-inflammatory effect by inhibiting the inflammatory mechanisms in the mouse thymocytes.
Collapse
Affiliation(s)
- Donatas Stakišaitis
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
- Laboratory of Molecular Oncology, National Cancer Institute, 08660 Vilnius, Lithuania
| | - Linas Kapočius
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
| | - Evelina Kilimaitė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
| | - Dovydas Gečys
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu Ave., 50161 Kaunas, Lithuania;
| | - Lina Šlekienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
| | - Jolita Palubinskienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
| | - Vaiva Lesauskaitė
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu Ave., 50161 Kaunas, Lithuania;
| |
Collapse
|
13
|
Long DL, McCall CE, Poole LB. Glutathionylation of pyruvate dehydrogenase complex E2 and inflammatory cytokine production during acute inflammation are magnified by mitochondrial oxidative stress. Redox Biol 2023; 65:102841. [PMID: 37566945 PMCID: PMC10440583 DOI: 10.1016/j.redox.2023.102841] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
Lipopolysaccharide (LPS) is a known inducer of inflammatory signaling which triggers generation of reactive oxygen species (ROS) and cell death in responsive cells like THP-1 promonocytes and freshly isolated human monocytes. A key LPS-responsive metabolic pivot point is the 9 MDa mitochondrial pyruvate dehydrogenase complex (PDC), which provides pyruvate dehydrogenase (E1), lipoamide-linked transacetylase (E2) and lipoamide dehydrogenase (E3) activities to produce acetyl-CoA from pyruvate. While phosphorylation-dependent decreases in PDC activity following LPS treatment or sepsis have been deeply investigated, redox-linked processes have received less attention. Data presented here demonstrate that LPS-induced reversible oxidation within PDC occurs in PDCE2 in both THP-1 cells and primary human monocytes. Knockout of PDCE2 by CRISPR and expression of FLAG-tagged PDCE2 in THP-1 cells demonstrated that LPS-induced glutathionylation is associated with wild type PDCE2 but not mutant protein lacking the lipoamide-linking lysine residues. Moreover, the mitochondrially-targeted electrophile MitoCDNB, which impairs both glutathione- and thioredoxin-based reductase systems, elevates ROS similar to LPS but does not cause PDCE2 glutathionylation. However, LPS and MitoCDNB together are highly synergistic for PDCE2 glutathionylation, ROS production, and cell death. Surprisingly, the two treatments together had differential effects on cytokine production; pro-inflammatory IL-1β production was enhanced by the co-treatment, while IL-10, an important anti-inflammatory cytokine, dropped precipitously compared to LPS treatment alone. This new information may expand opportunities to understand and modulate PDC redox status and activity and improve the outcomes of pathological inflammation.
Collapse
Affiliation(s)
- David L Long
- Department of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| | - Charles E McCall
- Department of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| | - Leslie B Poole
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
14
|
Long DL, McCall CE, Poole LB. Glutathionylation of Pyruvate Dehydrogenase Complex E2 and Inflammatory Cytokine Production During Acute Inflammation Are Magnified By Mitochondrial Oxidative Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525791. [PMID: 36747682 PMCID: PMC9900926 DOI: 10.1101/2023.01.26.525791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Lipopolysaccharide (LPS) is a known inducer of inflammatory signaling which triggers generation of reactive oxygen species (ROS) and cell death in responsive cells like THP-1 promonocytes and freshly isolated human monocytes. A key LPS-responsive metabolic pivot point is the 9 megadalton mitochondrial pyruvate dehydrogenase complex (PDC), which provides pyruvate dehydrogenase (E1), lipoamide-linked transacetylase (E2) and lipoamide dehydrogenase (E3) activities to produce acetyl-CoA from pyruvate. While phosphorylation-dependent decreases in PDC activity following LPS treatment or sepsis have been deeply investigated, redox-linked processes have received less attention. Data presented here demonstrate that LPS-induced reversible oxidation within PDC occurs in PDCE2 in both THP-1 cells and primary human monocytes. Knockout of PDCE2 by CRISPR and expression of FLAG-tagged PDCE2 in THP-1 cells demonstrated that LPS-induced glutathionylation is associated with wild type PDCE2 but not mutant protein lacking the lipoamide-linking lysine residues. Moreover, the mitochondrially-targeted electrophile MitoCDNB, which impairs both glutathione- and thioredoxin-based reductase systems, elevates ROS similar to LPS but does not cause PDCE2 glutathionylation. However, LPS and MitoCDNB together are highly synergistic for PDCE2 glutathionylation, ROS production, and cell death. Surprisingly, the two treatments together had differential effects on cytokine production; pro-inflammatory IL-1β production was enhanced by the co-treatment, while IL-10, an important anti-inflammatory cytokine, dropped precipitously compared to LPS treatment alone. This new information may expand opportunities to understand and modulate PDC redox status and activity and improve the outcomes of pathological inflammation.
Collapse
Affiliation(s)
- David L. Long
- Department of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Charles E. McCall
- Department of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Leslie B. Poole
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| |
Collapse
|
15
|
Xu K, Saaoud F, Shao Y, Lu Y, Wu S, Zhao H, Chen K, Vazquez-Padron R, Jiang X, Wang H, Yang X. Early hyperlipidemia triggers metabolomic reprogramming with increased SAH, increased acetyl-CoA-cholesterol synthesis, and decreased glycolysis. Redox Biol 2023; 64:102771. [PMID: 37364513 PMCID: PMC10310484 DOI: 10.1016/j.redox.2023.102771] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
To identify metabolomic reprogramming in early hyperlipidemia, unbiased metabolome was screened in four tissues from ApoE-/- mice fed with high fat diet (HFD) for 3 weeks. 30, 122, 67, and 97 metabolites in the aorta, heart, liver, and plasma, respectively, were upregulated. 9 upregulated metabolites were uremic toxins, and 13 metabolites, including palmitate, promoted a trained immunity with increased syntheses of acetyl-CoA and cholesterol, increased S-adenosylhomocysteine (SAH) and hypomethylation and decreased glycolysis. The cross-omics analysis found upregulation of 11 metabolite synthetases in ApoE‾/‾ aorta, which promote ROS, cholesterol biosynthesis, and inflammation. Statistical correlation of 12 upregulated metabolites with 37 gene upregulations in ApoE‾/‾ aorta indicated 9 upregulated new metabolites to be proatherogenic. Antioxidant transcription factor NRF2-/- transcriptome analysis indicated that NRF2 suppresses trained immunity-metabolomic reprogramming. Our results have provided novel insights on metabolomic reprogramming in multiple tissues in early hyperlipidemia oriented toward three co-existed new types of trained immunity.
Collapse
Affiliation(s)
- Keman Xu
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Fatma Saaoud
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Ying Shao
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Yifan Lu
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Sheng Wu
- Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Huaqing Zhao
- Medical Education and Data Science, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Kaifu Chen
- Computational Biology Program, Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Roberto Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33125, USA
| | - Xiaohua Jiang
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
16
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
17
|
Meyers AK, Wang Z, Han W, Zhao Q, Zabalawi M, Duan L, Liu J, Zhang Q, Manne RK, Lorenzo F, Quinn MA, Song Q, Fan D, Lin HK, Furdui CM, Locasale JW, McCall CE, Zhu X. Pyruvate dehydrogenase kinase supports macrophage NLRP3 inflammasome activation during acute inflammation. Cell Rep 2023; 42:111941. [PMID: 36640341 PMCID: PMC10117036 DOI: 10.1016/j.celrep.2022.111941] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 08/02/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Activating the macrophage NLRP3 inflammasome can promote excessive inflammation with severe cell and tissue damage and organ dysfunction. Here, we show that pharmacological or genetic inhibition of pyruvate dehydrogenase kinase (PDHK) significantly attenuates NLRP3 inflammasome activation in murine and human macrophages and septic mice by lowering caspase-1 cleavage and interleukin-1β (IL-1β) secretion. Inhibiting PDHK reverses NLRP3 inflammasome-induced metabolic reprogramming, enhances autophagy, promotes mitochondrial fusion over fission, preserves crista ultrastructure, and attenuates mitochondrial reactive oxygen species (ROS) production. The suppressive effect of PDHK inhibition on the NLRP3 inflammasome is independent of its canonical role as a pyruvate dehydrogenase regulator. Our study suggestsa non-canonical role of mitochondrial PDHK in promoting mitochondrial stress and supporting NLRP3 inflammasome activation during acute inflammation.
Collapse
Affiliation(s)
- Allison K Meyers
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Zhan Wang
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Wenzheng Han
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Qingxia Zhao
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Manal Zabalawi
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Likun Duan
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Juan Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Qianyi Zhang
- Department of Biology, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Rajesh K Manne
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Felipe Lorenzo
- Section on Endocrinology and Metabolism, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Matthew A Quinn
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Cristina M Furdui
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Charles E McCall
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Xuewei Zhu
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
18
|
McCall CE, Zhu X, Zabalawi M, Long D, Quinn MA, Yoza BK, Stacpoole PW, Vachharajani V. Sepsis, pyruvate, and mitochondria energy supply chain shortage. J Leukoc Biol 2022; 112:1509-1514. [PMID: 35866365 PMCID: PMC9796618 DOI: 10.1002/jlb.3mr0322-692rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 01/04/2023] Open
Abstract
Balancing high energy-consuming danger resistance and low energy supply of disease tolerance is a universal survival principle that often fails during sepsis. Our research supports the concept that sepsis phosphorylates and deactivates mitochondrial pyruvate dehydrogenase complex control over the tricarboxylic cycle and the electron transport chain. StimulatIng mitochondrial energetics in septic mice and human sepsis cell models can be achieved by inhibiting pyruvate dehydrogenase kinases with the pyruvate structural analog dichloroacetate. Stimulating the pyruvate dehydrogenase complex by dichloroacetate reverses a disruption in the tricarboxylic cycle that induces itaconate, a key mediator of the disease tolerance pathway. Dichloroacetate treatment increases mitochondrial respiration and ATP synthesis, decreases oxidant stress, overcomes metabolic paralysis, regenerates tissue, organ, and innate and adaptive immune cells, and doubles the survival rate in a murine model of sepsis.
Collapse
Affiliation(s)
- Charles E. McCall
- Department of MedicineWake Forest School of MedicineWinston SalemNCUSA
| | - Xuewei Zhu
- Department of MedicineWake Forest School of MedicineWinston SalemNCUSA
| | - Manal Zabalawi
- Department of MedicineWake Forest School of MedicineWinston SalemNCUSA
| | - David Long
- Department of MedicineWake Forest School of MedicineWinston SalemNCUSA
| | - Matthew A. Quinn
- Department of Pathology – Comparative MedicineWake Forest School of MedicineWinston SalemNCUSA
| | - Barbara K. Yoza
- Department of SurgeryWake Forest School of MedicineWinston SalemNCUSA
| | - Peter W. Stacpoole
- Department of Medicine and BiochemistryUniversity of Florida Medical SchoolGainesvilleFloridaUSA
| | - Vidula Vachharajani
- Department of Critical Care MedicineCleveland Clinic Lerner College of Medicine of CWRUClevelandOhioUSA
| |
Collapse
|
19
|
Oh TS, Zabalawi M, Jain S, Long D, Stacpoole PW, McCall CE, Quinn MA. Dichloroacetate improves systemic energy balance and feeding behavior during sepsis. JCI Insight 2022; 7:153944. [PMID: 35730570 PMCID: PMC9309051 DOI: 10.1172/jci.insight.153944] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by dysregulated host response to an infection. The metabolic aberrations associated with sepsis underly an acute and organism-wide hyperinflammatory response and multiple organ dysfunction; however, crosstalk between systemic metabolomic alterations and metabolic reprogramming at organ levels remains unknown. We analyzed substrate utilization by the respiratory exchange ratio, energy expenditure, metabolomic screening, and transcriptional profiling in a cecal ligation and puncture model to show that sepsis increases circulating free fatty acids and acylcarnitines but decreases levels of amino acids and carbohydrates, leading to a drastic shift in systemic fuel preference. Comparative analysis of previously published metabolomics from septic liver indicated a positive correlation with hepatic and plasma metabolites during sepsis. In particular, glycine deficiency was a common abnormality of the plasma and liver during sepsis. Interrogation of the hepatic transcriptome in septic mice suggested that the septic liver may contribute to systemic glycine deficiency by downregulating genes involved in glycine synthesis. Interestingly, intraperitoneal injection of the pyruvate dehydrogenase kinase (PDK) inhibitor dichloroacetate reversed sepsis-induced anorexia, energy imbalance, inflammation, dyslipidemia, hypoglycemia, and glycine deficiency. Collectively, our data indicated that PDK inhibition rescued systemic energy imbalance and metabolic dysfunction in sepsis partly through restoration of hepatic fuel metabolism.
Collapse
Affiliation(s)
- Tae Seok Oh
- Department of Pathology, Section on Comparative Medicine, and
| | - Manal Zabalawi
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Shalini Jain
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - David Long
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Peter W. Stacpoole
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine and Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Charles E. McCall
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Matthew A. Quinn
- Department of Pathology, Section on Comparative Medicine, and,Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
20
|
The Pyruvate Dehydrogenase Complex Mitigates LPS-Induced Endothelial Barrier Dysfunction by Metabolic Regulation. Shock 2022; 57:308-317. [PMID: 35759309 DOI: 10.1097/shk.0000000000001931] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
ABSTRACT Sepsis is a fatal health issue induced by an aberrant host response to infection, and it correlates with organ damage and a high mortality rate. Endothelial barrier dysfunction and subsequent capillary leakage play major roles in sepsis-induced multiorgan dysfunction. Anaerobic glycolysis is the primary metabolic mode in sepsis and the pyruvate dehydrogenase complex (PDHC) serves as a critical hub in energy regulation. Therefore, it is important to understand the role of PDHC in metabolic regulation during the development of sepsis-induced endothelial barrier dysfunction.In present study, human umbilical vein endothelial cells (HUVECs) and C57 BL/6 mice were treated with lipopolysaccharide (LPS) as models of endotoxemia. LPS increased basal glycolysis, compensatory glycolysis, and lactate secretion, indicating increased glycolysis level in endothelial cells (ECs). Activation of PDHC with dichloroacetate (DCA) reversed LPS-induced glycolysis, allowing PDHC to remain in the active dephosphorylated state, thereby preventing lactic acid production and HUVECs monolayers barrier dysfunction, as assessed by transendothelial electrical resistance and Fluorescein Isothiocyanate-labeled dextran. The in vivo study also showed that the lactate level and vascular permeability were increased in LPS-treated mice, but pretreatment with DCA attenuated these increases. The LPS-treated HUVEC model showed that DCA reversed LPS-induced phosphorylation of pyruvate dehydrogenase E1α Ser293 and Ser300 to restore PDHC activity. Immunoprecipitation results showed that LPS treatment increased the acetylation level of PDH E1α in HUVECs.Our study suggested that activation of PDHC may represent a therapeutic target for treatment of LPS-induced endothelial barrier dysfunction.
Collapse
|
21
|
Zhang X, Jia Y, Yuan Z, Wen Y, Zhang Y, Ren J, Ji P, Yao W, Hua Y, Wei Y. Sheng Mai San ameliorated heat stress-induced liver injury via regulating energy metabolism and AMPK/Drp1-dependent autophagy process. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 97:153920. [PMID: 35026620 DOI: 10.1016/j.phymed.2021.153920] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/21/2021] [Accepted: 12/30/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Liver damage is one of the most common complications in humans and animals after heat stress (HS). Sheng Mai San (SMS), a traditional Chinese medicine prescription that originated in the Jin Dynasty, exert a therapeutic effect on HS. However, how SMS prevents liver injury after heat exposure remains unknown. PURPOSE This study aimed to investigate the pharmacological effect and molecular mechanisms of SMS on HS-induced liver injury. STUDY DESIGN A comprehensive strategy via incorporating pharmacodynamics, targeted metabolomics, and molecular biology technology was adopted to investigate energy metabolism changes and the therapeutic mechanisms of SMS in HS-induced rat liver injury. METHODS First, Sprague-Dawley rats were subjected to HS (38 °C/ 75% RH/ 2 h/ day) for 7 consecutive days to establish the HS model, and SMS was given orally for treatment 2 h before heat exposure. Thereafter, liver function and pathological changes in liver tissue were evaluated. Finally, the underlying mechanisms of SMS were determined using targeted energy metabolomics to comprehensively analyze the metabolic pathways and were further verified through Western-blot and qRT-PCR assays. RESULTS Our results showed that SMS alleviated HS-induced liver dysfunction by reducing the alanine aminotransferase (ALT), aspartate aminotransferase (AST), and AST/ALT ratios in serum and improving hepatic pathological damage. Meanwhile, SMS suppressed inflammatory response, oxidative injury, and overexpression of heat shock proteins in liver tissue after heat exposure. With the help of targeted energy metabolomics, we found that SMS could effectively regulate glycolysis and tricarboxylic acid (TCA) cycle to relieve energy metabolism disorder. Furthermore, we confirmed that SMS can facilitate the phosphorylation of AMP-activated protein kinase (AMPK) to maintain mitochondrial homeostasis through a dynamin protein 1 (Drp1)-dependent mitophagy process. CONCLUSION On the basis of energy metabolomics, the present study for the first time systematically illustrated the protective effect of SMS on HS-induced liver injury, and preliminarily confirmed that an AMPK-mediated Drp1-dependent mitophagy and mitochondria rebuilding process plays an important role in SMS intervention on HS-induced rat liver. Together, our study lends further support to the use of SMS in treating HS condition.
Collapse
Affiliation(s)
- Xiaosong Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Yaqian Jia
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Ziwen Yuan
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Yanqiao Wen
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Yahui Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Jianmin Ren
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Peng Ji
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Wanling Yao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Yongli Hua
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Yanming Wei
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| |
Collapse
|
22
|
Zeng Z, Huang Q, Mao L, Wu J, An S, Chen Z, Zhang W. The Pyruvate Dehydrogenase Complex in Sepsis: Metabolic Regulation and Targeted Therapy. Front Nutr 2022; 8:783164. [PMID: 34970577 PMCID: PMC8712327 DOI: 10.3389/fnut.2021.783164] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022] Open
Abstract
Anaerobic glycolysis is the process by which glucose is broken down into pyruvate and lactate and is the primary metabolic pathway in sepsis. The pyruvate dehydrogenase complex (PDHC) is a multienzyme complex that serves as a critical hub in energy metabolism. Under aerobic conditions, pyruvate translocates to mitochondria, where it is oxidized into acetyl-CoA through the activation of PDHC, thereby accelerating aerobic oxidation. Both phosphorylation and acetylation affect PDHC activity and, consequently, the regulation of energy metabolism. The mechanisms underlying the protective effects of PDHC in sepsis involve the regulation on the balance of lactate, the release of inflammatory mediators, the remodeling of tricarboxylic acid (TCA) cycle, as well as on the improvement of lipid and energy metabolism. Therapeutic drugs that target PDHC activation for sepsis treatment include dichloroacetate, thiamine, amrinone, TNF-binding protein, and ciprofloxacin. In this review, we summarize the recent findings regarding the metabolic regulation of PDHC in sepsis and the therapies targeting PDHC for the treatment of this condition.
Collapse
Affiliation(s)
- Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaobing Huang
- Department of Pathophysiology, Guangdong Provincial Key Lab of Shock and Microcirculation, Southern Medical University, Guangzhou, China
| | - Liangfeng Mao
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Wu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sheng An
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weijin Zhang
- Department of Internal Medicine General Ward, Shantou Central Hospital, Shantou, China
| |
Collapse
|
23
|
Zhao X, Li S, Mo Y, Li R, Huang S, Zhang A, Ni X, Dai Q, Wang J. DCA Protects against Oxidation Injury Attributed to Cerebral Ischemia-Reperfusion by Regulating Glycolysis through PDK2-PDH-Nrf2 Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5173035. [PMID: 34712383 PMCID: PMC8548159 DOI: 10.1155/2021/5173035] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/06/2021] [Accepted: 10/04/2021] [Indexed: 12/19/2022]
Abstract
Cerebral ischemic stroke (IS) is still a difficult problem to be solved; energy metabolism failure is one of the main factors causing mitochondrion dysfunction and oxidation stress damage within the pathogenesis of cerebral ischemia, which produces considerable reactive oxygen species (ROS) and opens the blood-brain barrier. Dichloroacetic acid (DCA) can inhibit pyruvate dehydrogenase kinase (PDK). Moreover, DCA has been indicated with the capability of increasing mitochondrial pyruvate uptake and promoting oxidation of glucose in the course of glycolysis, thereby improving the activity of pyruvate dehydrogenase (PDH). As a result, pyruvate flow is promoted into the tricarboxylic acid cycle to expedite ATP production. DCA has a protective effect on IS and brain ischemia/reperfusion (I/R) injury, but the specific mechanism remains unclear. This study adopted a transient middle cerebral artery occlusion (MCAO) mouse model for simulating IS and I/R injury in mice. We investigated the mechanism by which DCA regulates glycolysis and protects the oxidative damage induced by I/R injury through the PDK2-PDH-Nrf2 axis. As indicated from the results of this study, DCA may improve glycolysis, reduce oxidative stress and neuronal death, damage the blood-brain barrier, and promote the recovery of oxidative metabolism through inhibiting PDK2 and activating PDH. Additionally, DCA noticeably elevated the neurological score and reduced the infarct volume, brain water content, and necrotic neurons. Moreover, as suggested from the results, DCA elevated the content of Nrf2 as well as HO-1, i.e., the downstream antioxidant proteins pertaining to Nrf2, while decreasing the damage of BBB and the degradation of tight junction proteins. To simulate the condition of hypoxia and ischemia in vitro, HBMEC cells received exposure to transient oxygen and glucose deprivation (OGD). The DCA treatment is capable of reducing the oxidative stress and blood-brain barrier of HBMEC cells after in vitro hypoxia and reperfusion (H/R). Furthermore, this study evidenced that HBMEC cells could exhibit higher susceptibility to H/R-induced oxidative stress after ML385 application, the specific inhibitor of Nrf2. Besides, the protection mediated by DCA disappeared after ML385 application. To sum up, as revealed from the mentioned results, DCA could exert the neuroprotective effect on oxidative stress and blood-brain barrier after brain I/R injury via PDK2-PDH-Nrf2 pathway activation. Accordingly, the PDK2-PDH-Nrf2 pathway may play a key role and provide a new pharmacology target in cerebral IS and I/R protection by DCA.
Collapse
Affiliation(s)
- Xiaoyong Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang 261021, China
| | - Shan Li
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yunchang Mo
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Ruru Li
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Shaoyi Huang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Anqi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Xuqing Ni
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Qinxue Dai
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| |
Collapse
|
24
|
Persistent hyperammonia and altered concentrations of urea cycle metabolites in a 5-day swine experiment of sepsis. Sci Rep 2021; 11:18430. [PMID: 34531431 PMCID: PMC8445921 DOI: 10.1038/s41598-021-97855-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/24/2021] [Indexed: 01/20/2023] Open
Abstract
We measured plasma and cerebrospinal fluid (CSF) metabolite concentrations in a 5-day porcine sepsis model of fecal peritonitis. The objectives were: (i) to verify whether the expected pathways that had emerged in previous studies pertain only to the early inflammatory response or persist for the subsequent days; (ii) to identify metabolic derangements that arise later; (iii) to verify whether CSF metabolite concentrations were altered and if these alterations were similar to those in the blood or delayed. We observed an early response to inflammation and cytokine storms with alterations in lipid and glucose metabolism. The arginine/asymmetric dimethylarginine (ADMA) and phenylalanine/tyrosine balances changed 24 h after resuscitation in plasma, and later in CSF. There was a rise in ammonia concentration, with altered concentrations of metabolites in the urea cycle. Whether persistent derangement of these pathways have a role not only on short-term outcomes but also on longer-term comorbidities, such as septic encephalopathy, should be addressed in further studies.
Collapse
|
25
|
van Doorn CLR, Schouten GK, van Veen S, Walburg KV, Esselink JJ, Heemskerk MT, Vrieling F, Ottenhoff THM. Pyruvate Dehydrogenase Kinase Inhibitor Dichloroacetate Improves Host Control of Salmonella enterica Serovar Typhimurium Infection in Human Macrophages. Front Immunol 2021; 12:739938. [PMID: 34552598 PMCID: PMC8450447 DOI: 10.3389/fimmu.2021.739938] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/23/2021] [Indexed: 01/11/2023] Open
Abstract
Global increases in the prevalence of antimicrobial resistance highlight the urgent need for novel strategies to combat infectious diseases. Recent studies suggest that host metabolic pathways play a key role in host control of intracellular bacterial pathogens. In this study we explored the potential of targeting host metabolic pathways for innovative host-directed therapy (HDT) against intracellular bacterial infections. Through gene expression profiling in human macrophages, pyruvate metabolism was identified as potential key pathway involved in Salmonella enterica serovar Typhimurium (Stm) infections. Next, the effect of targeting pyruvate dehydrogenase kinases (PDKs) - which are regulators of the metabolic checkpoint pyruvate dehydrogenase complex (PDC) - on macrophage function and bacterial control was studied. Chemical inhibition of PDKs by dichloroacetate (DCA) induced PDC activation and was accompanied with metabolic rewiring in classically activated macrophages (M1) but not in alternatively activated macrophages (M2), suggesting cell-type specific effects of dichloroacetate on host metabolism. Furthermore, DCA treatment had minor impact on cytokine and chemokine secretion on top of infection, but induced significant ROS production by M1 and M2. DCA markedly and rapidly reduced intracellular survival of Stm, but interestingly not Mycobacterium tuberculosis, in human macrophages in a host-directed manner. In conclusion, DCA represents a promising novel HDT compound targeting pyruvate metabolism for the treatment of Stm infections.
Collapse
|
26
|
Kolliniati O, Ieronymaki E, Vergadi E, Tsatsanis C. Metabolic Regulation of Macrophage Activation. J Innate Immun 2021; 14:51-68. [PMID: 34247159 DOI: 10.1159/000516780] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/19/2021] [Indexed: 11/19/2022] Open
Abstract
Macrophages, the central mediators of innate immune responses, being in the first-line of defense, they have to readily respond to pathogenic or tissue damage signals to initiate the inflammatory cascade. Such rapid responses require energy to support orchestrated production of pro-inflammatory mediators and activation of phagocytosis. Being a cell type that is present in diverse environments and conditions, macrophages have to adapt to different nutritional resources. Thus, macrophages have developed plasticity and are capable of utilizing energy at both normoxic and hypoxic conditions and in the presence of varying concentrations of glucose or other nutrients. Such adaptation is reflected on changes in signaling pathways that modulate responses, accounting for the different activation phenotypes observed. Macrophage metabolism has been tightly associated with distinct activation phenotypes within the range of M1-like and M2-like types. In the context of diseases, systemic changes also affect macrophage metabolism, as in diabetes and insulin resistance, which results in altered metabolism and distinct activation phenotypes in the adipose tissue or in the periphery. In the context of solid tumors, tumor-associated macrophages adapt in the hypoxic environment, which results in metabolic changes that are reflected on an activation phenotype that supports tumor growth. Coordination of environmental and pathogenic signals determines macrophage metabolism, which in turn shapes the type and magnitude of the response. Therefore, modulating macrophage metabolism provides a potential therapeutic approach for inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Ourania Kolliniati
- Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece.,Department of Pediatrics, Medical School, University of Crete, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | - Eleftheria Ieronymaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | - Eleni Vergadi
- Department of Pediatrics, Medical School, University of Crete, Heraklion, Greece
| | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| |
Collapse
|
27
|
Lin J, Ren J, Gao DS, Dai Y, Yu L. The Emerging Application of Itaconate: Promising Molecular Targets and Therapeutic Opportunities. Front Chem 2021; 9:669308. [PMID: 34055739 PMCID: PMC8149739 DOI: 10.3389/fchem.2021.669308] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/01/2021] [Indexed: 01/16/2023] Open
Abstract
Metabolites have recently been found to be involved in significant biological regulation and changes. Itaconate, an important intermediate metabolite isolated from the tricarboxylic acid cycle, is derived from cis-aconitate decarboxylation mediated by immune response gene 1 in mitochondrial matrix. Itaconate has emerged as a key autocrine regulatory component involved in the development and progression of inflammation and immunity. It could directly modify cysteine sites on functional substrate proteins which related to inflammasome, signal transduction, transcription, and cell death. Itaconate can be a connector among immunity, metabolism, and inflammation, which is of great significance for further understanding the mechanism of cellular immune metabolism. And it could be the potential choice for the treatment of inflammation and immune-related diseases. This study is a systematic review of the potential mechanisms of metabolite associated with different pathology conditions. We briefly summarize the structural characteristics and classical pathways of itaconate and its derivatives, with special emphasis on its promising role in future clinical application, in order to provide theoretical basis for future research and treatment intervention.
Collapse
Affiliation(s)
| | | | | | | | - Lina Yu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
28
|
Mainali R, Zabalawi M, Long D, Buechler N, Quillen E, Key CC, Zhu X, Parks JS, Furdui C, Stacpoole PW, Martinez J, McCall CE, Quinn MA. Dichloroacetate reverses sepsis-induced hepatic metabolic dysfunction. eLife 2021; 10:64611. [PMID: 33616039 PMCID: PMC7901874 DOI: 10.7554/elife.64611] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/17/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming between resistance and tolerance occurs within the immune system in response to sepsis. While metabolic tissues such as the liver are subjected to damage during sepsis, how their metabolic and energy reprogramming ensures survival is unclear. Employing comprehensive metabolomic, lipidomic, and transcriptional profiling in a mouse model of sepsis, we show that hepatocyte lipid metabolism, mitochondrial tricarboxylic acid (TCA) energetics, and redox balance are significantly reprogrammed after cecal ligation and puncture (CLP). We identify increases in TCA cycle metabolites citrate, cis-aconitate, and itaconate with reduced fumarate and triglyceride accumulation in septic hepatocytes. Transcriptomic analysis of liver tissue supports and extends the hepatocyte findings. Strikingly, the administration of the pyruvate dehydrogenase kinase (PDK) inhibitor dichloroacetate reverses dysregulated hepatocyte metabolism and mitochondrial dysfunction. In summary, our data indicate that sepsis promotes hepatic metabolic dysfunction and that targeting the mitochondrial PDC/PDK energy homeostat rebalances transcriptional and metabolic manifestations of sepsis within the liver.
Collapse
Affiliation(s)
- Rabina Mainali
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | - Manal Zabalawi
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | - David Long
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | - Nancy Buechler
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | - Ellen Quillen
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | - Chia-Chi Key
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | - Xuewei Zhu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | - John S Parks
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | - Cristina Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | - Peter W Stacpoole
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine and Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, United States
| | - Jennifer Martinez
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, Bethesda, United States
| | - Charles E McCall
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| | - Matthew A Quinn
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, United States.,Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, United States
| |
Collapse
|
29
|
Zhang C, Dong L, Wu J, Qiao S, Xu W, Ma S, Zhao B, Wang X. Intervention of resistant starch 3 on type 2 diabetes mellitus and its mechanism based on urine metabonomics by liquid chromatography-tandem mass spectrometry. Biomed Pharmacother 2020; 128:110350. [PMID: 32521455 DOI: 10.1016/j.biopha.2020.110350] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/24/2020] [Accepted: 06/01/2020] [Indexed: 12/24/2022] Open
Abstract
As a severe metabolic disease, type 2 diabetes mellitus (T2DM) has aroused increasing public attentions. Resistant starch 3 (RS3), as a starch resistant to enzymatic hydrolysis owing to its special structure, has a good effect on improving insulin resistance and reducing blood sugar in T2DM patients. However, the possible mechanisms were barely interpreted yet. In our research, we aimed to evaluate the effects and the possible mechanisms of RS3 on the treatment of T2DM. ICR mice treated with high-fat diet (HFD) for eight weeks, and then injected with streptozotocin (STZ) (100 mg/kg) to establish the T2DM. We choose the mice with the fast blood glucose (FBG) more than 11 mmol/L as T2DM. After treated for 11 weeks the relevant data was analyzed. According to the results, the FBG was dramatically reduced (p < 0.05), which also downregulated triglyceride (p < 0.01) and total cholesterol (p < 0.01). Additionally, the insulin resistance indexes were significantly reduced (p < 0.01), the homeostasis model assessment-β and insulin-sensitive index were significantly improved (p < 0.01) in RS3 group. Meanwhile, the metabolic profiles of urine were analyzed and 29 potential biomarkers were screened out, including amino acids and lipids. In conclusion, we speculated that the tricarboxylic acid cycle, amino acid metabolism and lipid metabolism played roles in the therapeutic mechanisms of RS3 on T2DM.
Collapse
Affiliation(s)
- Caijuan Zhang
- Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China
| | - Ling Dong
- Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China
| | - Jiahui Wu
- Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China
| | - Sanyang Qiao
- Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China
| | - Wenjuan Xu
- Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China
| | - Shuangshuang Ma
- Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China
| | - Baosheng Zhao
- Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China
| | - Xueyong Wang
- Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China.
| |
Collapse
|