1
|
Seyfried TN, Lee DC, Duraj T, Ta NL, Mukherjee P, Kiebish M, Arismendi-Morillo G, Chinopoulos C. The Warburg hypothesis and the emergence of the mitochondrial metabolic theory of cancer. J Bioenerg Biomembr 2025:10.1007/s10863-025-10059-w. [PMID: 40199815 DOI: 10.1007/s10863-025-10059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Otto Warburg originally proposed that cancer arose from a two-step process. The first step involved a chronic insufficiency of mitochondrial oxidative phosphorylation (OxPhos), while the second step involved a protracted compensatory energy synthesis through lactic acid fermentation. His extensive findings showed that oxygen consumption was lower while lactate production was higher in cancerous tissues than in non-cancerous tissues. Warburg considered both oxygen consumption and extracellular lactate as accurate markers for ATP production through OxPhos and glycolysis, respectively. Warburg's hypothesis was challenged from findings showing that oxygen consumption remained high in some cancer cells despite the elevated production of lactate suggesting that OxPhos was largely unimpaired. New information indicates that neither oxygen consumption nor lactate production are accurate surrogates for quantification of ATP production in cancer cells. Warburg also did not know that a significant amount of ATP could come from glutamine-driven mitochondrial substrate level phosphorylation in the glutaminolysis pathway with succinate produced as end product, thus confounding the linkage of oxygen consumption to the origin of ATP production within mitochondria. Moreover, new information shows that cytoplasmic lipid droplets and elevated aerobic lactic acid fermentation are both biomarkers for OxPhos insufficiency. Warburg's original hypothesis can now be linked to a more complete understanding of how OxPhos insufficiency underlies dysregulated cancer cell growth. These findings can also address several questionable assumptions regarding the origin of cancer thus allowing the field to advance with more effective therapeutic strategies for a less toxic metabolic management and prevention of cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA.
| | - Derek C Lee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Tomas Duraj
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Nathan L Ta
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | | | - Gabriel Arismendi-Morillo
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, Venezuela
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao (Bizkaia), Spain
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
2
|
Andrianova NV, Buyan MI, Brezgunova AA, Cherkesova KS, Zorov DB, Plotnikov EY. Hemorrhagic Shock and Mitochondria: Pathophysiology and Therapeutic Approaches. Int J Mol Sci 2025; 26:1843. [PMID: 40076469 PMCID: PMC11898946 DOI: 10.3390/ijms26051843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Severe injuries and some pathologies associated with massive bleeding, such as maternal hemorrhage, gastrointestinal and perioperative bleeding, and rupture of an aneurysm, often lead to major blood loss and the development of hemorrhagic shock. A sharp decrease in circulating blood volume triggers a vicious cycle of vasoconstriction and coagulopathy leading to ischemia of all internal organs and, in severe decompensated states, ischemia of the brain and heart. The basis of tissue damage and dysfunction in hemorrhagic shock is an interruption in the supply of oxygen and substrates for energy production to the cells, making the mitochondria a source and target of oxidative stress and proapoptotic signaling. Based on these mechanisms, different strategies are proposed to treat the multiple organ failure that occurs in shock. The main direction of such treatment is to provide the cells with a sufficient amount of substrates that utilize oxidative phosphorylation at different stages and increase the efficiency of energy production by the mitochondria. These strategies include restoring the efficiency of mitochondrial complexes, for example, by restoring the nicotinamide adenine dinucleotide (NAD) pool. Another direction is approaches to minimize oxidative stress as well as apoptosis, which are primarily dependent on the mitochondria. There are also a number of other methods to reduce mitochondrial dysfunction and improve the quality of the mitochondrial population. In this review, we consider such strategies for the treatment of hemorrhagic shock and show the promise of therapeutic approaches aimed at restoring the bioenergetic functions of the cell and protecting mitochondria.
Collapse
Affiliation(s)
- Nadezda V. Andrianova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
| | - Marina I. Buyan
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Anna A. Brezgunova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
| | - Kseniia S. Cherkesova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (N.V.A.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia
| |
Collapse
|
3
|
Qiu M, Geng H, Zou C, Zhao X, Zhao C, Xie J, Wang J, Zhang N, Hu Y, Fu Y, Wang J, Hu X. Intestinal inflammation exacerbates endometritis through succinate production by gut microbiota and SUCNR1-mediated proinflammatory response. Int Immunopharmacol 2025; 146:113919. [PMID: 39736240 DOI: 10.1016/j.intimp.2024.113919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025]
Abstract
Endometritis poses higher health risks to women. Clinical practice has found that gastrointestinal dysfunction is more likely to lead to the occurrence of endometritis. However, the mechanism is unclear. This study explored the influence and mechanism of DSS-induced intestinal inflammation on endometritis. Our findings demonstrate that DSS-induced intestinal inflammation can worsen LPS-induced endometritis in mice, and this effect is dependent on the gut microbiota, as depleting the gut microbiota eliminates this protective effect. Similarly, FMT from DSS-treated mice to recipient mice exacerbates LPS-induced endometritis. In addition, treatment of DSS disrupted an imbalance of succinate-producing and succinate-consuming bacteria and increased the levels of succinate in the gut and uterine tissues. Furthermore, treatment with succinate aggravates LPS-induced endometritis by activating the succinate receptor 1 (SUCNR1), evidenced by inhibition of the activation of SUCNR1 reversed the inflammatory response in uterine tissues induced by succinate during endometritis induced by LPS. Collectively, the results suggested that dysbiosis of the gut microbiota exacerbates LPS-induced endometritis by production and migration of succinate from gut to uterine tissues via the gut-uterus axis, then activates the SUCNR1. This identifies gut-derived succinate as a novel target for treating endometritis, and it indicates that targeting the gut microbiota and its metabolism could be a potential strategy for intervention in endometritis.
Collapse
Affiliation(s)
- Min Qiu
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Huafeng Geng
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Chenyu Zou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Xiaotong Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Caijun Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Jiaxin Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Jinnan Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Yubo Hu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Junrong Wang
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China.
| | - Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China.
| |
Collapse
|
4
|
Wei B, Zheng J, Chai J, Huang J, Duan H, Han S, Yang X, Zhang W, Hu F, Qu Y, Liu X, Liu T, Wu Y, Chi Y. Metabolomic and proteomic profiling of a burn-hemorrhagic shock swine model reveals a metabolomic signature associated with fatal outcomes. Eur J Med Res 2025; 30:10. [PMID: 39773520 PMCID: PMC11706163 DOI: 10.1186/s40001-024-02245-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Burn-hemorrhagic shock combined injury, a severe condition causing complex stress responses and metabolic disturbances that significantly affect clinical outcomes in both military and civilian settings, was modeled in swine to investigate the associated metabolomic and proteomic changes and identify potential biomarkers for disease prognosis. METHODS Eight clean-grade adult male Landrace pigs (4-5 months, average weight 60-70 kg) were used to model burn-hemorrhagic shock combined injury. Serum samples collected at 0 h and 2 h post-injury were analyzed using metabolomic and proteomic measurements. The metabolomic and proteomic data were processed through partial least squares-discriminant analysis (PLS-DA) and the KEGG enrichment etc. Furthermore, the integrate analysis of the metabolomic and proteomic data was generalized by canonical correlation discriminant analysis, and the correlation between metabolites and mortality of the swine model was predicted using a multiple linear regression model by Pearson analysis. RESULTS PLS-DA revealed a global shift in each of the metabolomic and proteomic profiles following injury. The levels of 87 signature metabolites including various types of amino acids, fatty acids and acyl-carnitines of different lengths, and many metabolites in the gluconeogenesis, glycolysis, and tricarboxylic acid (TCA) cycle are generally increased (P < 0.05) after injury and can be used as biomarkers. Pathways related to amino acids metabolism and TCA cycle were significantly enriched (P < 0.01). In proteome analysis, we found dramatically altered (P < 0.05) levels of matrix and red blood cell-related proteins, such as type I collagen and hemoglobin. Most importantly, we found that the markedly elevated (P < 0.01) succinic acid, glutaric acid, and malic acid are closely associated (r = 0.863, 0.861, and 0.821, respectively) with injury severity by Pearson analysis, and can predict mortality using a multiple linear regression model. CONCLUSIONS The study provides compelling observations that burn-shock swine model undergoes dramatic changes in the acute phase and present a valuable panel for clinical use of prognosis.
Collapse
Affiliation(s)
- Bin Wei
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
- The First Department of Surgery, Chinese People's Armed Police Force Hospital of Beijing, Beijing, 100027, China
| | - Jinguang Zheng
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Jiake Chai
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China.
| | - Jianxiang Huang
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Hongjie Duan
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Shaofang Han
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Xiaolin Yang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Wenjia Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Fangchao Hu
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Yirui Qu
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Xiangyu Liu
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Tian Liu
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Yushou Wu
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Yunfei Chi
- Department of Burns and Plastic Surgery, The Fourth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China.
| |
Collapse
|
5
|
Marhl M. What do stimulated beta cells have in common with cancer cells? Biosystems 2024; 242:105257. [PMID: 38876357 DOI: 10.1016/j.biosystems.2024.105257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
This study investigates the metabolic parallels between stimulated pancreatic beta cells and cancer cells, focusing on glucose and glutamine metabolism. Addressing the significant public health challenges of Type 2 Diabetes (T2D) and cancer, we aim to deepen our understanding of the mechanisms driving insulin secretion and cellular proliferation. Our analysis of anaplerotic cycles and the role of NADPH in biosynthesis elucidates their vital functions in both processes. Additionally, we point out that both cell types share an antioxidative response mediated by the Nrf2 signaling pathway, glutathione synthesis, and UCP2 upregulation. Notably, UCP2 facilitates the transfer of C4 metabolites, enhancing reductive TCA cycle metabolism. Furthermore, we observe that hypoxic responses are transient in beta cells post-stimulation but persistent in cancer cells. By synthesizing these insights, the research may suggest novel therapeutic targets for T2D, highlighting the shared metabolic strategies of stimulated beta cells and cancer cells. This comparative analysis not only illuminates the metabolic complexity of these conditions but also emphasizes the crucial role of metabolic pathways in cell function and survival, offering fresh perspectives for tackling T2D and cancer challenges.
Collapse
Affiliation(s)
- Marko Marhl
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia; Faculty of Education, University of Maribor, Koroška cesta 160, 2000, Maribor, Slovenia; Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000, Maribor, Slovenia.
| |
Collapse
|
6
|
LaCroix IS, Moore EE, Cralley A, Cendali FI, Dzieciatkowska M, Hom P, Mitra S, Cohen M, Silliman C, Hansen KC, D'Alessandro A. Multiomics Signatures of Coagulopathy in a Polytrauma Swine Model Contrasted with Severe Multisystem Injured Patients. J Proteome Res 2024; 23:1163-1173. [PMID: 38386921 DOI: 10.1021/acs.jproteome.3c00581] [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] [Indexed: 02/24/2024]
Abstract
Trauma-induced coagulopathy (TIC) is a leading contributor to preventable mortality in severely injured patients. Understanding the molecular drivers of TIC is an essential step in identifying novel therapeutics to reduce morbidity and mortality. This study investigated multiomics and viscoelastic responses to polytrauma using our novel swine model and compared these findings with severely injured patients. Molecular signatures of TIC were significantly associated with perturbed coagulation and inflammation systems as well as extensive hemolysis. These results were consistent with patterns observed in trauma patients who had multisystem injuries. Here, intervention using resuscitative endovascular balloon occlusion of the aorta following polytrauma in our swine model revealed distinct multiomics alterations as a function of placement location. Aortic balloon placement in zone-1 worsened ischemic damage and mitochondrial dysfunction, patterns that continued throughout the monitored time course. While placement in zone-III showed a beneficial effect on TIC, it showed an improvement in effective coagulation. Taken together, this study highlights the translational relevance of our polytrauma swine model for investigating therapeutic interventions to correct TIC in patients.
Collapse
Affiliation(s)
- Ian S LaCroix
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Ernest E Moore
- Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Ernest E Moore Shock Trauma Center at Denver Health, Denver, Colorado 80204, United States
| | - Alexis Cralley
- Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Francesca I Cendali
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Patrick Hom
- Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Sanchayita Mitra
- Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Mitchell Cohen
- Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Christopher Silliman
- Vitalant Research Institute, Denver, Colorado 80230, United States
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
7
|
Taghavi S, Campbell A, Engelhardt D, Duchesne J, Shaheen F, Pociask D, Kolls J, Jackson-Weaver O. Dimethyl malonate protects the lung in a murine model of acute respiratory distress syndrome. J Trauma Acute Care Surg 2024; 96:386-393. [PMID: 37934622 PMCID: PMC10922501 DOI: 10.1097/ta.0000000000004184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
BACKGROUND Succinate is a proinflammatory citric acid cycle metabolite that accumulates in tissues during pathophysiological states. Oxidation of succinate after ischemia-reperfusion leads to reversal of the electron transport chain and generation of reactive oxygen species. Dimethyl malonate (DMM) is a competitive inhibitor of succinate dehydrogenase, which has been shown to reduce succinate accumulation. We hypothesized that DMM would protect against inflammation in a murine model of ARDS. METHODS C57BL/6 mice were given ARDS via 67.7 μg of intratracheally administered lipopolysaccharide. Dimethyl malonate (50 mg/kg) was administered via tail vein injection 30 minutes after injury, then daily for 3 days. The animals were sacrificed on day 4 after bronchoalveolar lavage (BAL). Bronchoalveolar lavage cell counts were performed to examine cellular influx. Supernatant protein was quantified via Bradford protein assay. Animals receiving DMM (n = 8) were compared with those receiving sham injection (n = 8). Cells were fixed and stained with FITC-labeled wheat germ agglutinin to quantify the endothelial glycocalyx (EGX). RESULTS Total cell counts in BAL was less for animals receiving DMM (6.93 × 10 6 vs. 2.46 × 10 6 , p = 0.04). The DMM group had less BAL macrophages (168.6 vs. 85.1, p = 0.04) and lymphocytes (527.7 vs. 248.3; p = 0.04). Dimethyl malonate-treated animals had less protein leak in BAL than sham treated (1.48 vs. 1.15 μg/μl, p = 0.03). Treatment with DMM resulted in greater staining intensity of the EGX in the lung when compared with sham (12,016 vs. 15,186 arbitrary units, p = 0.03). Untreated animals had a greater degree of weight loss than treated animals (3.7% vs. 1.1%, p = 0.04). Dimethyl malonate prevented the upregulation of monocyte chemoattractant protein-1 (1.66 vs. 0.92 RE, p = 0.02) and ICAM-1 (1.40 vs. 1.01 RE, p = 0.05). CONCLUSION Dimethyl malonate reduces lung inflammation and capillary leak in ARDS. This may be mediated by protection of the EGX and inhibition of monocyte chemoattractant protein-1 and ICAM-1. Dimethyl malonate may be a novel therapeutic for ARDS.
Collapse
Affiliation(s)
- Sharven Taghavi
- Tulane University School of Medicine, Department of Surgery, New Orleans, Louisiana
| | - Alexandra Campbell
- Tulane University School of Medicine, Department of Surgery, New Orleans, Louisiana
| | - David Engelhardt
- Tulane University School of Medicine, Department of Surgery, New Orleans, Louisiana
| | - Juan Duchesne
- Tulane University School of Medicine, Department of Surgery, New Orleans, Louisiana
| | - Farhana Shaheen
- Tulane University School of Medicine, Department of Surgery, New Orleans, Louisiana
| | - Derek Pociask
- Tulane University School of Medicine, Department of Medicine, New Orleans, Louisiana
| | - Jay Kolls
- Tulane University School of Medicine, Center for Translational Research in Infection and Inflammation, New Orleans, LA
| | - Olan Jackson-Weaver
- Tulane University School of Medicine, Department of Surgery, New Orleans, Louisiana
| |
Collapse
|
8
|
LaCroix IS, Cralley A, Moore EE, Cendali FI, Dzieciatkowska M, Hom P, Mitra S, Cohen M, Silliman C, Sauaia A, Hansen KC, D’Alessandro A. Omics Signatures of Tissue Injury and Hemorrhagic Shock in Swine. Ann Surg 2023; 278:e1299-e1312. [PMID: 37334680 PMCID: PMC10728352 DOI: 10.1097/sla.0000000000005944] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
OBJECTIVE Advanced mass spectrometry methods were leveraged to analyze both proteomics and metabolomics signatures in plasma upon controlled tissue injury (TI) and hemorrhagic shock (HS)-isolated or combined-in a swine model, followed by correlation to viscoelastic measurements of coagulopathy via thrombelastography. BACKGROUND TI and HS cause distinct molecular changes in plasma in both animal models and trauma patients. However, the contribution to coagulopathy of trauma, the leading cause of preventable mortality in this patient population remains unclear. The recent development of a swine model for isolated or combined TI+HS facilitated the current study. METHODS Male swine (n=17) were randomized to either isolated or combined TI and HS. Coagulation status was analyzed by thrombelastography during the monitored time course. The plasma fractions of the blood draws (at baseline; end of shock; and at 30 minutes, 1, 2, and 4 hours after shock) were analyzed by mass spectrometry-based proteomics and metabolomics workflows. RESULTS HS-isolated or combined with TI-caused the most severe omic alterations during the monitored time course. While isolated TI delayed the activation of coagulation cascades. Correlation to thrombelastography parameters of clot strength (maximum amplitude) and breakdown (LY30) revealed signatures of coagulopathy which were supported by analysis of gene ontology-enriched biological pathways. CONCLUSION The current study provides a comprehensive characterization of proteomic and metabolomic alterations to combined or isolated TI and HS in a swine model and identifies early and late omics correlates to viscoelastic measurements in this system.
Collapse
Affiliation(s)
- Ian S. LaCroix
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Alexis Cralley
- Department of Surgery, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Ernest E. Moore
- Department of Surgery, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
- Ernest E Moore Shock Trauma Center at Denver Health, Denver, CO, USA
| | - Francesca I Cendali
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Patrick Hom
- Department of Surgery, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Sanchayita Mitra
- Department of Surgery, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | | | - Christopher Silliman
- Vitalant Research Institute, Denver, CO, USA
- Department of Pediatrics, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Angela Sauaia
- Ernest E Moore Shock Trauma Center at Denver Health, Denver, CO, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
9
|
Oh CJ, Kim MJ, Lee JM, Kim DH, Kim IY, Park S, Kim Y, Lee KB, Lee SH, Lim CW, Kim M, Lee JY, Pagire HS, Pagire SH, Bae MA, Chanda D, Thoudam T, Khang AR, Harris RA, Ahn JH, Jeon JH, Lee IK. Inhibition of pyruvate dehydrogenase kinase 4 ameliorates kidney ischemia-reperfusion injury by reducing succinate accumulation during ischemia and preserving mitochondrial function during reperfusion. Kidney Int 2023; 104:724-739. [PMID: 37399974 DOI: 10.1016/j.kint.2023.06.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 05/26/2023] [Accepted: 06/02/2023] [Indexed: 07/05/2023]
Abstract
Ischemia-reperfusion (IR) injury, a leading cause of acute kidney injury (AKI), is still without effective therapies. Succinate accumulation during ischemia followed by its oxidation during reperfusion leads to excessive reactive oxygen species (ROS) and severe kidney damage. Consequently, the targeting of succinate accumulation may represent a rational approach to the prevention of IR-induced kidney injury. Since ROS are generated primarily in mitochondria, which are abundant in the proximal tubule of the kidney, we explored the role of pyruvate dehydrogenase kinase 4 (PDK4), a mitochondrial enzyme, in IR-induced kidney injury using proximal tubule cell-specific Pdk4 knockout (Pdk4ptKO) mice. Knockout or pharmacological inhibition of PDK4 ameliorated IR-induced kidney damage. Succinate accumulation during ischemia, which is responsible for mitochondrial ROS production during reperfusion, was reduced by PDK4 inhibition. PDK4 deficiency established conditions prior to ischemia resulting in less succinate accumulation, possibly because of a reduction in electron flow reversal in complex II, which provides electrons for the reduction of fumarate to succinate by succinate dehydrogenase during ischemia. The administration of dimethyl succinate, a cell-permeable form of succinate, attenuated the beneficial effects of PDK4 deficiency, suggesting that the kidney-protective effect is succinate-dependent. Finally, genetic or pharmacological inhibition of PDK4 prevented IR-induced mitochondrial damage in mice and normalized mitochondrial function in an in vitro model of IR injury. Thus, inhibition of PDK4 represents a novel means of preventing IR-induced kidney injury, and involves the inhibition of ROS-induced kidney toxicity through reduction in succinate accumulation and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Chang Joo Oh
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Min-Ji Kim
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Ji-Min Lee
- Cell & Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Dong Hun Kim
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Republic of Korea
| | - Il-Young Kim
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea; Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Sanghee Park
- Department of Exercise Rehabilitation, Gachon University, Incheon, Republic of Korea
| | - Yeongmin Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Kyung-Bok Lee
- Center for Research Equipment (104-Dong), Korea Basic Science Institute, Ochang, Cheongju, Chungbuk, Republic of Korea
| | - Sang-Hee Lee
- Center for Research Equipment (104-Dong), Korea Basic Science Institute, Ochang, Cheongju, Chungbuk, Republic of Korea
| | - Chae Won Lim
- Department of Medicine, Graduate School, Daegu Catholic University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Myeongjin Kim
- Department of Medicine, Graduate School, Daegu Catholic University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Jung-Yi Lee
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Haushabhau S Pagire
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Suvarna H Pagire
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Myung Ae Bae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Dipanjan Chanda
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Ah Reum Khang
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University College of Medicine, Yangsan, Republic of Korea
| | - Robert A Harris
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea.
| | - Jae-Han Jeon
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea.
| | - In-Kyu Lee
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.
| |
Collapse
|
10
|
Cohen MJ, Erickson CB, Lacroix IS, Debot M, Dzieciatkowska M, Schaid TR, Hallas MW, Thielen ON, Cralley AL, Banerjee A, Moore EE, Silliman CC, D'Alessandro A, Hansen KC. Trans-Omics analysis of post injury thrombo-inflammation identifies endotypes and trajectories in trauma patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553446. [PMID: 37645811 PMCID: PMC10462097 DOI: 10.1101/2023.08.16.553446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Understanding and managing the complexity of trauma-induced thrombo-inflammation necessitates an innovative, data-driven approach. This study leveraged a trans-omics analysis of longitudinal samples from trauma patients to illuminate molecular endotypes and trajectories that underpin patient outcomes, transcending traditional demographic and physiological characterizations. We hypothesize that trans-omics profiling reveals underlying clinical differences in severely injured patients that may present with similar clinical characteristics but ultimately have very different responses to treatment and clinical outcomes. Here we used proteomics and metabolomics to profile 759 of longitudinal plasma samples from 118 patients at 11 time points and 97 control subjects. Results were used to define distinct patient states through data reduction techniques. The patient groups were stratified based on their shock severity and injury severity score, revealing a spectrum of responses to trauma and treatment that are fundamentally tied to their unique underlying biology. Ensemble models were then employed, demonstrating the predictive power of these molecular signatures with area under the receiver operating curves of 80 to 94% for key outcomes such as INR, ICU-free days, ventilator-free days, acute lung injury, massive transfusion, and death. The molecularly defined endotypes and trajectories provide an unprecedented lens to understand and potentially guide trauma patient management, opening a path towards precision medicine. This strategy presents a transformative framework that aligns with our understanding that trauma patients, despite similar clinical presentations, might harbor vastly different biological responses and outcomes.
Collapse
|
11
|
LaCroix IS, Cohen M, Moore EE, Dzieciatkowska M, Silliman CC, Hansen KC, D'Alessandro A. Omics markers of platelet transfusion in trauma patients. Transfusion 2023; 63:1447-1462. [PMID: 37466356 DOI: 10.1111/trf.17472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/26/2023] [Accepted: 05/30/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Even in the era of the COVID-19 pandemic, trauma remains the global leading cause of mortality under the age of 49. Trauma-induced coagulopathy is a leading driver of early mortality in critically ill patients, and transfusion of platelet products is a life-saving intervention to restore hemostasis in the bleeding patient. However, despite extensive functional studies based on viscoelastic assays, limited information is available about the impact of platelet transfusion on the circulating molecular signatures in trauma patients receiving platelet transfusion. MATERIALS AND METHODS To bridge this gap, we leveraged metabolomics and proteomics approaches to characterize longitudinal plasma samples (n = 118; up to 11 time points; total samples: 759) from trauma patients enrolled in the Control Of Major Bleeding After Trauma (COMBAT) study. Samples were collected in the field, in the emergency department (ED), and at intervals up to 168 h (7 days) post-hospitalization. Transfusion of platelet (PLT) products was performed (n = 30; total samples: 250) in the ED through 24 h post-hospitalization. Longitudinal plasma samples were subjected to mass spectrometry-based metabolomics and proteomics workflows. Multivariate analyses were performed to determine omics markers of transfusion of one, two, three, or more PLT transfusions. RESULTS Higher levels of tranexamic acid (TXA), inflammatory proteins, carnitines, and polyamines were detected in patients requiring PLT transfusion. Correlation of PLT units with omics data suggested sicker patients required more units and partially overlap with the population requiring transfusion of packed red blood cell products. Furthermore, platelet activation was likely increased in the most severely injured patients. Fatty acid levels were significantly lower in PLT transfusion recipients (at time of maximal transfusion: Hour 4) compared with non-recipients, while carnitine levels were significantly higher. Fatty acid levels restore later in the time course (e.g., post-PLT transfusion). DISCUSSION The present study provides the first multi-omics characterization of platelet transfusion efficacy in a clinically relevant cohort of trauma patients. Physiological alterations following transfusion were detected, highlighting the efficacy of mass spectrometry-based omics techniques to improve personalized transfusion medicine. More specialized clinical research studies focused on PLT transfusion, including organized pre and post transfusion sample collection and limitation to PLT products only, are required to fully understand subsequent metabolomic and proteomic alterations.
Collapse
Affiliation(s)
- Ian S LaCroix
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mitchell Cohen
- Department of Surgery, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ernest E Moore
- Department of Surgery, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
- "Ernest E Moore" Trauma Center at Denver Health, Denver, Colorado, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christopher C Silliman
- Vitalant Research Institute, Denver, Colorado, USA
- Department of Pediatrics, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
12
|
D'Alessandro A. Red Blood Cell Omics and Machine Learning in Transfusion Medicine: Singularity Is Near. Transfus Med Hemother 2023; 50:174-183. [PMID: 37434999 PMCID: PMC10331163 DOI: 10.1159/000529744] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/14/2023] [Indexed: 07/30/2023] Open
Abstract
Background Blood transfusion is a life-saving intervention for millions of recipients worldwide. Over the last 15 years, the advent of high-throughput, affordable omics technologies - including genomics, proteomics, lipidomics, and metabolomics - has allowed transfusion medicine to revisit the biology of blood donors, stored blood products, and transfusion recipients. Summary Omics approaches have shed light on the genetic and non-genetic factors (environmental or other exposures) impacting the quality of stored blood products and efficacy of transfusion events, based on the current Food and Drug Administration guidelines (e.g., hemolysis and post-transfusion recovery for stored red blood cells). As a treasure trove of data accumulates, the implementation of machine learning approaches promises to revolutionize the field of transfusion medicine, not only by advancing basic science. Indeed, computational strategies have already been used to perform high-content screenings of red blood cell morphology in microfluidic devices, generate in silico models of erythrocyte membrane to predict deformability and bending rigidity, or design systems biology maps of the red blood cell metabolome to drive the development of novel storage additives. Key Message In the near future, high-throughput testing of donor genomes via precision transfusion medicine arrays and metabolomics of all donated products will be able to inform the development and implementation of machine learning strategies that match, from vein to vein, donors, optimal processing strategies (additives, shelf life), and recipients, realizing the promise of personalized transfusion medicine.
Collapse
Affiliation(s)
- Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
13
|
Wang YH, Yan ZZ, Luo SD, Hu JJ, Wu M, Zhao J, Liu WF, Li C, Liu KX. Gut microbiota-derived succinate aggravates acute lung injury after intestinal ischaemia/reperfusion in mice. Eur Respir J 2023; 61:13993003.00840-2022. [PMID: 36229053 DOI: 10.1183/13993003.00840-2022] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 10/02/2022] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Acute lung injury (ALI) is a major cause of morbidity and mortality after intestinal ischaemia/reperfusion (I/R). The gut microbiota and its metabolic byproducts act as important modulators of the gut-lung axis. This study aimed to define the role of succinate, a key microbiota metabolite, in intestinal I/R-induced ALI progression. METHODS Gut and lung microbiota of mice subjected to intestinal I/R were analysed using 16S rRNA gene sequencing. Succinate level alterations were measured in germ-free mice or conventional mice treated with antibiotics. Succinate-induced alveolar macrophage polarisation and its effects on alveolar epithelial apoptosis were evaluated in succinate receptor 1 (Sucnr1)-deficient mice and in murine alveolar macrophages transfected with Sucnr1-short interfering RNA. Succinate levels were measured in patients undergoing cardiopulmonary bypass, including intestinal I/R. RESULTS Succinate accumulated in lungs after intestinal I/R, and this was associated with an imbalance of succinate-producing and succinate-consuming bacteria in the gut, but not the lungs. Succinate accumulation was absent in germ-free mice and was reversed by gut microbiota depletion with antibiotics, indicating that the gut microbiota is a source of lung succinate. Moreover, succinate promoted alveolar macrophage polarisation, alveolar epithelial apoptosis and lung injury during intestinal I/R. Conversely, knockdown of Sucnr1 or blockage of SUCNR1 in vitro and in vivo reversed the effects of succinate by modulating the phosphoinositide 3-kinase-AKT/hypoxia-inducible factor-1α pathway. Plasma succinate levels significantly correlated with intestinal I/R-related lung injury after cardiopulmonary bypass. CONCLUSION Gut microbiota-derived succinate exacerbates intestinal I/R-induced ALI through SUCNR1-dependent alveolar macrophage polarisation, identifying succinate as a novel target for gut-derived ALI in critically ill patients.
Collapse
Affiliation(s)
- Yi-Heng Wang
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Anaesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Yi-Heng Wang and Zheng-Zheng Yan contributed equally
| | - Zheng-Zheng Yan
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Yi-Heng Wang and Zheng-Zheng Yan contributed equally
| | - Si-Dan Luo
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Juan Hu
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Wu
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Zhao
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei-Feng Liu
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cai Li
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Cai Li and Ke-Xuan Liu contributed equally to this article as lead authors and supervised the work
| | - Ke-Xuan Liu
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Cai Li and Ke-Xuan Liu contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
14
|
Atallah R, Olschewski A, Heinemann A. Succinate at the Crossroad of Metabolism and Angiogenesis: Roles of SDH, HIF1α and SUCNR1. Biomedicines 2022; 10:3089. [PMID: 36551845 PMCID: PMC9775124 DOI: 10.3390/biomedicines10123089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Angiogenesis is an essential process by which new blood vessels develop from existing ones. While adequate angiogenesis is a physiological process during, for example, tissue repair, insufficient and excessive angiogenesis stands on the pathological side. Fine balance between pro- and anti-angiogenic factors in the tissue environment regulates angiogenesis. Identification of these factors and how they function is a pressing topic to develop angiogenesis-targeted therapeutics. During the last decade, exciting data highlighted non-metabolic functions of intermediates of the mitochondrial Krebs cycle including succinate. Among these functions is the contribution of succinate to angiogenesis in various contexts and through different mechanisms. As the concept of targeting metabolism to treat a wide range of diseases is rising, in this review we summarize the mechanisms by which succinate regulates angiogenesis in normal and pathological settings. Gaining a comprehensive insight into how this metabolite functions as an angiogenic signal will provide a useful approach to understand diseases with aberrant or excessive angiogenic background, and may provide strategies to tackle them.
Collapse
Affiliation(s)
- Reham Atallah
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Akos Heinemann
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
15
|
Seyfried TN, Arismendi-Morillo G, Zuccoli G, Lee DC, Duraj T, Elsakka AM, Maroon JC, Mukherjee P, Ta L, Shelton L, D'Agostino D, Kiebish M, Chinopoulos C. Metabolic management of microenvironment acidity in glioblastoma. Front Oncol 2022; 12:968351. [PMID: 36059707 PMCID: PMC9428719 DOI: 10.3389/fonc.2022.968351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma (GBM), similar to most cancers, is dependent on fermentation metabolism for the synthesis of biomass and energy (ATP) regardless of the cellular or genetic heterogeneity seen within the tumor. The transition from respiration to fermentation arises from the documented defects in the number, the structure, and the function of mitochondria and mitochondrial-associated membranes in GBM tissue. Glucose and glutamine are the major fermentable fuels that drive GBM growth. The major waste products of GBM cell fermentation (lactic acid, glutamic acid, and succinic acid) will acidify the microenvironment and are largely responsible for drug resistance, enhanced invasion, immunosuppression, and metastasis. Besides surgical debulking, therapies used for GBM management (radiation, chemotherapy, and steroids) enhance microenvironment acidification and, although often providing a time-limited disease control, will thus favor tumor recurrence and complications. The simultaneous restriction of glucose and glutamine, while elevating non-fermentable, anti-inflammatory ketone bodies, can help restore the pH balance of the microenvironment while, at the same time, providing a non-toxic therapeutic strategy for killing most of the neoplastic cells.
Collapse
Affiliation(s)
- Thomas N. Seyfried
- Biology Department, Boston College, Chestnut Hill, MA, United States
- *Correspondence: Thomas N. Seyfried,
| | - Gabriel Arismendi-Morillo
- Instituto de Investigaciones Biológicas, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Giulio Zuccoli
- The Program for the Study of Neurodevelopment in Rare Disorders (NDRD), University of Pittsburgh, Pittsburgh, PA, United States
| | - Derek C. Lee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Tomas Duraj
- Faculty of Medicine, Institute for Applied Molecular Medicine (IMMA), CEU San Pablo University, Madrid, Spain
| | - Ahmed M. Elsakka
- Neuro Metabolism, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Joseph C. Maroon
- Department of Neurosurgery, University of Pittsburgh, Medical Center, Pittsburgh, PA, United States
| | - Purna Mukherjee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Linh Ta
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | | | - Dominic D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, United States
| | | | | |
Collapse
|
16
|
Henriksen HH, Marín de Mas I, Herand H, Krocker J, Wade CE, Johansson PI. Metabolic systems analysis identifies a novel mechanism contributing to shock in patients with endotheliopathy of trauma (EoT) involving thromboxane A2 and LTC 4. Matrix Biol Plus 2022; 15:100115. [PMID: 35813244 PMCID: PMC9260291 DOI: 10.1016/j.mbplus.2022.100115] [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: 10/07/2021] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose Endotheliopathy of trauma (EoT), as defined by circulating levels of syndecan-1 ≥ 40 ng/mL, has been reported to be associated with significantly increased transfusion requirements and a doubled 30-day mortality. Increased shedding of the glycocalyx points toward the endothelial cell membrane composition as important for the clinical outcome being the rationale for this study. Results The plasma metabolome of 95 severely injured trauma patients was investigated by mass spectrometry, and patients with EoT vs. non-EoT were compared by partial least square-discriminant analysis, identifying succinic acid as the top metabolite to differentiate EoT and non-EoT patients (VIP score = 3). EoT and non-EoT patients' metabolic flux profile was inferred by integrating the corresponding plasma metabolome data into a genome-scale metabolic network reconstruction analysis and performing a functional study of the metabolic capabilities of each group. Model predictions showed a decrease in cholesterol metabolism secondary to impaired mevalonate synthesis in EoT compared to non-EoT patients. Intracellular task analysis indicated decreased synthesis of thromboxanA2 and leukotrienes, as well as a lower carnitine palmitoyltransferase I activity in EoT compared to non-EoT patients. Sensitivity analysis also showed a significantly high dependence of eicosanoid-associated metabolic tasks on alpha-linolenic acid as unique to EoT patients. Conclusions Model-driven analysis of the endothelial cells' metabolism identified potential novel targets as impaired thromboxane A2 and leukotriene synthesis in EoT patients when compared to non-EoT patients. Reduced thromboxane A2 and leukotriene availability in the microvasculature impairs vasoconstriction ability and may thus contribute to shock in EoT patients. These findings are supported by extensive scientific literature; however, further investigations are required on these findings.
Collapse
Key Words
- AA, Arachidonic acid
- CPT1, Carnitine palmitoyltransferase I
- EC, Endothelial cell
- EC-GEM, Genome-scale metabolic model of the microvascular endothelial cell
- ELISA, Enzyme-linked immunosorbent assay
- Eicosanoid
- Endotheliopathy
- EoT, Endotheliopathy of trauma
- FBA, Flux balance analysis
- GEMs, Genome-scale metabolic models
- Genome-scale metabolic model
- HMG-CoA, Hydroxymethylglutaryl-CoA
- ISS, Injury Severity Score
- LTC4, Leukotriene C4
- Metabolomics
- PCA, Principal Component Analysis
- PLS-DA, Partial least square-discriminant analysis
- Systems biology
- Trauma
Collapse
Affiliation(s)
- Hanne H. Henriksen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Igor Marín de Mas
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Denmark
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark
| | - Helena Herand
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Denmark
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark
| | - Joseph Krocker
- Center for Translational Injury Research, Department of Surgery, University of Texas Health Science Center, Houston, TX, USA
| | - Charles E. Wade
- Center for Translational Injury Research, Department of Surgery, University of Texas Health Science Center, Houston, TX, USA
| | - Pär I. Johansson
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Denmark
- Center for Translational Injury Research, Department of Surgery, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
17
|
Lv J, Yi Y, Qi Y, Yan C, Jin W, Meng L, Zhang D, Jiang W. Mitochondrial homeostasis regulates definitive endoderm differentiation of human pluripotent stem cells. Cell Death Discov 2022; 8:69. [PMID: 35177589 PMCID: PMC8854419 DOI: 10.1038/s41420-022-00867-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/21/2022] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular organelles play fundamental roles in almost all cell behaviors. Mitochondria have been reported to be functionally linked to various biological processes, including reprogramming and pluripotency maintenance. However, very little about the role of mitochondria has been revealed in human early development and lineage specification. Here, we reported the characteristics and function of mitochondria during human definitive endoderm differentiation. Using a well-established differentiation system, we first investigated the change of mitochondrial morphology by comparing undifferentiated pluripotent stem cells, the intermediate mesendoderm cells, and differentiated endoderm cells, and found that mitochondria were gradually elongated and matured along differentiation. We further analyzed the expression pattern of mitochondria-related genes by RNA-seq, indicating that mitochondria became active during differentiation. Supporting this notion, the production of adenosine triphosphate (ATP) and reactive oxygen species (ROS) was increased as well. Functionally, we utilized chemicals and genome editing techniques, which could interfere with mitochondrial homeostasis, to determine the role of mitochondria in human endoderm differentiation. Treatment with mitochondrial inhibitors, or genetic depletion of mitochondrial transcription factor A (TFAM), significantly reduced the differentiation efficiency of definitive endoderm. In addition, the defect in endoderm differentiation due to dysfunctional mitochondria could be restored to some extent by the addition of ATP. Moreover, the clearance of excessive ROS due to dysfunctional mitochondria by N-acetylcysteine (NAC) improved the differentiation as well. We further found that ATP and NAC could partially replace the growth factor activin A for definitive endoderm differentiation. Our study illustrates the essential role of mitochondria during human endoderm differentiation through providing ATP and regulating ROS levels, which may provide new insight for metabolic regulation of cell fate determination.
Collapse
Affiliation(s)
- Jing Lv
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Ying Yi
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yan Qi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Chenchao Yan
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Wenwen Jin
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Liming Meng
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China.
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
- Human Genetics Resource Preservation Center of Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
18
|
Sharma S, Agnihotri N, Kumar S. Targeting fuel pocket of cancer cell metabolism: A focus on glutaminolysis. Biochem Pharmacol 2022; 198:114943. [DOI: 10.1016/j.bcp.2022.114943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022]
|
19
|
Xia L, Zhang H, Wang X, Zhang X, Nie K. The Role of Succinic Acid Metabolism in Ovarian Cancer. Front Oncol 2021; 11:769196. [PMID: 34796116 PMCID: PMC8593202 DOI: 10.3389/fonc.2021.769196] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/15/2021] [Indexed: 11/19/2022] Open
Abstract
Ovarian cancer is one of the most common malignancies and the highest mortality among gynecological malignancy. The standard therapy options for patients with ovarian cancer are cytoreductive surgery and chemotherapy, and although most patients do better with standard treatment, it is easy to relapse and be resistant to chemotherapy. Therefore, it is important to find new therapeutic strategies. More recently, metabolic reprogramming has been recognized as a hallmark of cancer and has become a potential target for tumor therapy. Mutations of metabolic enzymes are closely related to the development of ovarian cancer. The metabolic reprogramming of ovarian cancer not only provides energy to tumor cells, but also participates in various biological processes as signaling molecules. Succinic acid (SA) is an important metabolic intermediate involved in a number of metabolic pathways, such as TCA cycle and glutamine metabolism, and is also widely present in a variety of plants and vegetables. Studies show abnormal SA metabolism in many tumors and affect tumor formation through a variety of mechanisms. But the role of SA in ovarian cancer is less studied. This paper reviews the role of SA and its abnormal metabolic pathway in ovarian cancer.
Collapse
Affiliation(s)
- Lei Xia
- Department of Pathology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hairong Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Third Hospital, Jinan, China
| | - Xuezhen Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaoyu Zhang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ke Nie
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
20
|
Zhou S, Xie J, Yu C, Feng Z, Cheng K, Ma J, Wang Y, Duan C, Zhang Y, Jin B, Yin W, Zhuang R. CD226 deficiency promotes glutaminolysis and alleviates mitochondria damage in vascular endothelial cells under hemorrhagic shock. FASEB J 2021; 35:e21998. [PMID: 34669985 DOI: 10.1096/fj.202101134r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/23/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022]
Abstract
Hemorrhagic shock (HS) is common in clinical emergencies, leading to millions of deaths each year globally. CD226 is a costimulatory adhesion molecule expressed on both immune cells and endothelial cells (ECs) to regulate their metabolic activity and function. As endothelial dysfunction occurs after HS, the roles CD226 plays in vascular EC metabolism were investigated. CD226fl/fl Tekcre mice were adopted to achieve vascular EC-specific knockout of CD226, and subjected to HS modelling. Serum levels of crucial intermediate metabolites were evaluated through liquid chromatography-mass spectrometry analysis. Human umbilical vein ECs (HUVECs) were used to study the effects of CD226 under hypoxia in vitro. Seahorse analysis evaluated the cellular glycolysis and mitochondria bioenergetics. Results showed that CD226 deficiency in vascular ECs alleviated HS-induced intestinal damage and inflammatory response in mice. Animal studies indicated an improved energy metabolism when CD226 was knocked out in ECs after HS, as evidenced by enhanced glutamine-glutamate metabolism and decreased lactic acid levels. Glut-1 was upregulated in mouse vascular ECs after HS and HUVECs under hypoxia, combined with decreased CD226. Moreover, HUVECs with CD226 knockdown exhibited relieved mitochondrial damage and early apoptosis under hypoxia, whereas CD226 overexpression showed opposite effects. Seahorse analysis showed that downregulated CD226 significantly increased mitochondrial ATP production and glucose uptake in HUVECs under hypoxia. Additionally, Erk/PHD2 signaling-mediated HIF-1α/Glut-1 and HIF-2α/ASCT2 pathways were involved in CD226 regulation on HUVEC glutaminolysis after hypoxia. Hence, CD226 deficiency promotes bypass energy supply to vascular ECs under ischemic or hypoxic stress, to ameliorate the stress-mediated metabolic disturbance.
Collapse
Affiliation(s)
- Shangxun Zhou
- Department of Immunology, Fourth Military Medical University, Xi'an, China.,Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiangang Xie
- Department of Immunology, Fourth Military Medical University, Xi'an, China.,Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chaoping Yu
- Department of Immunology, Fourth Military Medical University, Xi'an, China.,Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhusheng Feng
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kun Cheng
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Jingchang Ma
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Yuling Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Chujun Duan
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuan Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Boquan Jin
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Wen Yin
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi'an, China.,Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
21
|
Vigneshwar NG, Moore HB, Moore EE. Trauma-Induced Coagulopathy: Diagnosis and Management in 2020. CURRENT ANESTHESIOLOGY REPORTS 2021. [DOI: 10.1007/s40140-021-00438-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
22
|
Mark N, Lyubin A, Gerasi R, Ofir D, Tsur AM, Chen J, Bader T. Comparison of the Effects of Motion and Environment Conditions on Accuracy of Handheld and Finger-Based Pulse Oximeters. Mil Med 2021; 186:465-472. [PMID: 33499470 DOI: 10.1093/milmed/usaa314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/23/2020] [Accepted: 09/14/2020] [Indexed: 11/14/2022] Open
Abstract
INTRODUCTION The most common cause of preventable death on the battlefield is significant blood loss, eventually causing decrease in tissue oxygen delivery. Pulse oximeters (POs) are widely used by the Israeli Defense Forces to obtain fast and noninvasive information about peripheral oxygen saturation (SpO2). However, POs are produced by different manufacturers and therefore include different sensors and are based on distinctive algorithms. This makes them susceptible to different errors caused by factors varying from environmental conditions to the severity of injury. The objectives of this study were to compare the reliability of different devices and their accuracy under various conditions. MATERIAL AND METHODS Six POs underwent performance analysis. The finger-based category included: MightySat by Masimo, Onyx II by Nonin, and CMS50D by Contec. The handheld category comprised: RAD5 by Masimo, 9847 model by Nonin, and 3301 model by BCI. Several environmental and physiological parameters were altered using the ProSim8 simulator by Fluke biomedical, forming unique test cases under which the devices were tested in stationary and motion conditions. RESULTS All finger-based POs showed higher error rates of PO SpO2 and heart rate measurements in motion conditions, regardless of the manufacturer. However, newer devices in the handheld category were not affected. Results presented in Phase II showed that the SpO2 measurement error in all the devices was affected by pigmentation. However, the CMS50D, considered a low-cost device, had a significantly higher error size than other devices. In the devices that were influenced both by pigmentation and the finger cleanliness factors, the combined detected error size was clinically significant. The pigmentation, ambient light, and finger cleanliness also had a significant effect on the heart rate measurement in the CMS50D model, unlike the handheld devices, which were not affected. During Phase II, neither the Nonin nor the Masimo devices were deemed to have a significant advantage. CONCLUSION Considering measurement limitations of POs used is extremely important. Use of handheld devices should be favored for use in motion conditions. Technologically advanced and/or recently developed devices should be preferred because of evolving algorithms, which decrease or eliminate the error factors. The "dirty finger" effect on the measurement error cannot be neglected and therefore the action of finger cleaning should be considered part of the treatment protocol.
Collapse
Affiliation(s)
- Noy Mark
- Surgeon General's Headquarters, Israel Defense Forces Medical Corps, Military POB 02149, Tel Hashomer, Ramat Gan, Israel, Military Postal Code 01215
| | - Anat Lyubin
- Surgeon General's Headquarters, Israel Defense Forces Medical Corps, Military POB 02149, Tel Hashomer, Ramat Gan, Israel, Military Postal Code 01215
| | - Refael Gerasi
- Surgeon General's Headquarters, Israel Defense Forces Medical Corps, Military POB 02149, Tel Hashomer, Ramat Gan, Israel, Military Postal Code 01215
| | - Dror Ofir
- Israel Naval Medical Institute, Israel Defense Forces Medical Corps, Box 22, Rambam Health Care Campus, POB 9602, Haifa, Israel, Postal Code 3109601
| | - Avishai M Tsur
- Surgeon General's Headquarters, Israel Defense Forces Medical Corps, Military POB 02149, Tel Hashomer, Ramat Gan, Israel, Military Postal Code 01215
| | - Jacob Chen
- Surgeon General's Headquarters, Israel Defense Forces Medical Corps, Military POB 02149, Tel Hashomer, Ramat Gan, Israel, Military Postal Code 01215
| | - Tarif Bader
- Surgeon General's Headquarters, Israel Defense Forces Medical Corps, Military POB 02149, Tel Hashomer, Ramat Gan, Israel, Military Postal Code 01215.,Faculty of Medicine, Institute for Research in Military Medicine, The Hebrew University of Jerusalem and Israel Defense Forces Medical Corps, POB 12272, Jerusalem, Israel, Postal Code 9112102
| |
Collapse
|
23
|
Cyr A, Zhong Y, Reis SE, Namas RA, Amoscato A, Zuckerbraun B, Sperry J, Zamora R, Vodovotz Y, Billiar TR. Analysis of the Plasma Metabolome after Trauma, Novel Circulating Sphingolipid Signatures, and In-Hospital Outcomes. J Am Coll Surg 2021; 232:276-287.e1. [PMID: 33453380 PMCID: PMC11875205 DOI: 10.1016/j.jamcollsurg.2020.12.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Trauma is the leading cause of death and disability for individuals under age 55. Many severely injured trauma patients experience complicated clinical courses despite appropriate initial therapy. We sought to identify novel circulating metabolomic signatures associated with clinical outcomes following trauma. STUDY DESIGN Untargeted metabolomics and circulating plasma immune mediator analysis was performed on plasma collected during 3 post-injury time periods (<6 hours [h], 6 h-24h, day 2-day 5) in critically ill trauma patients enrolled between April 2004 and May 2013 at UPMC Presbyterian Hospital in Pittsburgh, PA. Inclusion criteria were age ≥ 18 years, blunt mechanism, ICU admission, and expected survival ≥ 24 h. Exclusion criteria were isolated head injury, spinal cord injury, and pregnancy. Exploratory endpoints included length of stay (overall and ICU), ventilator requirements, nosocomial infection, and Marshall organ dysfunction (MOD) score. The top 50 metabolites were isolated using repeated measures ANOVA and multivariate empirical Bayesian analysis for further study. RESULTS Eighty-six patients were included for analysis. Sphingolipids were enriched significantly (chi-square, p < 10-6) among the top 50 metabolites. Clustering of sphingolipid patterns identified 3 patient subclasses: nonresponders (no time-dependent change in sphingolipids, n = 41), sphingosine/sphinganine-enhanced (n = 24), and glycosphingolipid-enhanced (n = 21). Compared with the sphingolipid-enhanced subclasses, nonresponders had longer mean length of stay, more ventilator days, higher MOD scores, and higher circulating levels of proinflammatory immune mediators IL-6, IL-8, IL-10, MCP1/CCL2, IP10/CXCL10, and MIG/CXCL9 (all p < 0.05), despite similar Injury Severity Scores (p = 0.12). CONCLUSIONS Metabolomic analysis identified broad alterations in circulating plasma sphingolipids after blunt trauma. Circulating sphingolipid signatures and their association with both clinical outcomes and circulating inflammatory mediators suggest a possible link between sphingolipid metabolism and the immune response to trauma.
Collapse
Affiliation(s)
- Anthony Cyr
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Yanjun Zhong
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA; Critical Care, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Steven E Reis
- Clinical and Translational Science Institute and Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Rami A Namas
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Andrew Amoscato
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | | | - Jason Sperry
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA; Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA; Clinical and Translational Science Institute and Department of Medicine, University of Pittsburgh, Pittsburgh, PA; Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA; Clinical and Translational Science Institute and Department of Medicine, University of Pittsburgh, Pittsburgh, PA.
| |
Collapse
|
24
|
Xu P, Xu W, Gao S, Che Y, Zhao P, Lu S, Wang Y, Chen X, Zhang Q, Chen J, Zheng X, Huang G, He G, Liu X. Global metabolic profiling of hemorrhagic shock and resuscitation. Biomed Chromatogr 2021; 35:e5044. [PMID: 33283298 DOI: 10.1002/bmc.5044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 11/11/2022]
Abstract
Hemorrhagic shock (HS) is a medical emergency during trauma. Significant loss of tissue perfusion may result in cellular hypoxia, organ damage and death. The primary treatment of HS is control of the source of bleeding as soon as possible and fluid replacement (crystalloid solutions and blood transfusion). Metabolomics can identify novel biomarkers for various functional and organic diseases. Therefore, systematic exploration of the biological mechanisms of HS and blood transfusion enables the optimization of treatments for HS to reduce the occurrence of organ damage. In this study, a global metabolic profiling strategy is applied to evaluate metabolic changes in the HS rat model. A serum metabolic network with 58 significant metabolites was constructed for HS and resuscitation. Our investigation will offer insights into the pathogenesis.
Collapse
Affiliation(s)
- Ping Xu
- Clinical Research Centre, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
| | - Wei Xu
- Clinical Research Centre, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
| | - Songyan Gao
- Institute of Translation Medicine, Shanghai University, Shanghai, China.,Luodian Clinical Drug Research Centre, Institute for Translational Medicine Research, Shanghai University, Shanghai, China
| | - Yanting Che
- Institute of Translation Medicine, Shanghai University, Shanghai, China.,Luodian Clinical Drug Research Centre, Institute for Translational Medicine Research, Shanghai University, Shanghai, China
| | - Ping Zhao
- Clinical Research Centre, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
| | - Shengfang Lu
- Clinical Research Centre, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
| | - Yong Wang
- Clinical Research Centre, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
| | - Xiaoqing Chen
- Institute of Translation Medicine, Shanghai University, Shanghai, China.,Luodian Clinical Drug Research Centre, Institute for Translational Medicine Research, Shanghai University, Shanghai, China
| | - Qun Zhang
- Baoshan Geriatric Nursing Hospital, Shanghai, China
| | - Junjie Chen
- Institute of Translation Medicine, Shanghai University, Shanghai, China.,Luodian Clinical Drug Research Centre, Institute for Translational Medicine Research, Shanghai University, Shanghai, China
| | | | - Gang Huang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guibing He
- Clinical Research Centre, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
| | - Xinru Liu
- Institute of Translation Medicine, Shanghai University, Shanghai, China.,Luodian Clinical Drug Research Centre, Institute for Translational Medicine Research, Shanghai University, Shanghai, China
| |
Collapse
|
25
|
Clendenen N, Nunns GR, Moore EE, Gonzalez E, Chapman M, Reisz JA, Peltz E, Fragoso M, Nemkov T, Wither MJ, Sauaia A, Silliman CC, Hansen K, Banerjee A, D‘Alessandro A, Moore HB. Selective organ ischaemia/reperfusion identifies liver as the key driver of the post-injury plasma metabolome derangements. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2019; 17:347-356. [PMID: 30747701 PMCID: PMC6774928 DOI: 10.2450/2018.0188-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 11/20/2018] [Indexed: 04/17/2023]
Abstract
BACKGROUND Understanding the molecular mechanisms in perturbation of the metabolome following ischaemia and reperfusion is critical in developing novel therapeutic strategies to prevent the sequelae of post-injury shock. While the metabolic substrates fueling these alterations have been defined, the relative contribution of specific organs to the systemic metabolic reprogramming secondary to ischaemic or haemorrhagic hypoxia remains unclear. MATERIALS AND METHODS A porcine model of selected organ ischaemia was employed to investigate the relative contribution of liver, kidney, spleen and small bowel ischaemia/reperfusion to the plasma metabolic phenotype, as gleaned through ultra-high performance liquid chromatography-mass spectrometry-based metabolomics. RESULTS Liver ischaemia/reperfusion promotes glycaemia, with increases in circulating carboxylic acid anions and purine oxidation metabolites, suggesting that this organ is the dominant contributor to the accumulation of these metabolites in response to ischaemic hypoxia. Succinate, in particular, accumulates selectively in response to the hepatic ischemia, with levels 6.5 times spleen, 8.2 times small bowel, and 6 times renal levels. Similar trends, but lower fold-change increase in comparison to baseline values, were observed upon ischaemia/reperfusion of kidney, spleen and small bowel. DISCUSSION These observations suggest that the liver may play a critical role in mediating the accumulation of the same metabolites in response to haemorrhagic hypoxia, especially with respect to succinate, a metabolite that has been increasingly implicated in the coagulopathy and pro-inflammatory sequelae of ischaemic and haemorrhagic shock.
Collapse
Affiliation(s)
- Nathan Clendenen
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO
| | | | - Ernest E. Moore
- Department of Surgery, University of Colorado Denver, Aurora, CO
- Denver Health Medical Center, Denver, CO
| | - Eduardo Gonzalez
- Department of Surgery, University of Colorado Denver, Aurora, CO
| | - Michael Chapman
- Department of Surgery, University of Colorado Denver, Aurora, CO
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO
| | - Erik Peltz
- Department of Surgery, University of Colorado Denver, Aurora, CO
| | | | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO
| | - Matthew J. Wither
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO
| | - Angela Sauaia
- Department of Surgery, University of Colorado Denver, Aurora, CO
| | | | - Kirk Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO
| | - Anirban Banerjee
- Department of Surgery, University of Colorado Denver, Aurora, CO
| | - Angelo D‘Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO
| | - Hunter B. Moore
- Department of Surgery, University of Colorado Denver, Aurora, CO
| |
Collapse
|
26
|
Gehrke S, Rice S, Stefanoni D, Wilkerson RB, Nemkov T, Reisz JA, Hansen KC, Lucas A, Cabrales P, Drew K, D'Alessandro A. Red Blood Cell Metabolic Responses to Torpor and Arousal in the Hibernator Arctic Ground Squirrel. J Proteome Res 2019; 18:1827-1841. [PMID: 30793910 DOI: 10.1021/acs.jproteome.9b00018] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Arctic ground squirrels provide a unique model to investigate metabolic responses to hibernation in mammals. During winter months these rodents are exposed to severe hypothermia, prolonged fasting, and hypoxemia. In the light of their role in oxygen transport/off-loading and owing to the absence of nuclei and organelles (and thus de novo protein synthesis capacity), mature red blood cells have evolved metabolic programs to counteract physiological or pathological hypoxemia. However, red blood cell metabolism in hibernation has not yet been investigated. Here we employed targeted and untargeted metabolomics approaches to investigate erythrocyte metabolism during entrance to torpor to arousal, with a high resolution of the intermediate time points. We report that torpor and arousal promote metabolism through glycolysis and pentose phosphate pathway, respectively, consistent with previous models of oxygen-dependent metabolic modulation in mature erythrocytes. Erythrocytes from hibernating squirrels showed up to 100-fold lower levels of biomarkers of reperfusion injury, such as the pro-inflammatory dicarboxylate succinate. Altered tryptophan metabolism during torpor was here correlated to the accumulation of potentially neurotoxic catabolites kynurenine, quinolinate, and picolinate. Arousal was accompanied by alterations of sulfur metabolism, including sudden spikes in a metabolite putatively identified as thiorphan (level 1 confidence)-a potent inhibitor of several metalloproteases that play a crucial role in nociception and inflammatory complication to reperfusion secondary to ischemia or hemorrhage. Preliminary studies in rats showed that intravenous injection of thiorphan prior to resuscitation mitigates metabolic and cytokine markers of reperfusion injury, etiological contributors to inflammatory complications after shock.
Collapse
Affiliation(s)
- Sarah Gehrke
- Department of Biochemistry and Molecular Genetics , University of Colorado Denver - Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Sarah Rice
- Department of Chemistry and Biochemistry , University of Alaska Fairbanks , Fairbanks , Alaska 99775 , United States
| | - Davide Stefanoni
- Department of Biochemistry and Molecular Genetics , University of Colorado Denver - Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Rebecca B Wilkerson
- Department of Biochemistry and Molecular Genetics , University of Colorado Denver - Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics , University of Colorado Denver - Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics , University of Colorado Denver - Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics , University of Colorado Denver - Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| | - Alfredo Lucas
- Department of Bioengineering , University of California San Diego , La Jolla , California 92093 , United States
| | - Pedro Cabrales
- Department of Bioengineering , University of California San Diego , La Jolla , California 92093 , United States
| | - Kelly Drew
- Department of Chemistry and Biochemistry , University of Alaska Fairbanks , Fairbanks , Alaska 99775 , United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics , University of Colorado Denver - Anschutz Medical Campus , Aurora , Colorado 80045 , United States
| |
Collapse
|
27
|
D'Alessandro A, El Kasmi KC, Plecitá-Hlavatá L, Ježek P, Li M, Zhang H, Gupte SA, Stenmark KR. Hallmarks of Pulmonary Hypertension: Mesenchymal and Inflammatory Cell Metabolic Reprogramming. Antioxid Redox Signal 2018; 28. [PMID: 28637353 PMCID: PMC5737722 DOI: 10.1089/ars.2017.7217] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE The molecular events that promote the development of pulmonary hypertension (PH) are complex and incompletely understood. The complex interplay between the pulmonary vasculature and its immediate microenvironment involving cells of immune system (i.e., macrophages) promotes a persistent inflammatory state, pathological angiogenesis, and fibrosis that are driven by metabolic reprogramming of mesenchymal and immune cells. Recent Advancements: Consistent with previous findings in the field of cancer metabolism, increased glycolytic rates, incomplete glucose and glutamine oxidation to support anabolism and anaplerosis, altered lipid synthesis/oxidation ratios, increased one-carbon metabolism, and activation of the pentose phosphate pathway to support nucleoside synthesis are but some of the key metabolic signatures of vascular cells in PH. In addition, metabolic reprogramming of macrophages is observed in PH and is characterized by distinct features, such as the induction of specific activation or polarization states that enable their participation in the vascular remodeling process. CRITICAL ISSUES Accumulation of reducing equivalents, such as NAD(P)H in PH cells, also contributes to their altered phenotype both directly and indirectly by regulating the activity of the transcriptional co-repressor C-terminal-binding protein 1 to control the proliferative/inflammatory gene expression in resident and immune cells. Further, similar to the role of anomalous metabolism in mitochondria in cancer, in PH short-term hypoxia-dependent and long-term hypoxia-independent alterations of mitochondrial activity, in the absence of genetic mutation of key mitochondrial enzymes, have been observed and explored as potential therapeutic targets. FUTURE DIRECTIONS For the foreseeable future, short- and long-term metabolic reprogramming will become a candidate druggable target in the treatment of PH. Antioxid. Redox Signal. 28, 230-250.
Collapse
Affiliation(s)
- Angelo D'Alessandro
- 1 Department of Biochemistry and Molecular Genetics, University of Colorado - Denver , Colorado
| | - Karim C El Kasmi
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado.,3 Department of Pediatric Gastroenterology, University of Colorado - Denver , Colorado
| | - Lydie Plecitá-Hlavatá
- 4 Department of Mitochondrial Physiology, Institute of Physiology , Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Ježek
- 4 Department of Mitochondrial Physiology, Institute of Physiology , Czech Academy of Sciences, Prague, Czech Republic
| | - Min Li
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| | - Hui Zhang
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| | - Sachin A Gupte
- 5 Department of Pharmacology, School of Medicine, New York Medical College , Valhalla, New York
| | - Kurt R Stenmark
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| |
Collapse
|
28
|
Clendenen N, Nunns GR, Moore EE, Reisz JA, Gonzalez E, Peltz E, Silliman CC, Fragoso M, Nemkov T, Wither MJ, Hansen K, Banerjee A, Moore HB, D’Alessandro A. Hemorrhagic shock and tissue injury drive distinct plasma metabolome derangements in swine. J Trauma Acute Care Surg 2017; 83:635-642. [PMID: 28463938 PMCID: PMC5608631 DOI: 10.1097/ta.0000000000001504] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Tissue injury and hemorrhagic shock induce significant systemic metabolic reprogramming in animal models and critically injured patients. Recent expansions of the classic concepts of metabolomic aberrations in tissue injury and hemorrhage opened the way for novel resuscitative interventions based on the observed abnormal metabolic demands. We hypothesize that metabolic demands and resulting metabolic signatures in pig plasma will vary in response to isolated or combined tissue injury and hemorrhagic shock. METHODS A total of 20 pigs underwent either isolated tissue injury, hemorrhagic shock, or combined tissue injury and hemorrhagic shock referenced to a sham protocol (n = 5/group). Plasma samples were analyzed by UHPLC-MS. RESULTS Hemorrhagic shock promoted a hypermetabolic state. Tissue injury alone dampened metabolic responses in comparison to sham and hemorrhagic shock, and attenuated the hypermetabolic state triggered by shock with respect to energy metabolism (glycolysis, glutaminolysis, and Krebs cycle). Tissue injury and hemorrhagic shock had a more pronounced effect on nitrogen metabolism (arginine, polyamines, and purine metabolism) than hemorrhagic shock alone. CONCLUSION Isolated or combined tissue injury and hemorrhagic shock result in distinct plasma metabolic signatures. These findings indicate that optimized resuscitative interventions in critically ill patients are possible based on identifying the severity of tissue injury and hemorrhage.
Collapse
Affiliation(s)
- Nathan Clendenen
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO, USA
| | - Geoffrey R Nunns
- Department of Surgery - University of Colorado Denver, Aurora, CO, USA
| | | | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Eduardo Gonzalez
- Department of Surgery - University of Colorado Denver, Aurora, CO, USA
| | - Erik Peltz
- Department of Surgery - University of Colorado Denver, Aurora, CO, USA
| | - Christopher C Silliman
- Department of Surgery - University of Colorado Denver, Aurora, CO, USA
- Bonfils Blood Center, Denver, CO, USA
- Department of Pediatrics, University of Colorado Denver, Aurora, CO, USA
| | - Miguel Fragoso
- Department of Surgery - University of Colorado Denver, Aurora, CO, USA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Matthew J Wither
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | | | - Anirban Banerjee
- Department of Surgery - University of Colorado Denver, Aurora, CO, USA
| | - Hunter B Moore
- Department of Surgery - University of Colorado Denver, Aurora, CO, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
29
|
C-peptide attenuates acute lung inflammation in a murine model of hemorrhagic shock and resuscitation by reducing gut injury. J Trauma Acute Care Surg 2017; 83:256-262. [PMID: 28452895 DOI: 10.1097/ta.0000000000001539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The study aims to evaluate whether C-peptide can reduce gut injury during hemorrhagic shock (HS) and resuscitation (R) therefore attenuate shock-induced inflammation and subsequent acute lung injury. METHODS Twelve-week-old male mice (C57/BL6) were hemorrhaged (mean arterial blood pressure maintained at 35 mm Hg for 60 minutes) and then resuscitated with Ringer's lactate, followed by red blood cell transfusion with (HS/R) or without C-peptide (HS/R + C-peptide). Mouse gut permeability, bacterial translocation into the circulatory system and intestinal pathology, circulating HMGB1, and acute lung injury were assessed at different times after R. The mice in the control group underwent sham procedures without HS. RESULTS Compared to the sham group, the mice in the HS/R group showed increased gut permeability (6.07 ± 3.41 μg of FD4/mL) and bacterial translocation into the circulatory system (10.05 ± 4.92, lipopolysaccharide [LPS] of pg/mL), and increased gut damage; conversely, mice in the HS/R + C-peptide group showed significantly reduced gut permeability (1.59 ± 1.39 μg of FD4/mL; p < 0.05) and bacterial translocation (4.53 ± 1.08 pg of LPS/mL; p < 0.05) with reduced intestine damage. In addition, mice in the HS/R group had increased circulating HMGB1 (21.64 ± 14.17 ng/mL), lung myeloperoxidase) activity (34.4 ± 8.91 mU/g of tissue), and pulmonary protein leakage (2.33 ± 1.16 μg Evans blue/g tissue per minute). Mice in the HS/R + C-peptide group showed decreased HMGB1 (7.27 ± 1.93 ng/mL; p < 0.05), lung myeloperoxidase (23.73 ± 8.39 mU/g of tissue; p < 0.05), and pulmonary protein leakage (1.17 ± 0.42 Evans Blue/g tissue per minute; p < 0.05). CONCLUSION Our results indicate that C-peptide exerts beneficial effects to attenuate gut injury and dysfunction, therefore diminishing lung inflammation and subsequent injury in mice with HS and R.
Collapse
|
30
|
Clendenen N, Weitzel N. Predictors of Prolonged Mechanical Ventilation in Adults After Acute Type-A Aortic Dissection Repair-Implications for the Future. J Cardiothorac Vasc Anesth 2017; 31:1562-1563. [PMID: 28864159 DOI: 10.1053/j.jvca.2017.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Nathan Clendenen
- Department of Anesthesiology University of Colorado Denver School of Medicine Aurora, CO
| | - Nathaen Weitzel
- Department of Anesthesiology University of Colorado Denver School of Medicine Aurora, CO
| |
Collapse
|
31
|
Reisz JA, D'Alessandro A. Measurement of metabolic fluxes using stable isotope tracers in whole animals and human patients. Curr Opin Clin Nutr Metab Care 2017; 20:366-374. [PMID: 28768294 PMCID: PMC5794022 DOI: 10.1097/mco.0000000000000393] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Metabolic flux analysis using stable isotope labeled substrates allows for the tracing of carbon, nitrogen, and hydrogen atoms through metabolic pathways and is an invaluable tool for investigating dynamic metabolic changes occurring in health and disease. Studies of flux analysis in vivo are more technically challenging than in vitro or ex vivo but provide a highly detailed view of organ and/or systemic metabolism. We review here recent efforts in studies of diet and nutrition, non-small cell lung cancer, ischemia/reperfusion injury, and hemorrhagic shock where in vivo flux analysis was utilized to analyze metabolic modulation. RECENT FINDINGS Recent technical strides in the field of metabolomics afford sensitive and quantitative in vivo measurements of metabolic fluxes. Stable isotope tracing with C-glucose, C, N-glutamine, C-propionate, and other substrates are used in combination or in parallel to investigate the interplays among central carbon metabolic pathways and many other areas of the metabolome. SUMMARY Stable isotope tracing in vivo provides opportunities to investigate physiological processes in the context of the whole animal. These approaches, often NMR spectroscopy or mass spectrometry (MS)-based, are growing in use and will likely find key applications in studying systemic disease, sports physiology, cancer metabolism, and personalized medicine.
Collapse
Affiliation(s)
- Julie A Reisz
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | | |
Collapse
|
32
|
D’ALESSANDRO A, MOORE HB, MOORE EE, REISZ JA, WITHER MJ, GHASABYAN A, CHANDLER J, SILLIMAN CC, HANSEN KC, BANERJEE A. Plasma succinate is a predictor of mortality in critically injured patients. J Trauma Acute Care Surg 2017; 83:491-495. [PMID: 28590356 PMCID: PMC5573618 DOI: 10.1097/ta.0000000000001565] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Trauma is the leading cause of mortality under the age of 40 years. Recent observations on metabolic reprogramming during hypoxia and ischemia indicate that hypoxic mitochondrial uncoupling promotes the generation of succinate, which in turn mediates reperfusion injury and inflammatory sequelae upon reoxygenation. Plasma levels of succinate significantly increase in response to trauma and hemorrhage in experimental models and clinical samples, suggesting that succinate may represent a candidate marker of systemic perfusion in trauma. METHODS Quantitative mass spectrometry-based metabolomics was used to quantify succinate and lactate in 595 plasma samples from severely injured patients enrolled at the Denver Health Medical Center, a Level I trauma center in Denver, Colorado. RESULTS A total of 95 severely injured patients were sampled for up to 10 time points (595 total samples), from field blood to 7 days postinjury. Results indicate that plasma levels of succinate increased up to 25.9-fold in deceased patients versus the median of the surviving patients (p = 2.75e-100; receiver operating characteristic area under the curve, 0.911). On the other hand, only 2.4-fold changes increases in lactate were observed (p = 5.8e-21; area under the curve, 0.874). CONCLUSION Succinate represents a uniquely sensitive biomarker of postshock metabolic derangement and may be an important mediator of sequelae. LEVEL OF EVIDENCE Prognostic study, level III.
Collapse
Affiliation(s)
- Angelo D’ALESSANDRO
- Department of Biochemistry and Molecular Genetics, University of
Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | | | - Ernest E. MOORE
- Denver Health Hospital – Denver, CO, USA
- Department of Surgery, University of Colorado Denver –
Anschutz Medical Campus, Aurora, CO, USA
| | - Julie A. REISZ
- Department of Biochemistry and Molecular Genetics, University of
Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew J. WITHER
- Department of Biochemistry and Molecular Genetics, University of
Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Christopher C. SILLIMAN
- Department of Paediatrics, University of Colorado Denver –
Anschutz Medical Campus, Aurora, CO, USA
- Bonfils Blood Center – Denver, CO, USA
| | - Kirk C. HANSEN
- Department of Biochemistry and Molecular Genetics, University of
Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Anirban BANERJEE
- Department of Surgery, University of Colorado Denver –
Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
33
|
Red blood cells in hemorrhagic shock: a critical role for glutaminolysis in fueling alanine transamination in rats. Blood Adv 2017; 1:1296-1305. [PMID: 29296771 DOI: 10.1182/bloodadvances.2017007187] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/02/2017] [Indexed: 02/06/2023] Open
Abstract
Red blood cells (RBCs) are the most abundant host cell in the human body and play a critical role in oxygen transport and systemic metabolic homeostasis. Hypoxic metabolic reprogramming of RBCs in response to high-altitude hypoxia or anaerobic storage in the blood bank has been extensively described. However, little is known about the RBC metabolism following hemorrhagic shock (HS), the most common preventable cause of death in trauma, the global leading cause of total life-years lost. Metabolomics analyses were performed through ultra-high pressure liquid chromatography-mass spectrometry on RBCs from Sprague-Dawley rats undergoing HS (mean arterial pressure [MAP], <30 mm Hg) in comparison with sham rats (MAP, >80 mm Hg). Steady-state measurements were accompanied by metabolic flux analysis upon tracing of in vivo-injected 13C15N-glutamine or inhibition of glutaminolysis using the anticancer drug CB-839. RBC metabolic phenotypes recapitulated the systemic metabolic reprogramming observed in plasma from the same rodent model. Results indicate that shock RBCs rely on glutamine to fuel glutathione (GSH) synthesis and pyruvate transamination, whereas abrogation of glutaminolysis conferred early mortality and exacerbated lactic acidosis and systemic accumulation of succinate, a predictor of mortality in the military and civilian critically ill populations. Glutamine is here identified as an essential amine group donor in HS RBCs, plasma, liver, and lungs, providing additional rationale for the central role glutaminolysis plays in metabolic reprogramming and survival following severe hemorrhage.
Collapse
|
34
|
Lu Q, Lu Y, Zhang Y, Li Z, Xie X. Establishment and evaluation of rat trauma hemorrhagic liver injury model. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:7340-7349. [PMID: 31966574 PMCID: PMC6965275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/17/2016] [Indexed: 06/10/2023]
Abstract
Trauma hemorrhagic shock is a common and critical disease, which induces multiple organ failure, especially of the liver, when combined with fracture. However, no effective trauma hemorrhagic liver injury model that mimics the real-life condition has been developed so far. This study aims to develop an effective trauma hemorrhagic liver injury model based on a fracture and hemorrhage approach. The levels of the following proteins were determined by the enzyme-linked immunosorbent assay (ELISA) in our fracture and hemorrhage-based model system: serum alanine transaminase (ALT), aspartate aminotransferase (AST), inflammatory cytokines such as interleukin-1β, interleukin-6, and tumor necrosis factor-α, chemokines such as C-C motif ligand 2, C-C motif ligand 5, C-C motif ligand 13, and C-X-C motif ligand 2. Pathological changes in the liver and the numbers of CD45+ cells and polymorphic nuclear neutrophils (PMNs) in the liver parenchyma were analyzed by hematoxylin-eosin staining, periodic acid-Schiff staining, and flow cytometry, respectively. As expected, the serum levels of ALT and AST increased significantly with trauma time and peaked at 16 hrs post-trauma. Similarly, the levels of the inflammatory cytokines also increased significantly with trauma time, and peaked after 8 hrs or 16 hrs of trauma. Analysis of hepatic morphology at the time-point when the trauma was inflicted and at later time-points post-trauma, revealed invasion of inflammatory cells, formation of hyperchromatic nuclei, and presence of loose and irregular acinus and vacuolus; the phenotype was most severe at 16 hrs post-trauma. The number of CD45+ cells and PMNs increased significantly with trauma time and peaked after 16 hrs of trauma. These observations indicated that the trauma hemorrhagic liver injury model was successfully established and that it could provide an effective system to study the mechanisms of trauma hemorrhagic liver injury.
Collapse
Affiliation(s)
- Qingming Lu
- Department of Comprehensive Surgery of Nanlou, Chinese PLA General Hospital Beijing 100853, China
| | - Yan Lu
- Department of Comprehensive Surgery of Nanlou, Chinese PLA General Hospital Beijing 100853, China
| | - Ying Zhang
- Department of Comprehensive Surgery of Nanlou, Chinese PLA General Hospital Beijing 100853, China
| | - Zhaohui Li
- Department of Comprehensive Surgery of Nanlou, Chinese PLA General Hospital Beijing 100853, China
| | - Xiaohua Xie
- Department of Comprehensive Surgery of Nanlou, Chinese PLA General Hospital Beijing 100853, China
| |
Collapse
|