1
|
Chen D, Chen Y, Liu J, Liu X, Liu P, Zhan J, Chen Z, Gan Y, Huang M, Chen Z. In situ protein corona-camouflaged supramolecular assemblies remodel thrombotic microenvironment for improved arterial homeostasis. SCIENCE ADVANCES 2025; 11:eadu6676. [PMID: 40315315 PMCID: PMC12047436 DOI: 10.1126/sciadv.adu6676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
Arterial thrombosis is commonly accompanied by poor recanalization and high recurrence, typically caused by a fibrinolysis-resistant microenvironment. We identify elevated levels of plasminogen activator inhibitor-1 (PAI-1) and, notably, its strong correlation with inflammation in arterial thrombosis. To address this, small molecular inhibitors of PAI-1 and inflammation are used as bioregulators to restore vascular homeostasis. We design a carrier-free supramolecular system based on the bioregulators-tuned self-assembly of a near-infrared thrombus probe, which preferentially forms protein corona in situ to enhance plasma stability. Under acidic conditions and increased shear stress, the supramolecular assemblies disintegrate, enabling site-specific cargo release. In vivo, the probe accumulates 22.8-fold more in the thrombotic than contralateral artery. Functionally, this nanomedicine improves outcomes in mice with carotid artery thrombosis and chronic cerebral ischemia. Mechanistically, it down-regulates NF-κB signaling, inhibits NETosis and glycolysis, and up-regulates cGMP-mediated signaling, thereby alleviating inflammation and promoting fibrinolysis. This study offers an innovative codelivery strategy using supramolecular assemblies to advance therapies for arterial thrombosis.
Collapse
Affiliation(s)
- Dan Chen
- Department of Cardiology, Heart Center of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Yifan Chen
- Department of Cardiology, Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian 361004, China
| | - Jianwen Liu
- Department of Cardiology, Heart Center of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Xinyue Liu
- Department of Cardiology, Heart Center of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Peiwen Liu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Jiabing Zhan
- Department of Cardiology, Heart Center of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Zhiting Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Yong Gan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Zhaoyang Chen
- Department of Cardiology, Heart Center of Fujian Province, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| |
Collapse
|
2
|
Chen Z, Xu L, Yuan Y, Zhang S, Xue R. Metabolic crosstalk between platelets and cancer: Mechanisms, functions, and therapeutic potential. Semin Cancer Biol 2025; 110:65-82. [PMID: 39954752 DOI: 10.1016/j.semcancer.2025.02.001] [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: 12/06/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Platelets, traditionally regarded as passive mediators of hemostasis, are now recognized as pivotal regulators in the tumor microenvironment, establishing metabolic feedback loops with tumor and immune cells. Tumor-derived signals trigger platelet activation, which induces rapid metabolic reprogramming, particularly glycolysis, to support activation-dependent functions such as granule secretion, morphological changes, and aggregation. Beyond self-regulation, platelets influence the metabolic processes of adjacent cells. Through direct mitochondrial transfer, platelets reprogram tumor and immune cells, promoting oxidative phosphorylation. Additionally, platelet-derived cytokines, granules, and extracellular vesicles drive metabolic alterations in immune cells, fostering suppressive phenotypes that facilitate tumor progression. This review examines three critical aspects: (1) the distinctive metabolic features of platelets, particularly under tumor-induced activation; (2) the metabolic crosstalk between activated platelets and other cellular components; and (3) the therapeutic potential of targeting platelet metabolism to disrupt tumor-promoting networks. By elucidating platelet metabolism, this review highlights its essential role in tumor biology and its therapeutic implications.
Collapse
Affiliation(s)
- Zhixue Chen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lin Xu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yejv Yuan
- The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232001, China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
3
|
Gao C, Dai Y, Spezza PA, Boasiako P, Tang A, Rasquinha G, Zhong H, Shao B, Liu Y, Shi PA, Lobo CA, An X, Guo A, Mitchell WB, Manwani D, Yazdanbakhsh K, Mendelson A. Megakaryocytes transfer mitochondria to bone marrow mesenchymal stromal cells to lower platelet activation. J Clin Invest 2025; 135:e189801. [PMID: 40014405 PMCID: PMC11996913 DOI: 10.1172/jci189801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
Newly produced platelets acquire a low activation state, but whether the megakaryocyte plays a role in this outcome has not been fully uncovered. Mesenchymal stem cells (MSCs) were previously shown to promote platelet production and lower platelet activation. We found that healthy megakaryocytes transfer mitochondria to MSCs, which is mediated by connexin 43 (Cx43) gap junctions on MSCs and leads to platelets at a low energetic state with increased LYN activation, characteristic of resting platelets with increased LYN activation, characteristic of resting platelets. On the contrary, MSCs have a limited ability to transfer mitochondria to megakaryocytes. Sickle cell disease (SCD) is characterized by hemolytic anemia and results in heightened platelet activation, contributing to numerous disease complications. Platelets in SCD mice and human samples had a heightened energetic state with increased glycolysis. MSC exposure to heme in SCD led to decreased Cx43 expression and a reduced ability to uptake mitochondria from megakaryocytes. This prevented LYN activation in platelets and contributed to increased platelet activation at steady state. Altogether, our findings demonstrate an effect of hemolysis in the microenvironment leading to increased platelet activation in SCD. These findings have the potential to inspire new therapeutic targets to relieve thrombosis-related complications of SCD and other hemolytic conditions.
Collapse
Affiliation(s)
| | - Yitian Dai
- Laboratory of Stem Cell Biology and Engineering Research
| | - Paul A. Spezza
- Laboratory of Stem Cell Biology and Engineering Research
| | - Paul Boasiako
- Laboratory of Stem Cell Biology and Engineering Research
| | - Alice Tang
- Laboratory of Stem Cell Biology and Engineering Research
| | | | | | - Bojing Shao
- Laboratory of Vascular Inflammation and Thrombosis Research
| | | | | | - Cheryl A. Lobo
- Laboratory of Blood Borne Parasites, New York Blood Center, New York, New York, USA
| | | | - Anqi Guo
- Laboratory of Complement Biology
| | - William B. Mitchell
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children’s Hospital at Montefiore, Bronx, New York, USA
| | - Deepa Manwani
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children’s Hospital at Montefiore, Bronx, New York, USA
| | | | | |
Collapse
|
4
|
Li J, Chen S, Yang S, Zhang W, Huang X, Zhou L, Liu Y, Li M, Guo Y, Yin J, Xu K. Hypercoagulable state and gut microbiota dysbiosis as predictors of poor functional outcomes in acute ischemic stroke patients. mSystems 2025:e0149224. [PMID: 40202300 DOI: 10.1128/msystems.01492-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Stroke is the second leading cause of death worldwide. Acute ischemic stroke (AIS) patients often exhibit hypercoagulable state and gut microbiota dysbiosis. However, the association between coagulation abnormalities and gut microbiota dysbiosis in AIS patients and their predictive value for poor functional outcomes in AIS has not been investigated. Our study enrolled 95 AIS patients and 81 healthy controls, using 16S rRNA sequencing to analyze gut microbiota composition. Baseline fibrinogen level was found to be an independent risk factor for poor functional outcomes at 90-day follow-up (odds ratio = 2.16, 95% confidence interval: 1.02-4.59, P = 0.044). AIS patients showed significant gut microbiota dysbiosis, with significantly increased Parabacteroides and Alistipes, and decreased Prevotella and Roseburia, associated with coagulation indices. Furthermore, compared with AIS patients with normal coagulation function, those in a hypercoagulable state exhibited a significant increase in Alistipes and a decrease in Prevotella. We identified gut microbial biomarkers consisting of 15 bacteria that predicted poor functional outcome in AIS patients at 90-day follow-up. Coagulation indices improved the predictive performance of these biomarkers. In training and validation cohorts, area under the curve (AUC) values were 0.930 and 0.890 for microbial biomarkers alone, 0.691 and 0.751 for coagulation indices alone, and 0.943 and 0.944 for coagulation indices combined with gut microbial biomarkers. Our study showed that AIS patients with hypercoagulable state had gut microbiota dysbiosis, with Alistipes and Prevotella significantly associated with coagulation indices. A classification model based on coagulation indices and gut microbial biomarkers accurately predicted poor functional outcome in AIS patients at 90-day follow-up. IMPORTANCE Acute ischemic stroke (AIS) patients often exhibit hypercoagulable state and gut microbiota dysbiosis. However, the relationship between hypercoagulable state and gut microbiota dysbiosis in AIS patients and their predictive value for poor functional outcomes has not been fully explored. Our study of 95 AIS patients showed that baseline fibrinogen level was an independent risk factor for poor functional outcome at 90-day follow-up in AIS patients. Hypercoagulable state in AIS patients correlates with gut microbiota dysbiosis. AIS patients with hypercoagulable state had increased Alistipes abundance and decreased Prevotella abundance. A classification model based on coagulation indices and gut microbial biomarkers accurately predicted poor functional outcome in AIS patients at 90-day follow-up.
Collapse
Affiliation(s)
- Jie Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shengnan Chen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Siqi Yang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wen Zhang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoqi Huang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lang Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanchao Liu
- Department of Neurosurgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mengxi Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yonghui Guo
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jia Yin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kaiyu Xu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Xu B, Ye X, Sun K, Chen L, Wen Z, Lan Q, Chen J, Chen M, Shen M, Wang S, Xu Y, Zhang X, Zhao J, Wang J, Chen S. IRAP Drives Ribosomal Degradation to Refuel Energy for Platelet Activation during Septic Thrombosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411914. [PMID: 39853919 PMCID: PMC11967848 DOI: 10.1002/advs.202411914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/09/2025] [Indexed: 01/26/2025]
Abstract
Platelets play crucial roles in multiple pathophysiological processes after energy-dependent activation. It is puzzling how such a small cellular debris has abundant energy supply. In this study, it is shown that insulin-regulated aminopeptidase (IRAP), a type II transmembrane protein, is a key regulator for platelet activation by promoting energy regeneration during septic thrombosis. Through interaction with certain endosome membrane proteins, IRAP can not only promote granule release, but also facilitate lysosomal degradation of theoretically discarded ribosomes in an mTORC1- and S-acylation-dependent manner in activated platelets. Plentiful amino acids obtained from IRAP-mediated ribophagy are recruited to aerobic glycolysis and then promote energy metabolism reprogramming, thereby producing abundant energy for platelet life extension and prolonged activation. Consequently, targeted blocking IRAP can dramatically alleviate platelet hyperactivation and septic thrombosis.
Collapse
Affiliation(s)
- Baichuan Xu
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xianpeng Ye
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Kangfu Sun
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Liang Chen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Zhaoyang Wen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Qigang Lan
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jun Chen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Mo Chen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Mingqiang Shen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Song Wang
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Yang Xu
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xi Zhang
- Medical Center of HematologyXinqiao HospitalState Key Laboratory of Trauma and Chemical PoisoningArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jinghong Zhao
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Shilei Chen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| |
Collapse
|
6
|
Wang Y, Xu N, Ndzie Noah ML, Chen L, Zhan X. Pyruvate Kinase M1/2 Proteoformics for Accurate Insights into Energy Metabolism Abnormity to Promote the Overall Management of Ovarian Cancer Towards Predictive, Preventive, and Personalized Medicine Approaches. Metabolites 2025; 15:203. [PMID: 40137167 PMCID: PMC11944880 DOI: 10.3390/metabo15030203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/01/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Ovarian cancer (OC) is a global health problem that frequently presents at advanced stages, is predisposed to recurrence, readily develops resistance to platinum-based drugs, and has a low survival rate. Predictive, preventive, and personalized medicine (PPPM/3PM) offers an integrated solution with the use of genetic, proteomic, and metabolic biomarkers to identify high-risk individuals for early detection. Metabolic reprogramming is one of the key strategies employed by tumor cells to adapt to the microenvironment and support unlimited proliferation. Pyruvate kinases M1 and M2 (PKM1/2) are encoded by the PKM gene, a pivotal enzyme in the last step of the glycolytic pathway, which is at the crossroads of aerobic oxidation and the Warburg effect to serve as a potential regulator of glucose metabolism and influence cellular energy production and metabolic reprogramming. Commonly, the ratio of PKM1-to-PKM2 is changed in tumors compared to normal controls, and PKM2 is highly expressed in OC to induce a high glycolysis rate and participate in the malignant invasion and metastatic characteristics of cancer cells with epithelial/mesenchymal transition (EMT). PKM2 inhibitors suppress the migration and growth of OC cells by interfering with the Warburg effect. Proteoforms are the final structural and functional forms of a gene/protein, and the canonical protein PKM contains all proteoforms encoded by the same PKM gene. The complexity of PKM can be elucidated by proteoformics. The OC-specific PKM proteoform might represent a specific target for therapeutic interventions against OC. In the framework of PPPM/3PM, the OC-specific PKM proteoform might be the early warning and prognosis biomarker. It is important to clarify the molecular mechanisms of PKM proteoforms in cancer metabolism. This review analyzes the expression, function, and molecular mechanisms of PKM proteoforms in OC, which help identify specific biomarkers for OC.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China
- Department of Gynecology, Gaotang County Medical Center, Liaocheng 252800, China
| | - Nuo Xu
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
| | - Marie Louise Ndzie Noah
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
| | - Liang Chen
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China
| | - Xianquan Zhan
- Department of Gynecological Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China; (Y.W.); (N.X.); (M.L.N.N.)
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan 250117, China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China
| |
Collapse
|
7
|
Zhang W, Pei B, Zhou Y, Li H, Ma W, Zhou B, Zhou C, Jiang H, Ji X. Emerging Targets, Novel Directions, and Innovative Approaches in Thrombosis Therapy. Aging Dis 2025:AD.2024.1688. [PMID: 40153578 DOI: 10.14336/ad.2024.1688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/11/2025] [Indexed: 03/30/2025] Open
Abstract
In clinical practice, antiplatelet, anticoagulant and fibrinolytic drugs are the mainstay of thrombosis treatment, but their potential bleeding side effects limit their widespread use. Therefore, modifying these existing drugs or developing new therapies that mitigate bleeding risks while maintaining their efficacy and utilization is necessary. Since the critical role of platelets in thrombosis is closely related to their cell surface receptors, intracellular signaling pathways and metabolism, current research focuses on these three major classes of platelet targets to develop new antithrombotic drugs. The coagulation cascade has always been the main target of anticoagulant drugs, but since the role of molecules of the contact system is more critical in thrombosis than in hemostasis, molecules targeting the contact system, such as FXIa and FXIIa, have become the main direction of anticoagulant drug research at present. Moreover, since the inflammatory response has been found to be significantly associated with thrombosis in recent years, the development of drugs that target inflammatory pathways, such as inflammasome, has also become a hot topic. This article provides a detailed description of these targets or drug formulations that are currently being investigated, including their mode of action and antithrombotic efficiency, and also points out their existing shortcomings. Moreover, antithrombotic nanomedicines can achieve precise release of drugs, which can greatly improve the thrombolytic efficiency and reduce side effects. In conclusion, this review focuses on summarizing the current new targets and new methods of antithrombotic drug research, hoping to provide a little reference for future related research.
Collapse
Affiliation(s)
- Weiyue Zhang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Baoqing Pei
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Hui Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Bing Zhou
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Huimin Jiang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Xunming Ji
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| |
Collapse
|
8
|
Li Z, Chen P, Lin Y, Zhang J, Ding J, Ahmed RZ, Jin X, Zheng Y. Inhibition of platelet activation process upon tris (2-chloroethyl) phosphate exposure: Role of PFKP-mediated glycolysis and the pentose phosphate pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125714. [PMID: 39828204 DOI: 10.1016/j.envpol.2025.125714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Tris (2-chloroethyl) phosphate (TCEP), recognized as an emerging pollutant, has been frequently detected in human blood. Maintenance of blood homeostasis is indispensable for regulating various physiological states and overall health, yet hematological toxicology of TCEP has not been extensively investigated. Platelets, a vital component of blood, are fundamental in the processes of hemostasis and thrombosis through their activation; thus, this study was designed to elucidate the effects and underlying mechanisms of TCEP on platelet activation. Utilizing an in vivo model, we conducted a proteomic analysis of platelets and found that TCEP exposure inhibited platelet activation. An ex vivo platelet evaluation system was employed to further dissect the processes of platelet activation, revealing that TCEP predominantly suppressed platelet aggregation, degranulation and clot retraction. These processes were highly dependent on energy metabolism, and TCEP was found to decrease ATP levels, primarily by impairing glycolysis and pentose phosphate pathways. Subsequent investigation into the molecular mechanisms revealed that TCEP decreased the activity of phosphofructokinase platelet (PFKP) by enhancing O-linked N-acetylglucosamine (O-GlcNAc) transferase interaction with PFKP. This study is the first to uncover the disruptive effects of TCEP on platelet activation process, providing valuable insights into the assessment of hematologic health risks associated with TCEP-like emerging pollutants exposure.
Collapse
Affiliation(s)
- Ziyuan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Pu Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Yongfeng Lin
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Jingxu Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Jian Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | | | - Xiaoting Jin
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China.
| | - Yuxin Zheng
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Yvan-Charvet L, Barouillet T, Borowczyk C. Haematometabolism rewiring in atherosclerotic cardiovascular disease. Nat Rev Cardiol 2025:10.1038/s41569-024-01108-9. [PMID: 39743562 DOI: 10.1038/s41569-024-01108-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 01/04/2025]
Abstract
Atherosclerotic cardiovascular diseases are the most frequent cause of death worldwide. The clinical complications of atherosclerosis are closely linked to the haematopoietic and immune systems, which maintain homeostatic functions and vital processes in the body. The nodes linking metabolism and inflammation are receiving increasing attention because they are inextricably linked to inflammatory manifestations of non-communicable diseases, including atherosclerosis. Although metabolism and inflammation are essential to survival and involve all tissues, we still know little about how these processes influence each other. In an effort to understand these mechanisms, in this Review we explore whether and how potent cardiovascular risk factors and metabolic modifiers of atherosclerosis influence the molecular and cellular machinery of 'haematometabolism' (metabolic-dependent haematopoietic stem cell skewing) and 'efferotabolism' (metabolic-dependent efferocyte reprogramming). These changes might ultimately propagate a quantitative and qualitative drift of the macrophage supply chain and affect the clinical manifestations of atherosclerosis. Refining our understanding of the different metabolic requirements of these processes could open the possibility of developing therapeutics targeting haematometabolism that, in conjunction with improved dietary habits, help rebalance and promote efficient haematopoiesis and efferocytosis and decrease the risk of atherosclerosis complications.
Collapse
Affiliation(s)
- Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| | - Thibault Barouillet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | - Coraline Borowczyk
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| |
Collapse
|
10
|
Yin L, Mao L, Yin R, Lv C, Shi X, Yue C, Chen Y, Lu C, Wu Z, Xu K, Cao W. ACE Loss Drives Renal Cell Carcinoma Growth and Invasion by Modulating AKT-FOXO1. Biologics 2024; 18:397-412. [PMID: 39717370 PMCID: PMC11665188 DOI: 10.2147/btt.s485178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Purpose Emerging literature links the role of the renin-angiotensin-aldosterone system (RAAS) to the progression of cancers. However, the function of RAAS has not been verified in Clear-cell renal cell carcinoma (ccRCC). Methods ACE expression in ccRCC tissues was determined using RT-PCR, Western blot, and immunohistochemistry staining. The clinical significance of ACE was evaluated through Cox regression analysis. To assess the impact of ACE expression on ccRCC cell growth, metastasis, and glucose activity, CCK-8 assays, transwell assays, Seahorse detection, and xenograft models were utilized. The mechanisms of ACE and its upstream and downstream regulatory factors were investigated using RNA-seq, chromatin immunoprecipitation (ChIP), and luciferase reporter assays. Results RAAS-related gene Angiotensin-Converting Enzyme (ACE) was significantly under expressed in ccRCC cells and tissues. High ACE expression was positively associated with a favorable prognosis in ccRCC patients. Functional studies showed that ACE overexpression suppressed ccRCC cell line OS-RC-2 and A498 growth, metastasis, and glycolysis activities, while its knockdown had the opposite effect. Mechanistically, ACE inhibited ccRCC progression and epithelial-mesenchymal transition (EMT) by disrupting the AKT-FOXO1 signaling pathway. Furthermore, we provide evidence that ACE could enhance everolimus (approved agent for ccRCC) antitumor effect and ACE expression is transcriptionally regulated by ZBTB26. Conclusion Our findings investigated the roles and mechanisms of ACE in ccRCC. ACE inhibits the growth and metastasis of ccRCC cells in vitro and in vivo by promoting FOXO1 expression, which is the downstream target of PI3K-AKT pathway. Thus, this research suggests that ACE may be a promising target for new therapeutic strategy in ccRCC.
Collapse
Affiliation(s)
- Lei Yin
- Department of Urology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, People’s Republic of China
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Department of Urology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, People’s Republic of China
| | - Lixin Mao
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Rui Yin
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
| | - Chengxun Lv
- Department of Urology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, People’s Republic of China
| | - Xiaokai Shi
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Chuang Yue
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Yin Chen
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Chao Lu
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Zonglin Wu
- Department of Urology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, People’s Republic of China
| | - Kai Xu
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Wei Cao
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| |
Collapse
|
11
|
Cao Z, Jiang X, He Y, Zheng X. Metabolic landscape in venous thrombosis: insights into molecular biology and therapeutic implications. Ann Med 2024; 56:2401112. [PMID: 39297312 DOI: 10.1080/07853890.2024.2401112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 03/20/2024] [Accepted: 05/12/2024] [Indexed: 09/21/2024] Open
Abstract
The findings of the last decade suggest a complex link between inflammatory cells, coagulation, and the activation of platelets and their synergistic interaction to promote venous thrombosis. Inflammation is present throughout the process of venous thrombosis, and various metabolic pathways of erythrocytes, endothelial cells, and immune cells involved in venous thrombosis, including glucose metabolism, lipid metabolism, homocysteine metabolism, and oxidative stress, are associated with inflammation. While the metabolic microenvironment has been identified as a marker of malignancy, recent studies have revealed that for cancer thrombosis, alterations in the metabolic microenvironment appear to also be a potential risk. In this review, we discuss how the synergy between metabolism and thrombosis drives thrombotic disease. We also explore the great potential of anti-inflammatory strategies targeting venous thrombosis and the complex link between anti-inflammation and metabolism. Furthermore, we suggest how we can use our existing knowledge to reduce the risk of venous thrombosis.
Collapse
Affiliation(s)
- Zheng Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yiyu He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoxin Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
12
|
Blick-Nitko SK, Ture SK, Schafer XL, Munger JC, Livada AC, Li C, Maurya P, Rondina MT, Morrell CN. Platelet Ido1 expression is induced during Plasmodium yoelii infection, altering plasma tryptophan metabolites. Blood Adv 2024; 8:5814-5825. [PMID: 39133890 PMCID: PMC11609358 DOI: 10.1182/bloodadvances.2024013175] [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] [Received: 03/15/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 11/11/2024] Open
Abstract
ABSTRACT Platelets are immune responsive in many diseases as noted by changes in platelet messenger RNA in conditions such as sepsis, atherosclerosis, COVID-19, and many other inflammatory and infectious etiologies. The malaria causing Plasmodium parasite is a persistent public health threat and significant evidence shows that platelets participate in host responses to infection. Using a mouse model of nonlethal/uncomplicated malaria, non-lethal Plasmodium yoelii strain XNL (PyNL)-infected but not control mouse platelets expressed Ido1, a rate limiting enzyme in tryptophan metabolism that increases kynurenine at the expense of serotonin. Interferon-γ (IFN-γ) is a potent inducer of Ido1 and mice treated with recombinant IFN-γ had increased platelet Ido1 and IDO1 activity. PyNL-infected mice treated with anti-IFN-γ antibody had similar platelet Ido1 and metabolic profiles to that of uninfected controls. PyNL-infected mice become thrombocytopenic by day 7 after infection and transfusion of platelets from IFN-γ-treated wild-type mice but not Ido1-/- mice increased the plasma kynurenine-to-tryptophan ratio, indicating that platelets are a source of postinfection IDO1 activity. We generated platelet-specific Ido1 knockout mice to assess the contribution of platelet Ido1 during PyNL infection. Platelet-specific Ido1-/- mice had increased death and evidence of lung thrombi, which were not present in infected wild-type mice. Platelet Ido1 may be a significant contributor to plasma kynurenine in IFN-γ-driven immune processes and the loss of platelets may limit total Ido1, leading to immune and vascular dysfunction.
Collapse
Affiliation(s)
- Sara K. Blick-Nitko
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Sara K. Ture
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Xenia L. Schafer
- Department of Biochemistry, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Joshua C. Munger
- Department of Biochemistry, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Alison C. Livada
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Chen Li
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Preeti Maurya
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | | | - Craig N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
13
|
Kolenc A, Grundner M, Hostnik I, Maličev E. Monitoring Cell Activation and Extracellular Vesicle Generation in Platelet Concentrates for Transfusion. Int J Mol Sci 2024; 25:11577. [PMID: 39519129 PMCID: PMC11546954 DOI: 10.3390/ijms252111577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Platelets play a crucial role in blood transfusions, and understanding the changes that occur during their storage is important for maintaining the quality of preparations. In this study, we examined key alternating factors, with a particular focus on platelet activation and the release of extracellular vesicles. Additionally, we compared two detection methods-imaging flow cytometry (IFC) and nanoparticle tracking analysis (NTA)-for their effectiveness in detecting particles. Platelet concentrates were prepared by pooling buffy coats from five blood group-compatible donors in an additive solution. Samples were analysed after one, three, and seven days of storage for residual white blood cells (WBCs), glucose levels, platelet activation, and extracellular vesicle concentrations. Over the storage period, the total platelet concentration decreased slightly, while the residual WBC count remained stable. Glucose levels declined, whereas platelet activation and extracellular vesicle concentration increased, with a positive correlation between the two. The particle size remained relatively unchanged throughout the storage period. Ultimately, despite controlled processing and storage conditions, platelet activation, and the release of extracellular vesicles still occurred, which may have implications for transfusion recipients. Although an optimised method is still needed, IFC has proved to be specific and potentially appropriate for detecting extracellular vesicles in transfusion preparations.
Collapse
Affiliation(s)
- Ana Kolenc
- Blood Transfusion Center of Slovenia, Šlajmarjeva 6, 1000 Ljubljana, Slovenia; (A.K.)
| | - Maja Grundner
- Blood Transfusion Center of Slovenia, Šlajmarjeva 6, 1000 Ljubljana, Slovenia; (A.K.)
| | - Irma Hostnik
- Blood Transfusion Center of Slovenia, Šlajmarjeva 6, 1000 Ljubljana, Slovenia; (A.K.)
| | - Elvira Maličev
- Blood Transfusion Center of Slovenia, Šlajmarjeva 6, 1000 Ljubljana, Slovenia; (A.K.)
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, 1000 Ljubljana, Slovenia
| |
Collapse
|
14
|
Noone D, Preston RJS, Rehill AM. The Role of Myeloid Cells in Thromboinflammatory Disease. Semin Thromb Hemost 2024; 50:998-1011. [PMID: 38547918 DOI: 10.1055/s-0044-1782660] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Inflammation contributes to the development of thrombosis, but the mechanistic basis for this association remains poorly understood. Innate immune responses and coagulation pathways are activated in parallel following infection or injury, and represent an important host defense mechanism to limit pathogen spread in the bloodstream. However, dysregulated proinflammatory activity is implicated in the progression of venous thromboembolism and arterial thrombosis. In this review, we focus on the role of myeloid cells in propagating thromboinflammation in acute inflammatory conditions, such as sepsis and coronavirus disease 2019 (COVID-19), and chronic inflammatory conditions, such as obesity, atherosclerosis, and inflammatory bowel disease. Myeloid cells are considered key drivers of thromboinflammation via upregulated tissue factor activity, formation of neutrophil extracellular traps (NETs), contact pathway activation, and aberrant coagulation factor-mediated protease-activated receptor (PAR) signaling. We discuss how strategies to target the intersection between myeloid cell-mediated inflammation and activation of blood coagulation represent an exciting new approach to combat immunothrombosis. Specifically, repurposed anti-inflammatory drugs, immunometabolic regulators, and NETosis inhibitors present opportunities that have the potential to dampen immunothrombotic activity without interfering with hemostasis. Such therapies could have far-reaching benefits for patient care across many thromboinflammatory conditions.
Collapse
Affiliation(s)
- David Noone
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Aisling M Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| |
Collapse
|
15
|
Montecino-Garrido H, Trostchansky A, Espinosa-Parrilla Y, Palomo I, Fuentes E. How Protein Depletion Balances Thrombosis and Bleeding Risk in the Context of Platelet's Activatory and Negative Signaling. Int J Mol Sci 2024; 25:10000. [PMID: 39337488 PMCID: PMC11432290 DOI: 10.3390/ijms251810000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Platelets are small cell fragments that play a crucial role in hemostasis, requiring fast response times and fine signaling pathway regulation. For this regulation, platelets require a balance between two pathway types: the activatory and negative signaling pathways. Activatory signaling mediators are positive responses that enhance stimuli initiated by a receptor in the platelet membrane. Negative signaling regulates and controls the responses downstream of the same receptors to roll back or even avoid spontaneous thrombotic events. Several blood-related pathologies can be observed when these processes are unregulated, such as massive bleeding in activatory signaling inhibition or thrombotic events for negative signaling inhibition. The study of each protein and metabolite in isolation does not help to understand the role of the protein or how it can be contrasted; however, understanding the balance between active and negative signaling could help develop effective therapies to prevent thrombotic events and bleeding disorders.
Collapse
Affiliation(s)
- Hector Montecino-Garrido
- Centro de Estudios en Alimentos Procesados (CEAP), ANID-Regional, Gore Maule R0912001, Talca 3480094, Chile
| | - Andrés Trostchansky
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Yolanda Espinosa-Parrilla
- Interuniversity Center for Healthy Aging (CIES), Centro Asistencial, Docente e Investigación-CADI-UMAG, Escuela de Medicina, Universidad de Magallanes, Punta Arenas 6210427, Chile
| | - Iván Palomo
- Thrombosis and Healthy Aging Research Center, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| | - Eduardo Fuentes
- Thrombosis and Healthy Aging Research Center, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| |
Collapse
|
16
|
Tambralli A, Harbaugh A, NaveenKumar SK, Radyk MD, Rysenga CE, Sabb K, Hurley JM, Sule GJ, Yalavarthi S, Estes SK, Hoy CK, Smith T, Sarosh C, Madison JA, Schaefer JK, Sood SL, Zuo Y, Sawalha AH, Lyssiotis CA, Knight JS. Neutrophil glucose flux as a therapeutic target in antiphospholipid syndrome. J Clin Invest 2024; 134:e169893. [PMID: 38869951 PMCID: PMC11290966 DOI: 10.1172/jci169893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/11/2024] [Indexed: 06/15/2024] Open
Abstract
Neutrophil hyperactivity and neutrophil extracellular trap release (NETosis) appear to play important roles in the pathogenesis of the thromboinflammatory autoimmune disease known as antiphospholipid syndrome (APS). The understanding of neutrophil metabolism has advanced tremendously in the past decade, and accumulating evidence suggests that a variety of metabolic pathways guide neutrophil activities in health and disease. Our previous work characterizing the transcriptome of APS neutrophils revealed that genes related to glycolysis, glycogenolysis, and the pentose phosphate pathway (PPP) were significantly upregulated. Here, we found that neutrophils from patients with APS used glycolysis more avidly than neutrophils from people in the healthy control group, especially when the neutrophils were from patients with APS with a history of microvascular disease. In vitro, inhibiting either glycolysis or the PPP tempered phorbol myristate acetate- and APS IgG-induced NETosis, but not NETosis triggered by a calcium ionophore. In mice, inhibiting either glycolysis or the PPP reduced neutrophil reactive oxygen species production and suppressed APS IgG-induced NETosis ex vivo. When APS-associated thrombosis was evaluated in mice, inhibiting either glycolysis or the PPP markedly suppressed thrombosis and circulating NET remnants. In summary, these data identify a potential role for restraining neutrophil glucose flux in the treatment of APS.
Collapse
Affiliation(s)
- Ajay Tambralli
- Division of Rheumatology, Department of Internal Medicine
- Division of Pediatric Rheumatology, Department of Pediatrics
| | | | | | | | | | - Kaitlyn Sabb
- Division of Rheumatology, Department of Internal Medicine
| | | | - Gautam J. Sule
- Division of Rheumatology, Department of Internal Medicine
| | | | | | - Claire K. Hoy
- Division of Rheumatology, Department of Internal Medicine
| | - Tristin Smith
- Division of Rheumatology, Department of Internal Medicine
| | - Cyrus Sarosh
- Division of Rheumatology, Department of Internal Medicine
| | - Jacqueline A. Madison
- Division of Rheumatology, Department of Internal Medicine
- Division of Pediatric Rheumatology, Department of Pediatrics
| | - Jordan K. Schaefer
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Suman L. Sood
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yu Zuo
- Division of Rheumatology, Department of Internal Medicine
| | - Amr H. Sawalha
- Departments of Pediatrics, Medicine, and Immunology, and Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
17
|
Shah SS, Stone EF, Francis RO, Karafin MS. The global role of G6PD in infection and immunity. Front Immunol 2024; 15:1393213. [PMID: 38938571 PMCID: PMC11208698 DOI: 10.3389/fimmu.2024.1393213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) deficiency is the most common enzymopathy in humans. G6PD is an essential enzyme in the pentose phosphate pathway (PPP), generating NADPH needed for cellular biosynthesis and reactive oxygen species (ROS) homeostasis, the latter especially key in red blood cells (RBCs). Beyond the RBC, there is emerging evidence that G6PD exerts an immunologic role by virtue of its functions in leukocyte oxidative metabolism and anabolic synthesis necessary for immune effector function. We review these here, and consider the global immunometabolic role of G6PD activity and G6PD deficiency in modulating inflammation and immunopathology.
Collapse
Affiliation(s)
- Shivang S. Shah
- Department of Pediatrics, Columbia University, New York, NY, United States
| | - Elizabeth F. Stone
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Richard O. Francis
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Matthew S. Karafin
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
18
|
Akther F, Fallahi H, Zhang J, Nguyen NT, Ta HT. Evaluating thrombosis risk and patient-specific treatment strategy using an atherothrombosis-on-chip model. LAB ON A CHIP 2024; 24:2927-2943. [PMID: 38591995 DOI: 10.1039/d4lc00131a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Platelets play an essential role in thrombotic processes. Recent studies suggest a direct link between increased plasma glucose, lipids, and inflammatory cytokines with platelet activation and aggregation, resulting in an increased risk of atherothrombotic events in cardiovascular patients. Antiplatelet therapies are commonly used for the primary prevention of atherosclerosis. Transitioning from a population-based strategy to patient-specific care requires a better understanding of the risks and advantages of antiplatelet therapy for individuals. This proof-of-concept study evaluates the potential to assess an individual's risk of forming atherothrombosis using a dual-channel microfluidic model emulating multiple atherogenic factors in vitro, including high glucose, high cholesterol, and inflammatory cytokines along with stenosis vessel geometry. The model shows precise sensitivity toward increased plasma glucose, cholesterol, and tumour necrosis factor-alpha (TNF-α)-treated groups in thrombus formation. An in vivo-like dose-dependent increment in platelet aggregation is observed in different treated groups, benefiting the evaluation of thrombosis risk in the individual condition. Moreover, the model could help decide the effective dosing of aspirin in multi-factorial complexities. In the high glucose-treated group, a 50 μM dose of aspirin could significantly reduce platelet aggregation, while a 100 μM dose of aspirin was required to reduce platelet aggregation in the glucose-TNF-α-treated group, which proves the model's potentiality as a tailored tool for customised therapy.
Collapse
Affiliation(s)
- Fahima Akther
- Queensland Micro- and Nanotechnology, Griffith University, Nathan Campus, Brisbane, Queensland 4111, Australia.
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Hedieh Fallahi
- Queensland Micro- and Nanotechnology, Griffith University, Nathan Campus, Brisbane, Queensland 4111, Australia.
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Jun Zhang
- Queensland Micro- and Nanotechnology, Griffith University, Nathan Campus, Brisbane, Queensland 4111, Australia.
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology, Griffith University, Nathan Campus, Brisbane, Queensland 4111, Australia.
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Hang Thu Ta
- Queensland Micro- and Nanotechnology, Griffith University, Nathan Campus, Brisbane, Queensland 4111, Australia.
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| |
Collapse
|
19
|
Dash D. New Dimension to Study on Platelet Bioenergetics. Arterioscler Thromb Vasc Biol 2024; 44:417-418. [PMID: 38126171 DOI: 10.1161/atvbaha.123.320355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Affiliation(s)
- Debabrata Dash
- Centre for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221005, Uttar Pradesh, India
| |
Collapse
|
20
|
Zhu J, Wang R, Yang C, Shao X, Zhang Y, Hou J, Gao Y, Ou A, Chen M, Huang Y. Blocking tumor-platelet crosstalk to prevent tumor metastasis via reprograming glycolysis using biomimetic membrane-hybridized liposomes. J Control Release 2024; 366:328-341. [PMID: 38168561 DOI: 10.1016/j.jconrel.2023.12.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
Activated platelets promote tumor progression and metastasis through active interactions with cancer cells, especially in promoting epithelial-mesenchymal transition (EMT) of tumor cells and shedding tumor cells into the blood. Blocking platelet-tumor cell interactions can be a potential strategy to inhibit tumor metastasis. Platelet activation requires energy produced from aerobic glycolysis. Based on this, we propose a platelet suppression strategy by reprogramming glucose metabolism of platelets, which has an advantage over conventional antiplatelet treatment that has a risk of serious hemorrhage. We develop a biomimetic delivery system using platelet membrane-hybridized liposomes (PM-Lipo) for codelivery of quercetin and shikonin to simultaneously inhibit lactate transporter MCT-4 and a glycolytic enzyme PKM2 for achieving metabolic reprogramming of platelets and suppressing platelet activation. Notably, PM-Lipo can also inhibit glycolysis in cancer cells, which actually takes "two-birds-one-stone" action. Consequently, the platelet-tumor cell interactions are inhibited. Moreover, PM-Lipo can bind with circulating tumor cells and reduce their seeding in the premetastatic microenvironment. The in vivo studies further demonstrated that PM-Lipo can effectively suppress primary tumor growth and reduce lung metastasis without affecting inherited functions of platelets. Reprogramming glycolysis of platelets can remodel the tumor immune microenvironment, including suppression of Treg and stimulation of CTLs.
Collapse
Affiliation(s)
- Jie Zhu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rui Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; Nanchang University College of Pharmacy, 461 Bayi Rd, Nanchang 330006, China
| | - Chenxiao Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; Nanchang University College of Pharmacy, 461 Bayi Rd, Nanchang 330006, China
| | - Xinyue Shao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; Department of Implant Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jiazhen Hou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China
| | - Yanrong Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ante Ou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, SAR, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Zhongshan Institute for Drug Discovery, The Institutes of Drug Discovery and Development, Chinese Academy of Sciences, Zhongshan 528437, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| |
Collapse
|
21
|
Ghatge M, Flora GD, Nayak MK, Chauhan AK. Platelet Metabolic Profiling Reveals Glycolytic and 1-Carbon Metabolites Are Essential for GP VI-Stimulated Human Platelets-Brief Report. Arterioscler Thromb Vasc Biol 2024; 44:409-416. [PMID: 37942614 PMCID: PMC10880120 DOI: 10.1161/atvbaha.123.319821] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Evolving evidence suggests that besides signaling pathways, platelet activation involves a complex interplay between metabolic pathways to support thrombus growth. Selective targeting of metabolic checkpoints may inhibit platelet activation and provide a novel antiplatelet strategy. We, therefore, examined global metabolic changes that occur during the transition of human platelets from resting to an activated state to identify metabolites and associated pathways that contribute to platelet activation. METHODS We performed metabolic profiling of resting and convulxin-stimulated human platelet samples. The differential levels, pathway analysis, and PCA (principal component analysis) were performed using Metaboanalyst. Metascape was used for metabolite network construction. RESULTS Of the 401 metabolites identified, 202 metabolites were significantly upregulated, and 2 metabolites were downregulated in activated platelets. Of all the metabolites, lipids scored highly and constituted ≈50% of the identification. During activation, aerobic glycolysis supports energy demand and provides glycolytic intermediates required by metabolic pathways. Consistent with this, an important category of metabolites was carbohydrates, particularly the glycolysis intermediates that were significantly upregulated compared with resting platelets. We found that lysophospholipids such as 1-palmitoyl-GPA (glycero-3-phosphatidic acid), 1-stearoyl-GPS (glycero-3-phosphoserine), 1-palmitoyl-GPI (glycerophosphoinositol), 1-stearoyl-GPI, and 1-oleoyl-GPI were upregulated in activated platelets. We speculated that platelet activation could be linked to 1-carbon metabolism, a set of biochemical pathways that involve the transfer and use of 1-carbon units from amino acids, for cellular processes, including nucleotide and lysophospholipid synthesis. In alignment, based on pathway enrichment and network-based prioritization, the metabolites from amino acid metabolism, including serine, glutamate, and branched-chain amino acid pathway were upregulated in activated platelets, which might be supplemented by the high levels of glycolytic intermediates. CONCLUSIONS Metabolic analysis of resting and activated platelets revealed that glycolysis and 1-carbon metabolism are necessary to support platelet activation.
Collapse
Affiliation(s)
| | - Gagan D. Flora
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | | | | |
Collapse
|
22
|
Jacob S, Kosaka Y, Bhatlekar S, Denorme F, Benzon H, Moody A, Moody V, Tugolukova E, Hull G, Kishimoto N, Manne BK, Guo L, Souvenir R, Seliger BJ, Eustes AS, Hoerger K, Tolley ND, Fatahian AN, Boudina S, Christiani DC, Wei Y, Ju C, Campbell RA, Rondina MT, Abel ED, Bray PF, Weyrich AS, Rowley JW. Mitofusin-2 Regulates Platelet Mitochondria and Function. Circ Res 2024; 134:143-161. [PMID: 38156445 PMCID: PMC10872864 DOI: 10.1161/circresaha.123.322914] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 12/13/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Single-nucleotide polymorphisms linked with the rs1474868 T allele (MFN2 [mitofusin-2] T/T) in the human mitochondrial fusion protein MFN2 gene are associated with reduced platelet MFN2 RNA expression and platelet counts. This study investigates the impact of MFN2 on megakaryocyte and platelet biology. METHODS Mice with megakaryocyte/platelet deletion of Mfn2 (Mfn2-/- [Mfn2 conditional knockout]) were generated using Pf4-Cre crossed with floxed Mfn2 mice. Human megakaryocytes were generated from cord blood and platelets isolated from healthy subjects genotyped for rs1474868. Ex vivo approaches assessed mitochondrial morphology, function, and platelet activation responses. In vivo measurements included endogenous/transfused platelet life span, tail bleed time, transient middle cerebral artery occlusion, and pulmonary vascular permeability/hemorrhage following lipopolysaccharide-induced acute lung injury. RESULTS Mitochondria was more fragmented in megakaryocytes derived from Mfn2-/- mice and from human cord blood with MFN2 T/T genotype compared with control megakaryocytes. Human resting platelets of MFN2 T/T genotype had reduced MFN2 protein, diminished mitochondrial membrane potential, and an increased rate of phosphatidylserine exposure during ex vivo culture. Platelet counts and platelet life span were reduced in Mfn2-/- mice accompanied by an increased rate of phosphatidylserine exposure in resting platelets, especially aged platelets, during ex vivo culture. Mfn2-/- also decreased platelet mitochondrial membrane potential (basal) and activated mitochondrial oxygen consumption rate, reactive oxygen species generation, calcium flux, platelet-neutrophil aggregate formation, and phosphatidylserine exposure following dual agonist activation. Ultimately, Mfn2-/- mice showed prolonged tail bleed times, decreased ischemic stroke infarct size after cerebral ischemia-reperfusion, and exacerbated pulmonary inflammatory hemorrhage following lipopolysaccharide-induced acute lung injury. Analysis of MFN2 SNPs in the iSPAAR study (Identification of SNPs Predisposing to Altered ALI Risk) identified a significant association between MFN2 and 28-day mortality in patients with acute respiratory distress syndrome. CONCLUSIONS Mfn2 preserves mitochondrial phenotypes in megakaryocytes and platelets and influences platelet life span, function, and outcomes of stroke and lung injury.
Collapse
Affiliation(s)
- Shancy Jacob
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Yasuhiro Kosaka
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Seema Bhatlekar
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Frederik Denorme
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Haley Benzon
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Alexandra Moody
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Victoria Moody
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | | | - Grayson Hull
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Nina Kishimoto
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Bhanu K. Manne
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Li Guo
- Bloodworks Northwest Research Institute, Seattle, WA
- Division of Hematology and Oncology, University of Utah, Seattle, WA
| | - Rhonda Souvenir
- David Geffen School of Medicine and University of California, Los Angeles (UCLA), Health, Los Angeles, CA
| | | | | | - Kelly Hoerger
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Neal D. Tolley
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Amir N. Fatahian
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT
| | - Sihem Boudina
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT
| | - David C. Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA, 02115, USA
| | - Yongyue Wei
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, 100191, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, 100191, China
| | - Can Ju
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Robert A. Campbell
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
- Department of Pathology, University of Utah Heath, Salt Lake City, UT
| | - Matthew T. Rondina
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
- Department of Pathology, University of Utah Heath, Salt Lake City, UT
- Department of Internal Medicine and the GRECC, George E. Wahlen VAMC, Salt Lake City, UT
| | - E. Dale Abel
- David Geffen School of Medicine and University of California, Los Angeles (UCLA), Health, Los Angeles, CA
| | - Paul F. Bray
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Andrew S. Weyrich
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK
| | - Jesse W. Rowley
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
| |
Collapse
|
23
|
Roch B, Pisareva E, Mirandola A, Sanchez C, Pastor B, Tanos R, Frayssinoux F, Diab-Assaf M, Anker P, Al Amir Dache Z, Thierry AR. Impact of platelet activation on the release of cell-free mitochondria and circulating mitochondrial DNA. Clin Chim Acta 2024; 553:117711. [PMID: 38101467 DOI: 10.1016/j.cca.2023.117711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND Research on circulating mitochondrial DNA (cir-mtDNA) based diagnostic is insufficient, as to its function, origin, structural features, and particularly its standardization of isolation. To date, plasma preparation performed in previous studies do not take into consideration the potential bias resulting from the release of mitochondria by activated platelets. METHODS To tackle this, we compared the mtDNA amount determined by a standard plasma preparation method or a method optimally avoiding platelet activation. MtDNA extracted from the plasma of seven healthy individuals was quantified by Q-PCR in the course of the process of both methods submitted to filtration, freezing or differential centrifugation. RESULTS 98.7 to 99.4% of plasma mtDNA corresponded to extracellular mitochondria, either free or into large extracellular vesicles. Without platelet activation, the proportion of both types of entities remained preponderant (76-80%), but the amount of detected mtDNA decreased 67-fold. CONCLUSION We show the high capacity of platelets to release free mitochondria in "in vitro" conditions. This represents a potent confounding factor when extracting mtDNA for cir-mtDNA investigation. Platelet activation during pre-analytical conditions should therefore be avoided when studying cir-mtDNA. Our findings lead to a profound revision of the assumptions previously made by most works in this field. Overall, our data suggest the need to characterize or isolate mtDNA associated various structural forms, as well as to standardize plasma preparation, to better circumscribe cir-mtDNA's diagnostic capacity.
Collapse
Affiliation(s)
- Benoit Roch
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier F-34298, France; Thoracic Oncology Unit, Arnaud de Villeneuve Hospital, University Hospital of Montpellier, Montpellier F-34295, France
| | - Ekaterina Pisareva
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier F-34298, France
| | - Alexia Mirandola
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier F-34298, France
| | - Cynthia Sanchez
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier F-34298, France
| | - Brice Pastor
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier F-34298, France
| | - Rita Tanos
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier F-34298, France
| | - Florence Frayssinoux
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier F-34298, France
| | - Mona Diab-Assaf
- Faculty of Sciences II, Lebanese University Fanar, Beirut, Lebanon
| | - Philippe Anker
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier F-34298, France
| | - Zahra Al Amir Dache
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier F-34298, France
| | - Alain R Thierry
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier F-34298, France; ICM, Institut Régional du Cancer de Montpellier, Montpellier F-34298, France.
| |
Collapse
|
24
|
Spinello I, Saulle E, Quaranta MT, Pelosi E, Castelli G, Cerio A, Pasquini L, Morsilli O, Dupuis ML, Labbaye C. AC-73 and Syrosingopine Inhibit SARS-CoV-2 Entry into Megakaryocytes by Targeting CD147 and MCT4. Viruses 2024; 16:82. [PMID: 38257782 PMCID: PMC10818282 DOI: 10.3390/v16010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
Coagulation disorders are described in COVID-19 and long COVID patients. In particular, SARS-CoV-2 infection in megakaryocytes, which are precursors of platelets involved in thrombotic events in COVID-19, long COVID and, in rare cases, in vaccinated individuals, requires further investigation, particularly with the emergence of new SARS-CoV-2 variants. CD147, involved in the regulation of inflammation and required to fight virus infection, can facilitate SARS-CoV-2 entry into megakaryocytes. MCT4, a co-binding protein of CD147 and a key player in the glycolytic metabolism, could also play a role in SARS-CoV-2 infection. Here, we investigated the susceptibility of megakaryocytes to SARS-CoV-2 infection via CD147 and MCT4. We performed infection of Dami cells and human CD34+ hematopoietic progenitor cells induced to megakaryocytic differentiation with SARS-CoV-2 pseudovirus in the presence of AC-73 and syrosingopine, respective inhibitors of CD147 and MCT4 and inducers of autophagy, a process essential in megakaryocyte differentiation. Both AC-73 and syrosingopine enhance autophagy during differentiation but only AC-73 enhances megakaryocytic maturation. Importantly, we found that AC-73 or syrosingopine significantly inhibits SARS-CoV-2 infection of megakaryocytes. Altogether, our data indicate AC-73 and syrosingopine as inhibitors of SARS-CoV-2 infection via CD147 and MCT4 that can be used to prevent SARS-CoV-2 binding and entry into megakaryocytes, which are precursors of platelets involved in COVID-19-associated coagulopathy.
Collapse
Affiliation(s)
- Isabella Spinello
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (E.S.); (M.T.Q.); (M.L.D.)
| | - Ernestina Saulle
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (E.S.); (M.T.Q.); (M.L.D.)
| | - Maria Teresa Quaranta
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (E.S.); (M.T.Q.); (M.L.D.)
| | - Elvira Pelosi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.P.); (G.C.); (A.C.)
| | - Germana Castelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.P.); (G.C.); (A.C.)
| | - Annamaria Cerio
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.P.); (G.C.); (A.C.)
| | - Luca Pasquini
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Ornella Morsilli
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Ageing, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Luisa Dupuis
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (E.S.); (M.T.Q.); (M.L.D.)
| | - Catherine Labbaye
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (E.S.); (M.T.Q.); (M.L.D.)
| |
Collapse
|
25
|
Priya V, Samridhi, Singh N, Dash D, Muthu MS. Nattokinase Encapsulated Nanomedicine for Targeted Thrombolysis: Development, Improved in Vivo Thrombolytic Effects, and Ultrasound/Photoacoustic Imaging. Mol Pharm 2024; 21:283-302. [PMID: 38126777 DOI: 10.1021/acs.molpharmaceut.3c00830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Nattokinase (NK), a potent thrombolytic enzyme that dissolves blood clots, is highly used in the treatment of cardiovascular disorders. However, its effective delivery remains demanding because of stability and bioavailability problems owing to its high molecular weight and proteineous nature. In this research, we have developed novel NK-loaded nontargeted liposomes (NK-LS) and targeted liposomes (RGD-NK-LS and AM-NK-LS) by the reverse phase evaporation method. The physiochemical characterizations (particle size, polydispersity index, zeta potential, and morphology) were performed by a Zetasizer, SEM, TEM, and AFM. The Bradford assay and XPS analysis confirmed the successful surface conjugation of the targeting ligands. Platelet interaction studies by CLSM, photon imager optima, and flow cytometry showed significantly higher (P < 0.05) platelet binding affinity of targeted liposomes. In vitro evaluations were performed using human blood and a fibrinolysis study by CLSM imaging demonstrating the potent antithrombotic efficacy of AM-NK-LS. Furthermore, bleeding and clotting time studies revealed that the targeted liposomes were free from any bleeding complications. Moreover, the in vivo FeCl3 model on Sprague-Dawley (SD) rats using a Doppler flow meter and ultrasound/photoacoustic imaging indicated the increased % thrombolysis and potent affinity of targeted liposomes toward the thrombus site. Additionally, in vitro hemocompatibility and histopathology studies demonstrated the safety and biocompatibility of the nanoformulations.
Collapse
Affiliation(s)
- Vishnu Priya
- Department of Pharmaceutical Engineering and Technology, IIT (BHU), Varanasi 221005, U.P., India
| | - Samridhi
- Department of Pharmaceutical Engineering and Technology, IIT (BHU), Varanasi 221005, U.P., India
| | - Nitesh Singh
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Debabrata Dash
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, IIT (BHU), Varanasi 221005, U.P., India
| |
Collapse
|
26
|
Chaurasia SN, Singh V, Ekhlak M, Dash MK, Joshi N, Dash D. Ayurvedic preparations of Raudra Rasa inhibit agonist-mediated platelet activation and restrict thrombogenicity without affecting cell viability. FEBS Open Bio 2023; 13:2342-2355. [PMID: 37787005 PMCID: PMC10699108 DOI: 10.1002/2211-5463.13713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/01/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023] Open
Abstract
Ayurveda is considered to be one of the most ancient forms of medicine still practiced. The Ayurvedic preparation Raudra Rasa and its derivatives have been widely employed against cancer since the 12th century, but the effect of these traditional formulations on platelet function and signaling has not previously been examined. Here we demonstrate that Raudra Rasa and its derivatives significantly reduce thrombin-induced integrin activation and granule secretion in platelets, as observed by reduced PAC-1 binding and P-selectin externalization, respectively. These formulations also inhibited thrombin-stimulated phosphatidylserine exposure, mitochondrial reactive oxygen species generation, and mitochondrial transmembrane potential in platelets. Consistent with the above, Raudra Rasa significantly reduced thrombin-induced tyrosine phosphorylation of the platelet proteins, as well as phosphorylation of the enzymes AKT and GSK-3β. In summary, Raudra Rasa inhibits agonist-mediated platelet activation without affecting cell viability, suggesting it may have therapeutic potential as an anti-platelet/anti-thrombotic agent.
Collapse
Affiliation(s)
- Susheel Nidhi Chaurasia
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Vipin Singh
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Manoj Kumar Dash
- Department of Rasa Shastra & B KalpanaGovernment Ayurved CollegeRaipurIndia
| | - Namrata Joshi
- Department of Rasa Shastra, Faculty of Ayurveda, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| |
Collapse
|
27
|
Flora GD, Nayak MK, Ghatge M, Chauhan AK. Metabolic targeting of platelets to combat thrombosis: dawn of a new paradigm? Cardiovasc Res 2023; 119:2497-2507. [PMID: 37706546 PMCID: PMC10676458 DOI: 10.1093/cvr/cvad149] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/29/2023] [Accepted: 07/18/2023] [Indexed: 09/15/2023] Open
Abstract
Current antithrombotic therapies used in clinical settings target either the coagulation pathways or platelet activation receptors (P2Y12 or GPIIb/IIIa), as well as the cyclooxygenase (COX) enzyme through aspirin. However, they are associated with bleeding risk and are not suitable for long-term use. Thus, novel strategies which provide broad protection against platelet activation with minimal bleeding risks are required. Regardless of the nature of agonist stimulation, platelet activation is an energy-intensive and ATP-driven process characterized by metabolic switching toward a high rate of aerobic glycolysis, relative to oxidative phosphorylation (OXPHOS). Consequently, there has been considerable interest in recent years in investigating whether targeting metabolic pathways in platelets, especially aerobic glycolysis and OXPHOS, can modulate their activation, thereby preventing thrombosis. This review briefly discusses the choices of metabolic substrates available to platelets that drive their metabolic flexibility. We have comprehensively elucidated the relevance of aerobic glycolysis in facilitating platelet activation and the underlying molecular mechanisms that trigger this switch from OXPHOS. We have provided a detailed account of the antiplatelet effects of targeting vital metabolic checkpoints such as pyruvate dehydrogenase kinases (PDKs) and pyruvate kinase M2 (PKM2) that preferentially drive the pyruvate flux to aerobic glycolysis. Furthermore, we discuss the role of fatty acids and glutamine oxidation in mitochondria and their subsequent role in driving OXPHOS and platelet activation. While the approach of targeting metabolic regulatory mechanisms in platelets to prevent their activation is still in a nascent stage, accumulating evidence highlights its beneficial effects as a potentially novel antithrombotic strategy.
Collapse
Affiliation(s)
- Gagan D Flora
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, IA, USA
| | - Manasa K Nayak
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, IA, USA
| | - Madankumar Ghatge
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, IA, USA
| | - Anil K Chauhan
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
28
|
Grichine A, Jacob S, Eckly A, Villaret J, Joubert C, Appaix F, Pezet M, Ribba AS, Denarier E, Mazzega J, Rinckel JY, Lafanechère L, Elena-Herrmann B, Rowley JW, Sadoul K. The fate of mitochondria during platelet activation. Blood Adv 2023; 7:6290-6302. [PMID: 37624769 PMCID: PMC10589785 DOI: 10.1182/bloodadvances.2023010423] [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] [Received: 04/07/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Blood platelets undergo several successive motor-driven reorganizations of the cytoskeleton when they are recruited to an injured part of a vessel. These reorganizations take place during the platelet activation phase, the spreading process on the injured vessel or between fibrin fibers of the forming clot, and during clot retraction. All these steps require a lot of energy, especially the retraction of the clot when platelets develop strong forces similar to those of muscle cells. Platelets can produce energy through glycolysis and mitochondrial respiration. However, although resting platelets have only 5 to 8 individual mitochondria, they produce adenosine triphosphate predominantly via oxidative phosphorylation. Activated, spread platelets show an increase in size compared with resting platelets, and the question arises as to where the few mitochondria are located in these larger platelets. Using expansion microscopy, we show that the number of mitochondria per platelet is increased in spread platelets. Live imaging and focused ion beam-scanning electron microscopy suggest that a mitochondrial fission event takes place during platelet activation. Fission is Drp1 dependent because Drp1-deficient platelets have fused mitochondria. In nucleated cells, mitochondrial fission is associated with a shift to a glycolytic phenotype, and using clot retraction assays, we show that platelets have a more glycolytic energy production during clot retraction and that Drp1-deficient platelets show a defect in clot retraction.
Collapse
Affiliation(s)
- Alexei Grichine
- INSERM U1209, Centre National de la Recherche Scientifique Unité Mixed de Recherche 5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| | - Shancy Jacob
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Anita Eckly
- INSERM, EFS Grand Est, Biologie et Pharmacologie des Plaquettes Sanguines Unité Mixed de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, Strasbourg, France
| | - Joran Villaret
- INSERM U1209, Centre National de la Recherche Scientifique Unité Mixed de Recherche 5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| | - Clotilde Joubert
- INSERM U1209, Centre National de la Recherche Scientifique Unité Mixed de Recherche 5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| | - Florence Appaix
- INSERM U1209, Centre National de la Recherche Scientifique Unité Mixed de Recherche 5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| | - Mylène Pezet
- INSERM U1209, Centre National de la Recherche Scientifique Unité Mixed de Recherche 5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| | - Anne-Sophie Ribba
- INSERM U1209, Centre National de la Recherche Scientifique Unité Mixed de Recherche 5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| | - Eric Denarier
- INSERM U1216, Commissariat à l'Energie Atomique, Grenoble Institute of Neuroscience, University Grenoble Alpes, Grenoble, France
| | - Jacques Mazzega
- INSERM U1209, Centre National de la Recherche Scientifique Unité Mixed de Recherche 5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| | - Jean-Yves Rinckel
- INSERM, EFS Grand Est, Biologie et Pharmacologie des Plaquettes Sanguines Unité Mixed de Recherche-S 1255, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, Strasbourg, France
| | - Laurence Lafanechère
- INSERM U1209, Centre National de la Recherche Scientifique Unité Mixed de Recherche 5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| | - Bénédicte Elena-Herrmann
- INSERM U1209, Centre National de la Recherche Scientifique Unité Mixed de Recherche 5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| | - Jesse W. Rowley
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Karin Sadoul
- INSERM U1209, Centre National de la Recherche Scientifique Unité Mixed de Recherche 5309, Institute for Advanced Biosciences, University Grenoble Alpes, Grenoble, France
| |
Collapse
|
29
|
Fuentes E, Arauna D, Araya-Maturana R. Regulation of mitochondrial function by hydroquinone derivatives as prevention of platelet activation. Thromb Res 2023; 230:55-63. [PMID: 37639783 DOI: 10.1016/j.thromres.2023.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/07/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
Platelet activation plays an essential role in the pathogenesis of thrombotic events in different diseases (e.g., cancer, type 2 diabetes, Alzheimer's, and cardiovascular diseases, and even in patients diagnosed with coronavirus disease 2019). Therefore, antiplatelet therapy is essential to reduce thrombus formation. However, the utility of current antiplatelet drugs is limited. Therefore, identifying novel antiplatelet compounds is very important in developing new drugs. In this context, the involvement of mitochondrial function as an efficient energy source required for platelet activation is currently accepted; however, its contribution as an antiplatelet target still has little been exploited. Regarding this, the intramolecular hydrogen bonding of hydroquinone derivatives has been described as a structural motif that allows the reach of small molecules at mitochondria, which can exert antiplatelet activity, among others. In this review, we describe the role of mitochondrial function in platelet activation and how hydroquinone derivatives exert antiplatelet activity through mitochondrial regulation.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Universidad de Talca, Talca 3480094, Chile.
| | - Diego Arauna
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Universidad de Talca, Talca 3480094, Chile
| | - Ramiro Araya-Maturana
- Instituto de Química de Recursos Naturales, MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Universidad de Talca, Talca 3460000, Chile
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Platelet mitochondrial dysfunction is both caused by, as well as a source of oxidative stress. Oxidative stress is a key hallmark of metabolic disorders such as dyslipidemia and diabetes, which are known to have higher risks for thrombotic complications. RECENT FINDINGS Increasing evidence supports a critical role for platelet mitochondria beyond energy production and apoptosis. Mitochondria are key regulators of reactive oxygen species and procoagulant platelets, which both contribute to pathological thrombosis. Studies targeting platelet mitochondrial pathways have reported promising results suggesting antithrombotic effects with limited impact on hemostasis in animal models. SUMMARY Targeting platelet mitochondria holds promise for the reduction of thrombotic complications in patients with metabolic disorders. Future studies should aim at validating these preclinical findings and translate them to the clinic.
Collapse
Affiliation(s)
- Abigail Ajanel
- University of Utah Molecular Medicine Program, Salt Lake City, Utah
- Department Pathology, Division of Microbiology and Pathology, University of Utah, Salt Lake City, Utah
| | - Robert A. Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, Utah
- Department Pathology, Division of Microbiology and Pathology, University of Utah, Salt Lake City, Utah
- Department of Internal Medicine, Division of Hematology, University of Utah, Salt Lake City, Utah
| | - Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, Utah
- Department of Neurology, Division of Vascular Neurology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
31
|
Ushiki T, Mochizuki T, Suzuki K, Kamimura M, Ishiguro H, Suwabe T, Watanabe S, Omori G, Yamamoto N, Kawase T. Strategic analysis of body composition indices and resting platelet ATP levels in professional soccer players for better platelet-rich plasma therapy. Front Bioeng Biotechnol 2023; 11:1255860. [PMID: 37711445 PMCID: PMC10499317 DOI: 10.3389/fbioe.2023.1255860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/17/2023] [Indexed: 09/16/2023] Open
Abstract
Background: Autologous platelet-rich plasma (PRP) therapy is ambiguously thought to be more effective in elite athletes than in sedentary patients, although the possible importance of recipient responsiveness remains poorly understood. To address this issue, along with the well-known PRP quality, in this initial study, we evaluated two candidate biomarkers: body composition indices (BCIs), which reflect systemic physical conditions, and resting platelet ATP levels, which reflect platelet energy expenditure and the mass of energy generation units. Methods: In this cross-sectional cohort study, blood samples were collected from male professional soccer players (PSPs) on a local professional team during the off-season and platelet ATP levels were quantified using an ATP luminescence assay kit. BCIs were measured using the body mass impedance method. Age-matched male sedentary participants were used as the controls. Results: Among the BCIs, the body mass index, basal metabolic rate (BMR), and skeletal muscle weight levels were higher in the PSPs than in the controls. The platelet ATP levels in the PSPs group were significantly lower than those in the control group. The correlation between BMR and platelet ATP levels was moderately negative in the control group, but weakly positive in the PSPs group. Conclusion: Owing to regular physical exercise, PSPs had higher BMR levels and lower platelet ATP levels without a significant mutual correlation compared to sedentary controls. This study did not indicate the influence of these biomarkers on the success of PRP therapy but provided evidence for a better understanding of PRP therapy, particularly for elite athletes.
Collapse
Affiliation(s)
- Takashi Ushiki
- Division of Hematology and Oncology, Graduate School of Health Sciences, Niigata University, Niigata, Japan
- Department of Transfusion Medicine, Cell Therapy and Regenerative Medicine, Niigata University Medical and Dental Hospital, Niigata, Japan
- Department of Hematology, Endocrinology and Metabolism, Faculty of Medicine, Niigata University, Niigata, Japan
| | - Tomoharu Mochizuki
- Department of Orthopaedic Surgery, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Katsuya Suzuki
- Department of Transfusion Medicine, Cell Therapy and Regenerative Medicine, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Masami Kamimura
- Department of Transfusion Medicine, Cell Therapy and Regenerative Medicine, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Hajime Ishiguro
- Department of Orthopaedic Surgery, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tatsuya Suwabe
- Department of Orthopaedic Surgery, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Satoshi Watanabe
- Department of Orthopaedic Surgery, Niigata Medical Center, Niigata, Japan
| | - Go Omori
- Department of Health and Sports, Faculty of Health Sciences, Niigata University of Health and Welfare, Niigata, Japan
| | - Noriaki Yamamoto
- Department of Orthopaedic Surgery, Niigata Rehabilitation Hospital, Niigata, Japan
| | - Tomoyuki Kawase
- Division of Oral Bioengineering, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
32
|
Ghatge M, Nayak MK, Flora GD, Kumskova M, Jain A, Patel RB, Lin Z, Usachev YM, Chauhan AK. Mitochondrial calcium uniporter b deletion inhibits platelet function and reduces susceptibility to arterial thrombosis. J Thromb Haemost 2023; 21:2163-2174. [PMID: 37061131 DOI: 10.1016/j.jtha.2023.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/10/2023] [Accepted: 04/03/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND Mitochondrial calcium uniporter b (MCUb) is a negative regulator of the mitochondrial calcium uniporter (MCU) and is known to limit mitochondrial calcium ion (Ca2+) uptake. The role of MCUb in platelet function remains unclear. OBJECTIVES Utilizing MCUb-/- mice, we examined the role of MCUb in regulating platelet function and thrombosis. METHODS Platelet activation was evaluated in agonist-induced standardized in vitro assays. Susceptibility to arterial thrombosis was evaluated in FeCl3 injury-induced carotid artery and laser injury-induced mesenteric artery thrombosis models. The glycolytic proton efflux rate and oxygen consumption rate were measured to evaluate aerobic glycolysis. RESULTS Upon stimulation, MCUb-/- platelets exhibited reduced cytoplasmic Ca2+ responses concomitant with increased mitochondrial Ca2+ uptake. MCUb-/- platelets displayed reduced agonist-induced platelet aggregation and spreading on fibrinogen and decreased α and dense-granule secretion and clot retraction. MCUb-/- mice were less susceptible to arterial thrombosis in FeCl3 injury-induced carotid and laser injury-induced mesenteric thrombosis models with unaltered tail bleeding time. In adoptive transfer experiments, thrombocytopenic hIL-4Rα/GPIbα-transgenic mice transfused with MCUb-/- platelets were less susceptible to FeCl3 injury-induced carotid thrombosis compared with hIL-4Rα/GPIbα-Tg mice transfused with wild type platelets, suggesting a platelet-specific role of MCUb in thrombosis. MCUb-/- stimulated platelets exhibited reduced glucose uptake, decreased glycolytic rate, and lowered pyruvate dehydrogenase phosphorylation, suggesting that mitochondrial Ca2+ mediates bioenergetic changes in platelets. CONCLUSION Our findings suggest that mitochondrial Ca2+ signaling and glucose oxidation are functionally linked in activated platelets and reveal a novel role of MCUb in platelet activation and arterial thrombosis.
Collapse
Affiliation(s)
- Madankumar Ghatge
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA.
| | - Manasa K Nayak
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Gagan D Flora
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Mariia Kumskova
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Aditi Jain
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Rakesh B Patel
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Zhihong Lin
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | - Yuriy M Usachev
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | - Anil K Chauhan
- Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA.
| |
Collapse
|
33
|
Ekhlak M, Kulkarni PP, Singh V, Chaurasia SN, Mohapatra SK, Chaurasia RN, Dash D. Necroptosis executioner MLKL plays pivotal roles in agonist-induced platelet prothrombotic responses and lytic cell death in a temporal order. Cell Death Differ 2023; 30:1886-1899. [PMID: 37301927 PMCID: PMC10406901 DOI: 10.1038/s41418-023-01181-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Necroptosis is a form of programmed cell death executed by receptor-interacting serine/threonine protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like (MLKL). Platelets are circulating cells that play central roles in haemostasis and pathological thrombosis. In this study we demonstrate seminal contribution of MLKL in transformation of agonist-stimulated platelets to active haemostatic units progressing eventually to necrotic death on a temporal scale, thus attributing a yet unrecognized fundamental role to MLKL in platelet biology. Physiological agonists like thrombin instigated phosphorylation and subsequent oligomerization of MLKL in platelets in a RIPK3-independent but phosphoinositide 3-kinase (PI3K)/AKT-dependent manner. Inhibition of MLKL significantly curbed agonist-induced haemostatic responses in platelets that included platelet aggregation, integrin activation, granule secretion, procoagulant surface generation, rise in intracellular calcium, shedding of extracellular vesicles, platelet-leukocyte interactions and thrombus formation under arterial shear. MLKL inhibition, too, prompted impairment in mitochondrial oxidative phosphorylation and aerobic glycolysis in stimulated platelets, accompanied with disruption in mitochondrial transmembrane potential, augmented proton leak and drop in both mitochondrial calcium as well as ROS. These findings underscore the key role of MLKL in sustaining OXPHOS and aerobic glycolysis that underlie energy-intensive platelet activation responses. Prolonged exposure to thrombin provoked oligomerization and translocation of MLKL to plasma membranes forming focal clusters that led to progressive membrane permeabilization and decline in platelet viability, which was prevented by inhibitors of PI3K/MLKL. In summary, MLKL plays vital role in transitioning of stimulated platelets from relatively quiescent cells to functionally/metabolically active prothrombotic units and their ensuing progression to necroptotic death.
Collapse
Affiliation(s)
- Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Paresh P Kulkarni
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Vipin Singh
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Susheel N Chaurasia
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | | | - Rameshwar Nath Chaurasia
- Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
34
|
Ravera S, Signorello MG, Panfoli I. Platelet Metabolic Flexibility: A Matter of Substrate and Location. Cells 2023; 12:1802. [PMID: 37443836 PMCID: PMC10340290 DOI: 10.3390/cells12131802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Platelets are cellular elements that are physiologically involved in hemostasis, inflammation, thrombotic events, and various human diseases. There is a link between the activation of platelets and their metabolism. Platelets possess considerable metabolic versatility. Although the role of platelets in hemostasis and inflammation is known, our current understanding of platelet metabolism in terms of substrate preference is limited. Platelet activation triggers an oxidative metabolism increase to sustain energy requirements better than aerobic glycolysis alone. In addition, platelets possess extra-mitochondrial oxidative phosphorylation, which could be one of the sources of chemical energy required for platelet activation. This review aims to provide an overview of flexible platelet metabolism, focusing on the role of metabolic compartmentalization in substrate preference, since the metabolic flexibility of stimulated platelets could depend on subcellular localization and functional timing. Thus, developing a detailed understanding of the link between platelet activation and metabolic changes is crucial for improving human health.
Collapse
Affiliation(s)
- Silvia Ravera
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy;
| | | | - Isabella Panfoli
- Department of Pharmacy (DIFAR), University of Genoa, 16132 Genoa, Italy;
| |
Collapse
|
35
|
Kulkarni PP, Ekhlak M, Dash D. Energy metabolism in platelets fuels thrombus formation: Halting the thrombosis engine with small-molecule modulators of platelet metabolism. Metabolism 2023:155596. [PMID: 37244415 DOI: 10.1016/j.metabol.2023.155596] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
Platelets are circulating cells central to haemostasis that follows vessel injury, as well as thrombosis that ensues as a consequence of pathological stasis or plaque rupture. Platelet responses to various stimuli that mediate these processes are all energy-intensive. Hence, platelets need to adapt their energy metabolism to fulfil the requirements of clot formation while overcoming the adversities of the thrombus niche such as restricted access to oxygen and nutrient. In the present review, we describe the changes in energy metabolism of platelets upon agonist challenge and their underlying molecular mechanisms. We briefly discuss the metabolic flexibility and dependency of stimulated platelets in terms of choice of energy substrates. Finally, we discuss how targeting the metabolic vulnerabilities of stimulated platelets such as aerobic glycolysis and/or beta oxidation of fatty acids could forestall platelet activation and thrombus formation. Thus, we present a case for modulating platelet energy metabolism using small-molecules as a novel anti-platelet strategy in the management of vaso-occlusive disorders like acute myocardial infarction, ischemic stroke, deep vein thrombosis and pulmonary embolism.
Collapse
Affiliation(s)
- Paresh P Kulkarni
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India; Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, USA.
| | - Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
36
|
Li H, Chen J, Xing X, Lou D. Association of lactate detection with in-hospital mortality in critically ill patients with acute myocardial infarction: a retrospective cohort study. BMJ Open 2023; 13:e069129. [PMID: 37085300 PMCID: PMC10124257 DOI: 10.1136/bmjopen-2022-069129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/23/2023] Open
Abstract
OBJECTIVES To assess the associations of lactate level or lactate clearance at different time points with in-hospital mortality in critically ill patients with acute myocardial infarction (AMI). DESIGN A cohort study. SETTING The Medical Information Mart for Intensive Care III database. PARTICIPANT 490 AMI patients. INTERVENTION None. PRIMARY AND SECONDARY OUTCOME MEASURES In-hospital mortality of patients. RESULTS In total, 120 (24.49%) patients died at the end of follow-up. After adjusting for confounders, increased risk of in-hospital mortality in patients with AMI was observed in those with high lactate level (24 hours) (HR=1.156, 95%CI: 1.002 to 1.333). Increased lactate clearance (24 hours) was correlated with a decreased risk of in-hospital mortality in patients with AMI (HR=0.995, 95% CI: 0.994 to 0.997). The area under the curves (AUCs) of lactate level (24 hours) and lactate clearance (24 hours) were 0.689 (95% CI: 0.655 to 0.723) and 0.672 (95% CI: 0.637 to 0.706), respectively. The AUC of lactate level (24 hours) and lactate clearance (24 hours) was higher than lactate level (baseline). CONCLUSIONS Increased lactate level (24 hours) was associated with an elevated risk of in-hospital mortality in patients with AMI and increased lactate clearance (24 hours) was correlated with a decreased risk of in-hospital mortality in patients with AMI despite the age and genders.
Collapse
Affiliation(s)
- Hongwu Li
- Department of Cardiology, Peking Union Medical College Hospital, Dongcheng-qu, Beijing, China
| | - Jingyi Chen
- Emergency Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyue Xing
- Emergency Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danfei Lou
- Emergency Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
37
|
Tofovic SP. Purine Nucleoside Phosphorylase: A New Pharmacological Target in Sickle Cell Disease and Hemolytic Vasculopathy. Med Hypotheses 2023. [DOI: 10.1016/j.mehy.2023.111045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
|
38
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
39
|
Kulkarni PP, Ekhlak M, Singh V, Kailashiya V, Singh N, Dash D. Fatty acid oxidation fuels agonist-induced platelet activation and thrombus formation: Targeting β-oxidation of fatty acids as an effective anti-platelet strategy. FASEB J 2023; 37:e22768. [PMID: 36624703 DOI: 10.1096/fj.202201321rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023]
Abstract
Platelet mitochondria possess remarkable plasticity for oxidation of energy substrates, where metabolic dependency on glucose or fatty acids is higher than glutamine. Since platelets metabolize nearly the entire pool of glucose to lactate rather than fluxing through mitochondrial tricarboxylic acid cycle, we posit that majority of mitochondrial ATP, which is essential for platelet granule secretion and thrombus formation, is sourced from oxidation of fatty acids. We performed a comprehensive analysis of bioenergetics and function of stimulated platelets in the presence of etomoxir, trimetazidine and oxfenicine, three pharmacologically distinct inhibitors of β-oxidation. Each of them significantly impaired oxidative phosphorylation in unstimulated as well as thrombin-stimulated platelets leading to a small but consistent drop in ATP level in activated cells due to a lack of compensation from glycolytic ATP. Trimetazidine and oxfenicine attenuated platelet aggregation, P-selectin externalization and integrin αIIb β3 activation. Both etomoxir and trimetazidine impeded agonist-induced dense granule release and platelet thrombus formation on collagen under arterial shear. The effect of inhibitors on platelet aggregation and dense granule release was dose- and incubation time- dependent with significant inhibition at higher doses and prolonged incubation times. Neither of the inhibitors could protect mice from collagen-epinephrine-induced pulmonary embolism or prolong mouse tail bleeding times. However, mice pre-administered with etomoxir, trimetazidine and oxfenicine were protected from ferric chloride-induced mesenteric thrombosis. In conclusion, β-oxidation of fatty acids sustains ATP level in stimulated platelets and is therefore essential for energy-intensive agonist-induced platelet responses. Thus, fatty acid oxidation may constitute an attractive therapeutic target for novel antiplatelet agents.
Collapse
Affiliation(s)
- Paresh P Kulkarni
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vipin Singh
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vikas Kailashiya
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Nitesh Singh
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
40
|
Tumor Microenvironment and Metabolism: Role of the Mitochondrial Melatonergic Pathway in Determining Intercellular Interactions in a New Dynamic Homeostasis. Int J Mol Sci 2022; 24:ijms24010311. [PMID: 36613754 PMCID: PMC9820362 DOI: 10.3390/ijms24010311] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
There is a growing interest in the role of alterations in mitochondrial metabolism in the pathoetiology and pathophysiology of cancers, including within the array of diverse cells that can form a given tumor microenvironment. The 'exhaustion' in natural killer cells and CD8+ t cells as well as the tolerogenic nature of dendritic cells in the tumor microenvironment seems determined by variations in mitochondrial function. Recent work has highlighted the important role played by the melatonergic pathway in optimizing mitochondrial function, limiting ROS production, endogenous antioxidants upregulation and consequent impacts of mitochondrial ROS on ROS-dependent microRNAs, thereby impacting on patterned gene expression. Within the tumor microenvironment, the tumor, in a quest for survival, seeks to 'dominate' the dynamic intercellular interactions by limiting the capacity of cells to optimally function, via the regulation of their mitochondrial melatonergic pathway. One aspect of this is the tumor's upregulation of kynurenine and the activation of the aryl hydrocarbon receptor, which acts to metabolize melatonin and increase the N-acetylserotonin/melatonin ratio, with effluxed N-acetylserotonin acting as a brain-derived neurotrophic factor (BDNF) mimic via its activation of the BDNF receptor, TrkB, thereby increasing the survival and proliferation of tumors and cancer stem-like cells. This article highlights how many of the known regulators of cells in the tumor microenvironment can be downstream of the mitochondrial melatonergic pathway regulation. Future research and treatment implications are indicated.
Collapse
|
41
|
Nunn AVW, Guy GW, Brysch W, Bell JD. Understanding Long COVID; Mitochondrial Health and Adaptation-Old Pathways, New Problems. Biomedicines 2022; 10:3113. [PMID: 36551869 PMCID: PMC9775339 DOI: 10.3390/biomedicines10123113] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/04/2022] Open
Abstract
Many people infected with the SARS-CoV-2 suffer long-term symptoms, such as "brain fog", fatigue and clotting problems. Explanations for "long COVID" include immune imbalance, incomplete viral clearance and potentially, mitochondrial dysfunction. As conditions with sub-optimal mitochondrial function are associated with initial severity of the disease, their prior health could be key in resistance to long COVID and recovery. The SARs virus redirects host metabolism towards replication; in response, the host can metabolically react to control the virus. Resolution is normally achieved after viral clearance as the initial stress activates a hormetic negative feedback mechanism. It is therefore possible that, in some individuals with prior sub-optimal mitochondrial function, the virus can "tip" the host into a chronic inflammatory cycle. This might explain the main symptoms, including platelet dysfunction. Long COVID could thus be described as a virally induced chronic and self-perpetuating metabolically imbalanced non-resolving state characterised by mitochondrial dysfunction, where reactive oxygen species continually drive inflammation and a shift towards glycolysis. This would suggest that a sufferer's metabolism needs to be "tipped" back using a stimulus, such as physical activity, calorie restriction, or chemical compounds that mimic these by enhancing mitochondrial function, perhaps in combination with inhibitors that quell the inflammatory response.
Collapse
Affiliation(s)
- Alistair V. W. Nunn
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London W1W 6UW, UK
| | - Geoffrey W. Guy
- The Guy Foundation, Chedington Court, Beaminster, Dorset DT8 3HY, UK
| | | | - Jimmy D. Bell
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London W1W 6UW, UK
| |
Collapse
|
42
|
Modulation of Glycoprotein VI and Its Downstream Signaling Pathways as an Antiplatelet Target. Int J Mol Sci 2022; 23:ijms23179882. [PMID: 36077280 PMCID: PMC9456422 DOI: 10.3390/ijms23179882] [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: 05/16/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
Antiplatelet therapy aims to reduce the risk of thrombotic events while maintaining hemostasis. A promising current approach is the inhibition of platelet glycoprotein GPVI-mediated adhesion pathways; pathways that do not involve coagulation. GPVI is a signaling receptor integral for collagen-induced platelet activation and participates in the thrombus consolidation process, being a suitable target for thrombosis prevention. Considering this, the blocking or antibody-mediated depletion of GPVI is a promising antiplatelet therapy for the effective and safe treatment of thrombotic diseases without a significant risk of bleeding and impaired hemostatic plug formation. This review describes the current knowledge concerning pharmaceutical approaches to platelet GPVI modulation and its downstream signaling pathways in this context.
Collapse
|
43
|
Ribeiro HC, Sen P, Dickens A, Santa Cruz EC, Orešič M, Sussulini A. Metabolomic and proteomic profiling in bipolar disorder patients revealed potential molecular signatures related to hemostasis. Metabolomics 2022; 18:65. [PMID: 35922643 DOI: 10.1007/s11306-022-01924-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/19/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Bipolar disorder (BD) is a mood disorder characterized by the occurrence of depressive episodes alternating with episodes of elevated mood (known as mania). There is also an increased risk of other medical comorbidities. OBJECTIVES This work uses a systems biology approach to compare BD treated patients with healthy controls (HCs), integrating proteomics and metabolomics data using partial correlation analysis in order to observe the interactions between altered proteins and metabolites, as well as proposing a potential metabolic signature panel for the disease. METHODS Data integration between proteomics and metabolomics was performed using GC-MS data and label-free proteomics from the same individuals (N = 13; 5 BD, 8 HC) using generalized canonical correlation analysis and partial correlation analysis, and then building a correlation network between metabolites and proteins. Ridge-logistic regression models were developed to stratify between BD and HC groups using an extended metabolomics dataset (N = 28; 14 BD, 14 HC), applying a recursive feature elimination for the optimal selection of the metabolites. RESULTS Network analysis demonstrated links between proteins and metabolites, pointing to possible alterations in hemostasis of BD patients. Ridge-logistic regression model indicated a molecular signature comprising 9 metabolites, with an area under the receiver operating characteristic curve (AUROC) of 0.833 (95% CI 0.817-0.914). CONCLUSION From our results, we conclude that several metabolic processes are related to BD, which can be considered as a multi-system disorder. We also demonstrate the feasibility of partial correlation analysis for integration of proteomics and metabolomics data in a case-control study setting.
Collapse
Affiliation(s)
- Henrique Caracho Ribeiro
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Institute of Chemistry, University of Campinas, PO Box 6154, Campinas, SP, 13083-970, Brazil
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland
| | - Partho Sen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland
- School of Medical Sciences, Örebro University, 702 81, Örebro, Sweden
| | - Alex Dickens
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland
- Department of Chemistry, University of Turku, 20520, Turku, Finland
| | - Elisa Castañeda Santa Cruz
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Institute of Chemistry, University of Campinas, PO Box 6154, Campinas, SP, 13083-970, Brazil
| | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland
- School of Medical Sciences, Örebro University, 702 81, Örebro, Sweden
| | - Alessandra Sussulini
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Institute of Chemistry, University of Campinas, PO Box 6154, Campinas, SP, 13083-970, Brazil.
- Instituto Nacional de Ciência e Tecnologia de Bioanalítica (INCTBio), Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, 13083-970, Brazil.
| |
Collapse
|
44
|
Targeted analysis of sugar phosphates from glycolysis pathway by phosphate methylation with liquid chromatography coupled to tandem mass spectrometry. Anal Chim Acta 2022; 1221:340099. [DOI: 10.1016/j.aca.2022.340099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 11/18/2022]
|
45
|
Non-canonical Sonic Hedgehog signaling amplifies platelet reactivity and thrombogenicity. Blood Adv 2022; 6:5024-5040. [PMID: 35704688 PMCID: PMC9631642 DOI: 10.1182/bloodadvances.2021006560] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 05/19/2022] [Indexed: 11/20/2022] Open
Abstract
Sonic Hedgehog signaling amplifies platelet activation. Targeting Shh signaling attenuates hemostasis and thrombosis.
Sonic Hedgehog (Shh) is a morphogen in vertebrate embryos that is also associated with organ homeostasis in adults. We report here that human platelets, though enucleate, synthesize Shh from preexisting mRNAs upon agonist stimulation, and mobilize it for surface expression and release on extracellular vesicles, thus alluding to its putative role in platelet activation. Shh, in turn, induced a wave of noncanonical signaling in platelets leading to activation of small GTPase Ras homolog family member A and phosphorylation of myosin light chain in activated protein kinase-dependent manner. Remarkably, agonist-induced thrombogenic responses in platelets, which include platelet aggregation, granule secretion, and spreading on immobilized fibrinogen, were significantly attenuated by inhibition of Hedgehog signaling, thus, implicating inputs from Shh in potentiation of agonist-mediated platelet activation. In consistence, inhibition of the Shh pathway significantly impaired arterial thrombosis in mice. Taken together, the above observations strongly support a feed-forward loop of platelet stimulation triggered locally by Shh, similar to ADP and thromboxane A2, that contributes significantly to the stability of occlusive arterial thrombus and that can be investigated as a potential therapeutic target in thrombotic disorders.
Collapse
|
46
|
Yang K, Holt M, Fan M, Lam V, Yang Y, Ha T, Williams DL, Li C, Wang X. Cardiovascular Dysfunction in COVID-19: Association Between Endothelial Cell Injury and Lactate. Front Immunol 2022; 13:868679. [PMID: 35401579 PMCID: PMC8984030 DOI: 10.3389/fimmu.2022.868679] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/01/2022] [Indexed: 12/27/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), an infectious respiratory disease propagated by a new virus known as Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), has resulted in global healthcare crises. Emerging evidence from patients with COVID-19 suggests that endothelial cell damage plays a central role in COVID-19 pathogenesis and could be a major contributor to the severity and mortality of COVID-19. Like other infectious diseases, the pathogenesis of COVID-19 is closely associated with metabolic processes. Lactate, a potential biomarker in COVID-19, has recently been shown to mediate endothelial barrier dysfunction. In this review, we provide an overview of cardiovascular injuries and metabolic alterations caused by SARS-CoV-2 infection. We also propose that lactate plays a potential role in COVID-19-driven endothelial cell injury.
Collapse
Affiliation(s)
- Kun Yang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Matthew Holt
- James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Min Fan
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Victor Lam
- College of Arts and Science, New York University, New York City, NY, United States
| | - Yong Yang
- James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Tuanzhu Ha
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - David L. Williams
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Chuanfu Li
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Xiaohui Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
47
|
Sake CL, Metcalf AJ, Meagher M, Paola JD, Neeves KB, Boyle NR. Isotopically nonstationary 13C metabolic flux analysis in resting and activated human platelets. Metab Eng 2022; 69:313-322. [PMID: 34954086 PMCID: PMC8905147 DOI: 10.1016/j.ymben.2021.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 01/03/2023]
Abstract
Platelet metabolism is linked to platelet hyper- and hypoactivity in numerous human diseases. Developing a detailed understanding of the link between metabolic shifts and platelet activation state is integral to improving human health. Here, we show the first application of isotopically nonstationary 13C metabolic flux analysis to quantitatively measure carbon fluxes in both resting and thrombin activated platelets. Metabolic flux analysis results show that resting platelets primarily metabolize glucose to lactate via glycolysis, while acetate is oxidized to fuel the tricarboxylic acid cycle. Upon activation with thrombin, a potent platelet agonist, platelets increase their uptake of glucose 3-fold. This results in an absolute increase in flux throughout central metabolism, but when compared to resting platelets they redistribute carbon dramatically. Activated platelets decrease relative flux to the oxidative pentose phosphate pathway and TCA cycle from glucose and increase relative flux to lactate. These results provide the first report of reaction-level carbon fluxes in platelets and allow us to distinguish metabolic fluxes with much higher resolution than previous studies.
Collapse
Affiliation(s)
- Cara L. Sake
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO, 80401 USA
| | - Alexander J. Metcalf
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO, 80401 USA
| | - Michelle Meagher
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jorge Di Paola
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Keith B. Neeves
- Department of Bioengineering, University of Colorado, Aurora, CO, 80045 USA,Hemophilia and Thrombosis Center, University of Colorado, Aurora, CO, 80045 USA,Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant University of Colorado, Aurora, CO, 80045 USA
| | - Nanette R. Boyle
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO, 80401 USA,Correspondence: , 423 Alderson Hall; 1613 Illinois St.; Golden, CO 80401
| |
Collapse
|
48
|
Barrett TJ, Bilaloglu S, Cornwell M, Burgess HM, Virginio VW, Drenkova K, Ibrahim H, Yuriditsky E, Aphinyanaphongs Y, Lifshitz M, Xia Liang F, Alejo J, Smith G, Pittaluga S, Rapkiewicz AV, Wang J, Iancu-Rubin C, Mohr I, Ruggles K, Stapleford KA, Hochman J, Berger JS. Platelets contribute to disease severity in COVID-19. J Thromb Haemost 2021; 19:3139-3153. [PMID: 34538015 PMCID: PMC8646651 DOI: 10.1111/jth.15534] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/27/2021] [Accepted: 09/16/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Heightened inflammation, dysregulated immunity, and thrombotic events are characteristic of hospitalized COVID-19 patients. Given that platelets are key regulators of thrombosis, inflammation, and immunity they represent prime candidates as mediators of COVID-19-associated pathogenesis. The objective of this study was to understand the contribution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to the platelet phenotype via phenotypic (activation, aggregation) and transcriptomic characterization. APPROACH AND RESULTS In a cohort of 3915 hospitalized COVID-19 patients, we analyzed blood platelet indices collected at hospital admission. Following adjustment for demographics, clinical risk factors, medication, and biomarkers of inflammation and thrombosis, we find platelet count, size, and immaturity are associated with increased critical illness and all-cause mortality. Bone marrow, lung tissue, and blood from COVID-19 patients revealed the presence of SARS-CoV-2 virions in megakaryocytes and platelets. Characterization of COVID-19 platelets found them to be hyperreactive (increased aggregation, and expression of P-selectin and CD40) and to have a distinct transcriptomic profile characteristic of prothrombotic large and immature platelets. In vitro mechanistic studies highlight that the interaction of SARS-CoV-2 with megakaryocytes alters the platelet transcriptome, and its effects are distinct from the coronavirus responsible for the common cold (CoV-OC43). CONCLUSIONS Platelet count, size, and maturity associate with increased critical illness and all-cause mortality among hospitalized COVID-19 patients. Profiling tissues and blood from COVID-19 patients revealed that SARS-CoV-2 virions enter megakaryocytes and platelets and associate with alterations to the platelet transcriptome and activation profile.
Collapse
Affiliation(s)
- Tessa J Barrett
- Department of Medicine, New York University Langone Health, New York, New York, USA
| | - Seda Bilaloglu
- Department of Population Health, New York University Langone Health, New York, New York, USA
| | - Macintosh Cornwell
- Department of Medicine, New York University Langone Health, New York, New York, USA
| | - Hannah M Burgess
- Department of Microbiology, New York University Langone Health, New York, New York, USA
| | - Vitor W Virginio
- Department of Medicine, New York University Langone Health, New York, New York, USA
| | - Kamelia Drenkova
- Department of Medicine, New York University Langone Health, New York, New York, USA
| | - Homam Ibrahim
- Department of Medicine, New York University Langone Health, New York, New York, USA
| | - Eugene Yuriditsky
- Department of Medicine, New York University Langone Health, New York, New York, USA
| | - Yin Aphinyanaphongs
- Department of Population Health, New York University Langone Health, New York, New York, USA
| | - Mark Lifshitz
- Department of Pathology, New York University Langone Health, New York, New York, USA
| | - Feng Xia Liang
- DART Microscopy Laboratory, New York University Langone Health, New York, New York, USA
| | - Julie Alejo
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Grace Smith
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stefania Pittaluga
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Amy V Rapkiewicz
- Department of Pathology, NYU Winthrop Hospital, New York University Langone Health, Mineola, New York, USA
| | - Jun Wang
- Department of Pathology, New York University Langone Health, New York, New York, USA
| | - Camelia Iancu-Rubin
- Department of Pathology, Molecular and Cell-Based Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ian Mohr
- Department of Microbiology, New York University Langone Health, New York, New York, USA
| | - Kelly Ruggles
- Department of Medicine, New York University Langone Health, New York, New York, USA
| | - Kenneth A Stapleford
- Department of Microbiology, New York University Langone Health, New York, New York, USA
| | - Judith Hochman
- Department of Medicine, New York University Langone Health, New York, New York, USA
| | - Jeffrey S Berger
- Department of Medicine, New York University Langone Health, New York, New York, USA
- Department of Surgery, New York University Langone Health, New York, New York, USA
| |
Collapse
|
49
|
Nayak MK, Ghatge M, Flora GD, Dhanesha N, Jain M, Markan KR, Potthoff MJ, Lentz SR, Chauhan AK. The metabolic enzyme pyruvate kinase M2 regulates platelet function and arterial thrombosis. Blood 2021; 137:1658-1668. [PMID: 33027814 PMCID: PMC7995287 DOI: 10.1182/blood.2020007140] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/23/2020] [Indexed: 02/08/2023] Open
Abstract
Very little is known about the role of metabolic regulatory mechanisms in platelet activation and thrombosis. Dimeric pyruvate kinase M2 (PKM2) is a crucial regulator of aerobic glycolysis that facilitates the production of lactate and metabolic reprogramming. Herein, we report that limiting PKM2 dimer formation, using the small molecule inhibitor ML265, negatively regulates lactate production and glucose uptake in human and murine stimulated platelets. Furthermore, limiting PKM2 dimer formation reduced agonist-induced platelet activation, aggregation, clot retraction, and thrombus formation under arterial shear stress in vitro in both human and murine platelets. Mechanistically, limiting PKM2 dimerization downregulated phosphatidylinositol 3-kinase (PI3K)-mediated protein kinase B or serine/threonine-specific protein kinase (Akt)/glycogen synthase kinase 3 (GSK3) signaling in human and murine platelets. To provide further evidence for the role of PKM2 in platelet function, we generated a megakaryocyte or platelet-specific PKM2-/- mutant strain (PKM2fl/flPF4Cre+). Platelet-specific PKM2-deficient mice exhibited impaired agonist-induced platelet activation, aggregation, clot retraction, and PI3K-mediated Akt/GSK3 signaling and were less susceptible to arterial thrombosis in FeCl3 injury-induced carotid- and laser injury-induced mesenteric artery thrombosis models, without altering hemostasis. Wild-type mice treated with ML265 were less susceptible to arterial thrombosis with unaltered tail bleeding times. These findings reveal a major role for PKM2 in coordinating multiple aspects of platelet function, from metabolism to cellular signaling to thrombosis, and implicate PKM2 as a potential target for antithrombotic therapeutic intervention.
Collapse
Affiliation(s)
- Manasa K Nayak
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Madankumar Ghatge
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Gagan D Flora
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Nirav Dhanesha
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Manish Jain
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Kathleen R Markan
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA; and
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA; and
- Department of Veterans Affairs Medical Center, Iowa City, IA
| | - Steven R Lentz
- Department of Internal Medicine, Division of Hematology/Oncology, and
| | - Anil K Chauhan
- Department of Internal Medicine, Division of Hematology/Oncology, and
| |
Collapse
|
50
|
Schuster S, Ewald J, Kaleta C. Modeling the energy metabolism in immune cells. Curr Opin Biotechnol 2021; 68:282-291. [PMID: 33770632 DOI: 10.1016/j.copbio.2021.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/16/2021] [Accepted: 03/01/2021] [Indexed: 02/08/2023]
Abstract
In this review, we summarize and briefly discuss various approaches to modeling the metabolism in human immune cells, with a focus on energy metabolism. These approaches include metabolic reconstruction, elementary modes, and flux balance analysis, which are often subsumed under constraint-based modeling. Further approaches are evolutionary game theory and kinetic modeling. Many immune cells such as macrophages show the Warburg effect, meaning that glycolysis is upregulated upon activation. We outline a minimal model for explaining that effect using optimization. The effect of a confrontation with pathogen cells on immunometabolism is highlighted. Models describing the differences between M1 and M2 macrophages, ROS production in neutrophils, and tryptophan metabolism are discussed. Obstacles and future prospects are outlined.
Collapse
Affiliation(s)
- Stefan Schuster
- Department of Bioinformatics, Matthias Schleiden Institute, Friedrich Schiller University Jena, Ernst-Abbe-Pl. 2, 07743 Jena, Germany.
| | - Jan Ewald
- Department of Bioinformatics, Matthias Schleiden Institute, Friedrich Schiller University Jena, Ernst-Abbe-Pl. 2, 07743 Jena, Germany
| | - Christoph Kaleta
- Medical Systems Biology Group, Institute of Experimental Medicine, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|