1
|
He SS, Huang XD, Zhang SZ, Zhuang QQ, Chen XX, Wang HD, Mao XL. A prospective cohort study on prognostic implications of serum platelet derived microparticles levels in acute cerebral infarction. Sci Rep 2025; 15:14197. [PMID: 40268995 PMCID: PMC12018963 DOI: 10.1038/s41598-025-92732-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 03/03/2025] [Indexed: 04/25/2025] Open
Abstract
Platelet-derived microparticles (PDMPs) participate in ischemic brain injury. We further determined the relationships between serum PDMPs levels and early neurological deterioration (END) as well as functional outcome after acute cerebral infarction (ACI). In this prospective cohort study, serum PDMPs levels were measured in 125 controls and 621 patients with ACI. Univariate analysis and multivariate analysis were sequentially applied to investigate the relations of serum PDMPs levels to END and poor prognosis (modified Rankin scale score > 2) at six months after ACI. Serum PDMPs levels were significantly higher in patients than in controls (median, 14.00 ng/L vs. 27.00 ng/L; P < 0.001). Serum PDMPs levels were strongly correlated with infarction volume (ρ = 0.532, P < 0.001), National Institutes of Health Stroke Scale score (ρ = 0.627, P < 0.001) and modified Rankin scale score (ρ = 0.528, P < 0.001). It was independently associated with END [odds ratio (OR) 1.117, 95% confidence interval (CI) 1.008-1.238; P = 0.001] and poor prognosis (OR 1.092, 95% CI 1.066-1.119; P = 0.001). There were linear relationships between serum PDMPs levels and risks of poor prognosis (P for non-linear = 0.055) plus END (P for non-linear = 0.061) under restricted cubic spline. Using subgroup analysis, significant interaction existed between serum PDMPs levels and age in association of poor prognosis (P for interaction = 0.006), as well as between serum PDMPs levels and coronary heart disease in association of END (P for interaction = 0.017). Serum PDMPs levels significantly discriminated the development of poor prognosis (Area under curve 0.705, 95% CI 0.632-0.778; P < 0.001) and END (The area 0.733, 95% CI 0.664-0.803; P < 0.001). Serum PDMPs levels may predict the risk of END and 6-month poor prognosis in patients with ACI.
Collapse
Affiliation(s)
- Si-Si He
- Department of Neurology, The Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, 252 Bailidong Road, Wenzhou, 325000, China
- Panvascular Disease Management Center, The Wenzhou Central Hospital, 252 Bailidong Road, Wenzhou, 325000, China
| | - Xiang-Dong Huang
- Department of Neurology, The Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, 252 Bailidong Road, Wenzhou, 325000, China
- Panvascular Disease Management Center, The Wenzhou Central Hospital, 252 Bailidong Road, Wenzhou, 325000, China
| | - Shi-Zheng Zhang
- Department of Neurology, The Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, 252 Bailidong Road, Wenzhou, 325000, China
- Panvascular Disease Management Center, The Wenzhou Central Hospital, 252 Bailidong Road, Wenzhou, 325000, China
| | - Qing-Qing Zhuang
- Department of Neurology, The Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, 252 Bailidong Road, Wenzhou, 325000, China
- Panvascular Disease Management Center, The Wenzhou Central Hospital, 252 Bailidong Road, Wenzhou, 325000, China
| | - Xin-Xin Chen
- Department of Neurology, The Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, 252 Bailidong Road, Wenzhou, 325000, China
- Panvascular Disease Management Center, The Wenzhou Central Hospital, 252 Bailidong Road, Wenzhou, 325000, China
| | - Hao-Dong Wang
- Department of Neurology, The Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, 252 Bailidong Road, Wenzhou, 325000, China
- Panvascular Disease Management Center, The Wenzhou Central Hospital, 252 Bailidong Road, Wenzhou, 325000, China
| | - Xin-Lei Mao
- Department of Neurology, The Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, 252 Bailidong Road, Wenzhou, 325000, China.
- Panvascular Disease Management Center, The Wenzhou Central Hospital, 252 Bailidong Road, Wenzhou, 325000, China.
| |
Collapse
|
2
|
Liu P, Yuan X, Liu Y, Guo M, Xu L, Dong Q, Fan T, Lv Y, Pei X, Qu M, Yue W, Xie X. RHOB regulates megakaryocytic and erythroid differentiation by altering the cell cycle and cytoskeleton. Sci Rep 2025; 15:13159. [PMID: 40240762 PMCID: PMC12003728 DOI: 10.1038/s41598-025-95946-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
RHOB, a member of the RHO GTPase family, is a target of miR-1915-3p. miR-1915-3p was proven to promote the differentiation of megakaryocytes while inhibiting that of the erythroid lineage. Although RHOB has been shown to be involved in proplatelet production in mice, its role in early haematopoiesis, especially megakaryocytic and erythroid differentiation in humans, has not yet been elucidated. In this study, we demonstrated the function of RHOB by knocking down the gene in progenitor cells and inducing these cells to differentiate into erythroid or megakaryocytic cells. The silencing of RHOB significantly decreased the proportion of erythroid cells and inhibited the expression of related genes during erythroid differentiation while increasing the proportion of megakaryocytic and polyploid cells and promoting the expression of megakaryocytic lineage-specific genes in megakaryocyte-induced cells. RNA sequencing indicated that RHOB is involved in differentiation by regulating the cell cycle and cytoskeleton. Accordingly, we found that RHOB could differentially affect the expression of cyclin-dependent kinases and cyclins to alter the proportions of G0/G1-or S-phase cells and affect the expression or aggregation of beta-actin (ACTB) during erythroid or megakaryocytic differentiation. Directly overexpressing RHOA/RHOC in RHOB-downregulated cells showed that the effect of RHOB could be moderately compensated for by its homologs RHOA or RHOC. Taken together, these findings show that RHOB plays an important role in megakaryocytic and erythroid differentiation via the cell cycle and cytoskeleton regulation.
Collapse
Affiliation(s)
- Pengcong Liu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xin Yuan
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yiming Liu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Meishan Guo
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Lei Xu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Qian Dong
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Tao Fan
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yang Lv
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xuetao Pei
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Mingyi Qu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Wen Yue
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Xiaoyan Xie
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
3
|
Pilard M, Babran S, Martel C. Regulation of Platelet Function by HDL. Arterioscler Thromb Vasc Biol 2025. [PMID: 40207365 DOI: 10.1161/atvbaha.124.318260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Over the past decade, increasing the capacity of HDL (high-density lipoprotein) cholesterol to mediate macrophage reverse cholesterol transport has been a target of interest in the treatment of cardiovascular diseases (CVDs). However, clinical studies reporting the limited efficacy of HDL or its main apolipoprotein, APOA1, in reducing cardiovascular events have emerged. Although HDL cholesterol is unlikely to play a direct causal role in CVD, its inverse, albeit modest, association with CVD risk, consistently observed in large population studies, suggests it may influence alternative pathways beyond cholesterol metabolism. Given the diverse functions of HDL and its components, it is conceivable that its impact on CVD occurs through less direct mechanisms. A potential hypothesis is that HDL modulates platelet function, a crucial player in the initiation and progression of atherothrombosis, which may contribute to its observed relationship with CVD risk. In this review, we focus on how HDL and its components, with an emphasis on APOA1, interact with platelets (and their precursors or activation products) to modulate atherothrombotic responses.
Collapse
Affiliation(s)
- Marion Pilard
- Department of Medicine, Faculty of Medicine, Université de Montréal, Canada
- Montreal Heart Institute, Canada
| | - Sara Babran
- Department of Medicine, Faculty of Medicine, Université de Montréal, Canada
- Montreal Heart Institute, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Canada
- Montreal Heart Institute, Canada
| |
Collapse
|
4
|
Ma Y, Meng F, Lin Z, Chen Y, Lan T, Yang Z, Diao R, Zhang X, Chen Q, Zhang C, Tian Y, Li C, Fang W, Liang X, Zhang X. Bioengineering Platelets Presenting PD-L1, Galectin-9 and BTLA to Ameliorate Type 1 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2501139. [PMID: 40019367 PMCID: PMC12021092 DOI: 10.1002/advs.202501139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/14/2025] [Indexed: 03/01/2025]
Abstract
Autoimmune destruction of pancreatic β-cells leads to impaired insulin production and onset of type 1 diabetes (T1D). Hence, immunomodulation of pancreas-infiltrated immune cells especially the β-cells autoreactive-T cells is a promising way to hinder and reverse the progress of T1D. Herein, megakaryocytes are primed with interferon-γ (IFN-γ) to produce platelets presenting high levels of immunosuppressive checkpoint ligands including programmed death-ligand 1 (PD-L1), Programmed Death-Ligand 2 (PD-L2), the B and T lymphocyte attenuator (BTLA) and Galectin-9 (Gal-9), termed as IFN-γ platelets. The IFN-γ platelets bound and interacted with T cells through immune checkpoint ligands and receptors, which efficaciously induced T cell exhaustion and apoptosis in vitro. Virtually, NOD diabetes mice received IFN-γ platelets treatments prominently preserved β-cell integrity and insulin production, ultimately hindering the progress to hyperglycemia. Intriguingly, both the amount and activity of the pancreas infiltrate-T cells intensively reduced, whereas the magnitude of regulatory T cells (Tregs) remarkably increased, which is attributed to IFN-γ platelets treatments. Moreover, IFN-γ platelets treatment instigated macrophage polarization toward an anti-inflammatory M2 phenotype that may stimulate pancreatic angiogenesis, and promote β-cell proliferation, consequently ameliorating the new-onset T1D.
Collapse
Grants
- 32371425 National Natural Science Foundation of China
- 32201084 National Natural Science Foundation of China
- JCYJ20240813151128037 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- RCYX20200714114643121 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- JCYJ20200109142610136 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- JCYJ20180507181654186 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- ZDSYS20220606100803007 Science, Technology & Innovation Commission of Shenzhen Municipality, Shenzhen Science and Technology Program
- 2022A1515012289 Natural Science Foundation of Guangdong Province
- GDMUB2022037 Doctoral personnel scientific research start-up Fund project of Guangdong Medical University
- 2024ZDZX2069 Key Field Special Programs of Guangdong Provincial Ordinary Colleges and Universities
- GDMULCJC2024114 Special Project for Clinical and Basic Sci & Tech Innovation of Guangdong Medical University
- National Natural Science Foundation of China
- Natural Science Foundation of Guangdong Province
Collapse
Affiliation(s)
- Yumeng Ma
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Fanqiang Meng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Zhongda Lin
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Yanjun Chen
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Tianyu Lan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Zhaoxin Yang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Rui Diao
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Xiaozhou Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Qi Chen
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsKey Laboratory of Stem Cell and Regenerative Tissue EngineeringSchool of Basic Medical SciencesGuangdong Medical UniversityDongguan523808P. R. China
- The Affiliated Dongguan Songshan Lake Central HospitalGuangdong Medical UniversityDongguanGuangdong523806P. R. China
| | - Chi Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Yishi Tian
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Chanjuan Li
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Wenli Fang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| | - Xin Liang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsKey Laboratory of Stem Cell and Regenerative Tissue EngineeringSchool of Basic Medical SciencesGuangdong Medical UniversityDongguan523808P. R. China
- The Affiliated Dongguan Songshan Lake Central HospitalGuangdong Medical UniversityDongguanGuangdong523806P. R. China
| | - Xudong Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of MedicineShenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityShenzhenGuangdong518107P. R. China
- Department of PharmacologyMolecular Cancer Research CenterSchool of MedicineShenzhen Campus of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdong518107P. R. China
| |
Collapse
|
5
|
Wang Y, Jiang L, Wang J, Huang Y, Dong Y. Utilization of TEP miRNAs in tumor proliferation, diagnostic evaluation, therapeutic intervention, and prognostic assessment. Mol Biol Rep 2025; 52:343. [PMID: 40140156 DOI: 10.1007/s11033-025-10433-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025]
Abstract
According to the most recent 2022 statistics, China accounts for 4.82 million cancer patients, leading globally in prevalence. Early detection and intervention remain the most effective strategies for tumor prevention, treatment, and mortality reduction. There is an urgent need to enhance capabilities in cancer diagnosis and prevention. This study examines the association between tumor-educated platelet (TEP) microRNAs (miRNAs) and malignancies, as well as the role of TEP miRNAs in common cancers. TEP miRNAs offer significant advantages over tissue biopsies, conventional tumor biomarkers, and circulating miRNAs, including simplified sampling procedures, efficient monitoring, and longitudinal assessment of therapeutic dynamics. These advantages are instrumental in advancing tumor screening, diagnosis, treatment, and monitoring.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Clinical Laboratory, Luzhou Longmatan District People's Hospital, Luzhou, 646000, China
| | - Ling Jiang
- Department of Transfusion, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jie Wang
- Department of Transfusion, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yuanshuai Huang
- Department of Transfusion, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ya Dong
- Department of Oncology, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Chunhui Road 182#, Longmatan District, Luzhou, 646000, Sichuan Province, China.
| |
Collapse
|
6
|
Pan Y, Hu D, Chen H, Wang M, Dong X, Wan G, Tang H, Wang H, Chen H. PLGA nanocarriers biomimetic of platelet membranes and their interactions with the placental barrier. Int J Pharm 2025; 671:125225. [PMID: 39824267 DOI: 10.1016/j.ijpharm.2025.125225] [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: 07/20/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
This study focuses on the preparation and characterization of platelet membrane biomimetic nanocarriers (P-PLGA NPs) and investigates their interactions with the transplacental barrier. Poly (lactic-co-glycolic acid) nanoparticles (PLGA NPs) were coated with platelet membrane (PLTM) to construct P-PLGA NPs. Additionally, fluorinated polyethylenimine (F-PEI) was grafted onto PLGA NPs to prepare F-PEI-PLGA NPs, which were compared with PLGA NPs. In vitro placental barrier cell models using human choriocarcinoma cells (BeWo b30) and in vivo pregnancy animal models using ICR mice were utilized to evaluate the transplacental barrier efficiency of the PLGA NPs with different surface modifications. The results demonstrated that all three types of nanoparticles could accumulate in the uterus and placenta. The transplacental barrier efficiency of F-PEI-PLGA NPs was found to be the highest at 4 h, while P-PLGA NPs exhibited the highest transplacental barrier efficiency at 12 and 24 h. Furthermore, there were no significant effects on the main organs, structure, and quantity of uterine spiral arteries, indicating the safety of the nanoparticles (NPs). P-PLGA NPs are expected to be a safe and effective nano-delivery system for perinatal drug delivery. This study provides insights into the transplacental barrier mechanism of NPs with different surface characteristics.
Collapse
Affiliation(s)
- Yuxue Pan
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Danhui Hu
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Honglei Chen
- The Clinical Laboratory Center of Anyang People's Hospital, Anyang 455000, China
| | - Mengyuan Wang
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Xiaocong Dong
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Guoyun Wan
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Hongbo Tang
- Department of Pharmacy, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing 100026, China.
| | - Haijiao Wang
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China.
| | - Hongli Chen
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
7
|
de Castro Sampaio SS, Ramalho MCC, de Souza CS, de Almeida Rodrigues B, de Mendonça GRS, Lazarini M. RHO subfamily of small GTPases in the development and function of hematopoietic cells. J Cell Physiol 2025; 240:e31469. [PMID: 39434451 DOI: 10.1002/jcp.31469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/16/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024]
Abstract
RHOA, RHOB, and RHOC comprise a subfamily of RHO GTPase proteins famed for controlling cytoskeletal dynamics. RHO proteins operate downstream of multiple signals emerging from the microenvironment, leading to diverse cell responses, such as proliferation, adhesion, and migration. Therefore, RHO signaling has been centrally placed in the regulation of blood cells. Despite their high homology, unique roles of RHOA, RHOB, and RHOC have been described in hematopoietic cells. In this article, we overview the contribution of RHO proteins in the development and function of each blood cell lineage. Additionally, we highlight the aberrations of the RHO signaling pathways found in hematological malignancies, providing clues for the identification of new therapeutic targets.
Collapse
Affiliation(s)
| | | | - Caroline Santos de Souza
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Mariana Lazarini
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Hua T, Yao F, Wang H, Liu W, Zhu X, Yao Y. Megakaryocyte in sepsis: the trinity of coagulation, inflammation and immunity. Crit Care 2024; 28:442. [PMID: 39741325 DOI: 10.1186/s13054-024-05221-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/18/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Megakaryocytes are traditionally recognized as cells responsible for platelet production. However, beyond their role in thrombopoiesis, megakaryocytes also participate in inflammatory responses and regulate immune system functions. Sepsis, characterized by life-threatening organ dysfunction due to a dysregulated response to infection, prominently features coagulopathy, severe inflammation, and immune dysfunction as key pathophysiological aspects. AIM OF REVIEW Given the diverse functions of megakaryocytes, we explore their roles in coagulation in the context of sepsis, and also in inflammatory and immune regulation. We try to infer future research directions and potential strategies for sepsis prevention and treatment based on the properties of megakaryocytes. KEY SCIENTIFIC CONCEPTS OF REVIEW The purpose of this review is to both highlight and provide an update on the functions of megakaryocytes and pathophysiological changes in sepsis. Specific emphasis is given to the role of megakaryocytes in sepsis, which suggests value of future research and clinical application.
Collapse
Affiliation(s)
- Tianzhen Hua
- Department of Burns and Plastic Surgery, The Fourth Medical Center, Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China
- Chinese PLA Medical School, Beijing, 100853, China
| | - Fenghua Yao
- Department of Nephrology, First Medical Center of Chinese, PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, 100853, China
| | - Haitao Wang
- Medical Innovation Research Division and Fourth Medical Center of Chinese, PLA General Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Beijing, 100853, China
- Department of Hematology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100071, China
| | - Wei Liu
- Department of Burns and Plastic Surgery, The Fourth Medical Center, Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China.
- Chinese PLA Medical School, Beijing, 100853, China.
| | - Xiaomei Zhu
- Medical Innovation Research Division and Fourth Medical Center of Chinese, PLA General Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Beijing, 100853, China.
| | - Yongming Yao
- Medical Innovation Research Division and Fourth Medical Center of Chinese, PLA General Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Beijing, 100853, China.
| |
Collapse
|
9
|
Fan Z, Gan Y, Hu Y. The potential utilization of platelet-derived extracellular vesicles in clinical treatment. Platelets 2024; 35:2397592. [PMID: 39287127 DOI: 10.1080/09537104.2024.2397592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/09/2024] [Accepted: 08/02/2024] [Indexed: 09/19/2024]
Abstract
Platelet-derived extracellular vesicles (PEVs) are released by platelets in the blood circulation, which carry a rich bio-molecular cargo influential in intercellular communications. PEVs can enter the lymph, bone marrow, and synovial fluid as nano-sized particles, while platelets cannot cross tissue barriers. Considering the advantages of PEVs such as low immunogenicity, high regulation of signal transduction, and easy obtainment, PEVs may be promising therapeutic tools for medical applications. The exceptional functional roles played by PEVs explain the recent interest in exploring new cell-free therapies that could address needs in angiogenesis, regenerative medicine, and targeted drug delivery. The review takes a critical look at the main advances of PEVs in the treatment of diseases by presenting the latest knowledge from the performed studies, in order to enhance the further translation of the PEVs research into feasible therapeutic applications.
Collapse
Affiliation(s)
- Zhijia Fan
- Department of Laboratory Medicine, Beijing Chaoyang Hospital, Beijing Center for Clinical Laboratories, The Third Clinical Medical College of Capital Medical University, Beijing, PR China
| | - Yixiao Gan
- Department of Transfusion Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Yanwei Hu
- Department of Laboratory Medicine, Beijing Chaoyang Hospital, Beijing Center for Clinical Laboratories, The Third Clinical Medical College of Capital Medical University, Beijing, PR China
| |
Collapse
|
10
|
Yao WD, Zhou JN, Tang C, Zhang JL, Chen ZY, Li Y, Gong XJ, Qu MY, Zeng Q, Jia YL, Wang HY, Fan T, Ren J, Guo LL, Xi JF, Pei XT, Han Y, Yue W. Hydrogel Microneedle Patches Loaded with Stem Cell Mitochondria-Enriched Microvesicles Boost the Chronic Wound Healing. ACS NANO 2024; 18:26733-26750. [PMID: 39238258 PMCID: PMC11447894 DOI: 10.1021/acsnano.4c06921] [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: 05/24/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Rescuing or compensating mitochondrial function represents a promising therapeutic avenue for radiation-induced chronic wounds. Adult stem cell efficacies are primarily dependent on the paracrine secretion of mitochondria-containing extracellular vesicles (EVs). However, effective therapeutic strategies addressing the quantity of mitochondria and mitochondria-delivery system are lacking. Thus, in this study, we aimed to design an effective hydrogel microneedle patch (MNP) loaded with stem cell-derived mitochondria-rich EVs to gradually release and deliver mitochondria into the wound tissues and boost wound healing. We, first, used metformin to enhance mitochondrial biogenesis and thereby increasing the secretion of mitochondria-containing EVs (termed "Met-EVs") in adipose-derived stem cells. To verify the therapeutic effects of Met-EVs, we established an in vitro and an in vivo model of X-ray-induced mitochondrial dysfunction. The Met-EVs ameliorated the mitochondrial dysfunction by rescuing mitochondrial membrane potential, increasing adenosine 5'-triphosphate levels, and decreasing reactive oxygen species production by transferring active mitochondria. To sustain the release of EVs into damaged tissues, we constructed a Met-EVs@Decellularized Adipose Matrix (DAM)/Hyaluronic Acid Methacrylic Acid (HAMA)-MNP. Met-EVs@DAM/HAMA-MNP can load and gradually release Met-EVs and their contained mitochondria into wound tissues to alleviate mitochondrial dysfunction. Moreover, we found Met-EVs@DAM/HAMA-MNP can markedly promote macrophage polarization toward the M2 subtype with anti-inflammatory and regenerative functions, which can, in turn, enhance the healing process in mice with skin wounds combined radiation injuries. Collectively, we successfully fabricated a delivery system for EVs, Met-EVs@DAM/HAMA-MNP, to effectively deliver stem cell-derived mitochondria-rich EVs. The effectiveness of this system has been demonstrated, holding great potential for chronic wound treatments in clinic.
Collapse
Affiliation(s)
- Wen-De Yao
- School
of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
- Department
of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing 100853, China
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Jun-Nian Zhou
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Chao Tang
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Ju-Lei Zhang
- Department
of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing 100853, China
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Zhao-Yang Chen
- Department
of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing 100853, China
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Yan Li
- Department
of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing 100853, China
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Xiao-Jing Gong
- Department
of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing 100853, China
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Ming-Yi Qu
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Quan Zeng
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Ya-Li Jia
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Hai-Yang Wang
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Tao Fan
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Jing Ren
- Department
of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing 100853, China
| | - Ling-Li Guo
- Department
of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing 100853, China
| | - Jia-Fei Xi
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Xue-Tao Pei
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| | - Yan Han
- School
of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
- Department
of Plastic and Reconstructive Surgery, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Street, Beijing 100853, China
| | - Wen Yue
- Beijing
Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
11
|
Katana Z, Sianidou K, Kaiopoulos G, Deligianni F, Tsetsakos S, Kouvatsi A, Sakellari I, Kritis A, Touraki M, Sotiropoulos D, Xagorari A. Molecular and biochemical evaluation of oxidative effects of cord blood CD34+ MPs on hematopoietic cells. Blood Cells Mol Dis 2024; 108:102871. [PMID: 39013336 DOI: 10.1016/j.bcmd.2024.102871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/18/2024]
Abstract
A graft source for allogeneic hematopoietic stem cell transplantation is umbilical cord blood, which contains umbilical cord blood mononuclear cells (MNCs and mesenchymal stem cells, both an excellent source of extracellular microparticles (MPs). MPs act as cell communication mediators, which are implicated in reactive oxygen species formation or detoxification depending on their origin. Oxidative stress plays a crucial role in both the development of cancer and its treatment by triggering apoptotic mechanisms, in which CD34+ cells are implicated. The aim of this work is to investigate the oxidative stress status and the apoptosis of HL-60 and mononuclear cells isolated from umbilical cord blood (UCB) following a 24- and 48-hour exposure to CD34 + microparticles (CD34 + MPs). The activity of superoxide dismutase, glutathione reductase, and glutathione S-transferase, as well as lipid peroxidation in the cells, were employed as oxidative stress markers. A 24- and 48-hour exposure of leukemic and mononuclear cells to CD34 + -MPs resulted in a statistically significant increase in the antioxidant activity and lipid peroxidation in both cells types. Moreover, CD34 + MPs affect the expression of BCL2 and FAS and related proteins and downregulate the hematopoietic differentiation program in both HL-60 and mononuclear cells. Our results indicate that MPs through activation of antioxidant enzymes in both homozygous and nonhomozygous cells might serve as a means for graft optimization and enhancement.
Collapse
Affiliation(s)
- Zoi Katana
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kyriaki Sianidou
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gregory Kaiopoulos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Fani Deligianni
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sarantis Tsetsakos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Anastasia Kouvatsi
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Sakellari
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Aristeidis Kritis
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Touraki
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Damianos Sotiropoulos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Angeliki Xagorari
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece.
| |
Collapse
|
12
|
Yuan X, Liu P, Xu L, Liang L, Dong Q, Fan T, Yue W, Qu M, Pei X, Xie X. miR-1915-3p regulates megakaryocytic and erythroid differentiation by targeting SOCS4. Thromb J 2024; 22:74. [PMID: 39123189 PMCID: PMC11316338 DOI: 10.1186/s12959-024-00615-6] [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: 01/31/2024] [Accepted: 05/13/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Proper control of the lineage bias of megakaryocytic and erythroid progenitor cells (MEPs) is of significant importance, the disorder of which will lead to abnormalities in the number and function of platelets and erythrocytes. Unfortunately, the signaling pathways regulating MEP differentiation largely remain to be elucidated. This study aimed to analyze the role and the underlying molecular mechanism of miR-1915-3p in megakaryocytic and erythroid differentiation. METHODS We utilized miRNA mimics and miRNA sponge to alter the expression of miR-1915-3p in megakaryocytic and/or erythroid potential cells; siRNA and overexpression plasmid to change the expression of SOCS4, a potential target of miR-1915-3p. The expression of relevant surface markers was detected by flow cytometry. We scanned for miR-1915-3p target genes by mRNA expression profiling and bioinformatic analysis, and confirmed the targeting by dual-luciferase reporter assay, western blot and gain- and lost-of-function studies. One-way ANOVA and t-test were used to analyze the statistical significance. RESULTS In this study, overexpression or knockdown of miR-1915-3p inhibited or promoted erythroid differentiation, respectively. Accordingly, we scanned for miR-1915-3p target genes and confirmed that SOCS4 is one of the direct targets of miR-1915-3p. An attentive examination of the endogenous expression of SOCS4 during megakaryocytic and erythroid differentiation suggested the involvement of SOCS4 in erythroid/megakaryocytic lineage determination. SOCS4 knockdown lessened erythroid surface markers expression, as well as improved megakaryocytic differentiation, similar to the effects of miR-1915-3p overexpression. While SOCS4 overexpression resulted in reversed effects. SOCS4 overexpression in miR-1915-3p upregulated cells rescued the effect of miR-1915-3p. CONCLUSIONS miR-1915-3p acts as a negative regulator of erythropoiesis, and positively in thrombopoiesis. SOCS4 is one of the key mediators of miR-1915-3p during the differentiation of MEPs.
Collapse
Affiliation(s)
- Xin Yuan
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Pengcong Liu
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Lei Xu
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Liqing Liang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Qian Dong
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Tao Fan
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Wen Yue
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Mingyi Qu
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.
| | - Xuetao Pei
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.
| | - Xiaoyan Xie
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.
| |
Collapse
|
13
|
Zhao J, Tian H, Zhao X, Lan L, Liu H, Sun Y, Yu F. PKCα Induced the Generation of Extracellular Vesicles in Activated Platelets to Promote Breast Cancer Metastasis. Int J Biol Sci 2024; 20:3956-3971. [PMID: 39113702 PMCID: PMC11302887 DOI: 10.7150/ijbs.89822] [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: 09/04/2023] [Accepted: 07/05/2024] [Indexed: 08/10/2024] Open
Abstract
Platelet extracellular vesicles (PEVs) play an important role in tumor development. However, the mechanisms underlying their biogenesis have not been fully elucidated. Protein kinase Cα (PKCα) is an important regulator of platelet activation, but the effect of PKCα on EV generation is unclear. We used small-particle flow cytometry and found that the number of PEVs was increased in patients with breast cancer compared to those with benign breast disease. This was accompanied by increased levels of activated PKCα in breast cancer platelets. Treating platelets with the PKCα agonist phorbol 12-myristate 13-acetate (PMA) increased the phosphorylation PKCα and induced PEV production, while the PKCα inhibitor GÖ6976 showed the opposite effects. Notably, incubating platelets from patients with benign tumors with the culture supernatant of MDA-MB-231 cells induced PKCα phosphorylation in the platelets. Mass spectrometry and coimmunoprecipitation assays showed that Dynamin 2 (DNM2), a member of the guanosine-triphosphate-binding protein family, might cooperate with activated PKCα to regulate PEV production by breast cancer platelets. Similar results were observed in a mouse model of lung metastasis. In addition, PEVs were engulfed by breast cancer cells and promoted cancer cell migration and invasion via miR-1297 delivery. These findings suggested that PKCα cooperates with DNM2 to induce PEV generation, and PEV release might triggered by factors in the breast cancer environment.
Collapse
Affiliation(s)
- Jinghua Zhao
- Dept of Breast Surgery, Yat-Sen Breast Tumor Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Huan Tian
- Dept of Breast Surgery, Yat-Sen Breast Tumor Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Dept of Breast Surgery, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaona Zhao
- Dept of Breast Surgery, Yat-Sen Breast Tumor Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Lan Lan
- School of Life sciences, Guangzhou University, Guangzhou, Guangdong, China
| | - Huanhuan Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Plastic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat- sen University, Guangzhou, Guangdong, China
| | - Yi Sun
- Department of Breast Surgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Fengyan Yu
- Dept of Breast Surgery, Yat-Sen Breast Tumor Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Mao Z, Wu Y, Kong L, Zhou L, Zhang X, Geng A, Cai J, Yang H, Peili H. Changes in cargoes of platelet derived extracellular vesicles heterogeneous subpopulations induced by PM 0.1--Undisclosed cardiovascular injury communication mechanism. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 348:123845. [PMID: 38522605 DOI: 10.1016/j.envpol.2024.123845] [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: 11/01/2023] [Revised: 02/28/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Epidemiological evidence has indicated a closely link between PM0.1 exposure and the incidence rate of cardiovascular diseases. This study explores the underlying communication roles of platelet-derived extracellular vesicles (PEVs) heterogeneous subpopulations in cardiovascular injury. PEVs and PMEVs which were extracted from platelet-rich plasma (PRP) un-exposure or exposure to PM0.1 by TIM4 affinity beads. By optimizing separation conditions, replacing pipelines, and resetting injection procedures, Asymmetric flow field-flow fractionation (AF4) was employed to separate, purify, characterize, and enrich PEVs and PMEVs heterogeneous subpopulations (small PEVs, PEVs-S/PMEVs-S: <100 nm; medium PEVs, PEVs-M/PMEVs-M: 100-200 nm; and large PEVs, PEVs-L/PMEVs-L: >200 nm). The results showed that the cargoes of PMEVs heterogeneous subpopulations which were released by PRP stimulated by PM0.1 were changed obviously. Moreover, compared with PEVs, PMEVs can lead to a decrease in the survival rate of Human Umbilical Vein Endothelial Cells (HUVECs). In PMEVs-S subpopulations, the alterations of lipids associated with membrane fusion and cell signaling transport (such as PC, Cer), as well as miRNAs related to inflammation, angiogenesis, and migration (miR-223, miR-22, miR-126, and miR-150), are similar to those in PMEVs-M subpopulations but distinct from PMEVs-L subpopulations. This study revealed the diverse communication mechanisms underlying PM0.1-induced cardiovascular injury, thereby offering potential avenues for the development of new biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Zhen Mao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yingting Wu
- School of Pharmacy, Capital Medical University, No. 10 Xitoutiao You An Men, Beijing, 100069, China
| | - Ling Kong
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Central Laboratory, Xuanwu Hospital Capital Medical University, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing Institute for Brain Disorders, National Clinical Research Center for Geriatric Disorders, Beijing, 100053, China
| | - Lihong Zhou
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xiaodan Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Aobo Geng
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Jin Cai
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Hong Yang
- Yanjing Medical College, Capital Medical University, No.4 Dadong Road, Shunyi District, Beijing, 101300, China
| | - Huang Peili
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
15
|
Hamid MHBA, Cespedes PF, Jin C, Chen JL, Gileadi U, Antoun E, Liang Z, Gao F, Teague R, Manoharan N, Maldonado-Perez D, Khalid-Alham N, Cerundolo L, Ciaoca R, Hester SS, Pinto-Fernández A, Draganov SD, Vendrell I, Liu G, Yao X, Kvalvaag A, Dominey-Foy DCC, Nanayakkara C, Kanellakis N, Chen YL, Waugh C, Clark SA, Clark K, Sopp P, Rahman NM, Verrill C, Kessler BM, Ogg G, Fernandes RA, Fisher R, Peng Y, Dustin ML, Dong T. Unconventional human CD61 pairing with CD103 promotes TCR signaling and antigen-specific T cell cytotoxicity. Nat Immunol 2024; 25:834-846. [PMID: 38561495 PMCID: PMC11065694 DOI: 10.1038/s41590-024-01802-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
Cancer remains one of the leading causes of mortality worldwide, leading to increased interest in utilizing immunotherapy strategies for better cancer treatments. In the past decade, CD103+ T cells have been associated with better clinical prognosis in patients with cancer. However, the specific immune mechanisms contributing toward CD103-mediated protective immunity remain unclear. Here, we show an unexpected and transient CD61 expression, which is paired with CD103 at the synaptic microclusters of T cells. CD61 colocalization with the T cell antigen receptor further modulates downstream T cell antigen receptor signaling, improving antitumor cytotoxicity and promoting physiological control of tumor growth. Clinically, the presence of CD61+ tumor-infiltrating T lymphocytes is associated with improved clinical outcomes, mediated through enhanced effector functions and phenotype with limited evidence of cellular exhaustion. In conclusion, this study identified an unconventional and transient CD61 expression and pairing with CD103 on human immune cells, which potentiates a new target for immune-based cellular therapies.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Antigens, CD/metabolism
- Antigens, CD/immunology
- Apyrase
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Integrin alpha Chains/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction/immunology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Megat H B A Hamid
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Pablo F Cespedes
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Chen Jin
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ji-Li Chen
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unity, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Uzi Gileadi
- MRC Translational Immune Discovery Unity, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Elie Antoun
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Zhu Liang
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Fei Gao
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Renuka Teague
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Nikita Manoharan
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - David Maldonado-Perez
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Nasullah Khalid-Alham
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
- Oxford National Institute of Health Research (NIHR) Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Lucia Cerundolo
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Raul Ciaoca
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Svenja S Hester
- Target Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Adán Pinto-Fernández
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Simeon D Draganov
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Iolanda Vendrell
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Guihai Liu
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Xuan Yao
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Audun Kvalvaag
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Department of Molecular Cell Biology, Institute of Cancer Research, Oslo University Hospital, Oslo, Norway
| | | | - Charunya Nanayakkara
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nikolaos Kanellakis
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford National Institute of Health Research (NIHR) Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
- Laboratory of Pleural and Lung Cancer Translational Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Respiratory Medicine, Churchill Hospital, Oxford University Hospitals, Oxford, UK
| | - Yi-Ling Chen
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unity, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Craig Waugh
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Sally-Ann Clark
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Kevin Clark
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul Sopp
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Najib M Rahman
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford National Institute of Health Research (NIHR) Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
- Laboratory of Pleural and Lung Cancer Translational Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Respiratory Medicine, Churchill Hospital, Oxford University Hospitals, Oxford, UK
| | - Clare Verrill
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- Oxford National Institute of Health Research (NIHR) Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Benedikt M Kessler
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Graham Ogg
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unity, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ricardo A Fernandes
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Roman Fisher
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Yanchun Peng
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unity, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Michael L Dustin
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Tao Dong
- CAMS Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- MRC Translational Immune Discovery Unity, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
16
|
Launder D, Dillon JT, Wuescher LM, Glanz T, Abdul-Aziz N, Yi EMC, Naglik JR, Worth RG, Conti HR. Immunity to pathogenic mucosal C. albicans infections mediated by oral megakaryocytes activated by IL-17 and candidalysin. Mucosal Immunol 2024; 17:182-200. [PMID: 38246240 PMCID: PMC11034721 DOI: 10.1016/j.mucimm.2024.01.003] [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: 10/25/2023] [Revised: 01/05/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
The fungus Candida albicans can cause mucosal infections including oropharyngeal candidiasis (OPC) in immunocompromised patients. In humans, an increased risk of fungal infections correlates with thrombocytopenia. However, our understanding of platelets and megakaryocytes (Mks) in mucosal fungal infections is almost entirely unknown. When megakaryocyte- and platelet-depleted mice were infected with OPC, the tongue showed higher fungal burden, due to decreased neutrophil accumulation. Protection depended on a distinct population of oral-resident Mks. Interleukin-17, important in antifungal immunity, was required since mice lacking the IL-17 receptor had decreased circulating platelets and their oral Mks did not expand during OPC. The secretion of the peptide toxin candidalysin activated human Mks to release platelets with antifungal capacity. Infection with a candidalysin-deficient strain resulted in decreased expansion of tongue Mks during OPC. This is the first time that a distinct megakaryocyte population was identified in the oral mucosa which is critical for immunity against fungal infection.
Collapse
Affiliation(s)
- Dylan Launder
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States
| | - John T Dillon
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States
| | - Leah M Wuescher
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine & Life Sciences, Toledo, Ohio, United States
| | - Trevor Glanz
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States
| | - Nora Abdul-Aziz
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States
| | - Elise Mein-Chiain Yi
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States
| | - Julian R Naglik
- Center for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Randall G Worth
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine & Life Sciences, Toledo, Ohio, United States
| | - Heather R Conti
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States.
| |
Collapse
|
17
|
Munley JA, Willis ML, Gillies GS, Kannan KB, Polcz VE, Balch JA, Barrios EL, Wallet SM, Bible LE, Efron PA, Maile R, Mohr AM. Exosomal microRNA following severe trauma: Role in bone marrow dysfunction. J Trauma Acute Care Surg 2024; 96:548-556. [PMID: 38151766 PMCID: PMC10978306 DOI: 10.1097/ta.0000000000004225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
INTRODUCTION Severe trauma disrupts bone marrow function and is associated with persistent anemia and altered hematopoiesis. Previously, plasma-derived exosomes isolated after trauma have been shown to suppress in vitro bone marrow function. However, the cargo contained in these vesicles has not been examined. We hypothesized that trauma plasma-derived exosomes exhibit microRNA (miRNA) changes that impact bone marrow function after severe injury. METHODS Plasma was collected from a prospective cohort study of trauma patients (n = 15; 7 males, 8 females) with hip and/or femur fractures and an Injury Severity Score of ≥15; elective total hip arthroplasty (THA) patients (n = 8; 4 males, 4 females) served as operative controls. Exosomes were isolated from plasma with the Invitrogen Total Exosome Isolation Kit (Thermo Fisher Scientific, Waltham, MA), and RNA was isolated using a miRNeasy Mini Kit (Qiagen, Hilden, Germany). Direct quantification of miRNA was performed by NanoString Technologies on a human miRNA gene panel and analyzed with nSolver with significance defined as p < 0.05. RESULTS There were no differences in age or sex distribution between trauma and THA groups; the average Injury Severity Score was 23. Trauma plasma-derived exosomes had 60 miRNA identities that were significantly downregulated and 3 miRNAs that were upregulated when compared with THA ( p < 0.05). Twelve of the downregulated miRNAs have a direct role in hematopoiesis regulation. Furthermore, male trauma plasma-derived exosomes demonstrated downregulation of 150 miRNAs compared with male THA ( p < 0.05). Female trauma plasma-derived exosomes demonstrated downregulation of only four miRNAs and upregulation of two miRNAs compared with female THA ( p < 0.05). CONCLUSION We observed downregulation of 12 miRNAs linked to hematopoiesis along with sexual dimorphism in miRNA expression from plasma-derived exosomes following severe trauma. Understanding sexually dimorphic miRNA expression provides new insight into sex-based changes in postinjury systemic inflammation, immune system dysregulation, and bone marrow dysfunction and will aid us in more precise future potential therapeutic strategies. LEVEL OF EVIDENCE Prognostic and Epidemiological; Level III.
Collapse
Affiliation(s)
- Jennifer A. Munley
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Micah L. Willis
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Gwendolyn S. Gillies
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Kolenkode B. Kannan
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Valerie E. Polcz
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Jeremy A. Balch
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Evan L. Barrios
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Shannon M. Wallet
- Department of Oral Biology, University of Florida College of Medicine, Gainesville, Florida
| | - Letitia E. Bible
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Philip A. Efron
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Robert Maile
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Alicia M. Mohr
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
18
|
Guo J, Cui B, Zheng J, Yu C, Zheng X, Yi L, Zhang S, Wang K. Platelet-derived microparticles and their cargos: The past, present and future. Asian J Pharm Sci 2024; 19:100907. [PMID: 38623487 PMCID: PMC11016590 DOI: 10.1016/j.ajps.2024.100907] [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: 07/05/2023] [Revised: 01/05/2024] [Accepted: 01/31/2024] [Indexed: 04/17/2024] Open
Abstract
All eukaryotic cells can secrete extracellular vesicles, which have a double-membrane structure and are important players in the intercellular communication involved in a variety of important biological processes. Platelets form platelet-derived microparticles (PMPs) in response to activation, injury, or apoptosis. This review introduces the origin, pathway, and biological functions of PMPs and their importance in physiological and pathological processes. In addition, we review the potential applications of PMPs in cancer, vascular homeostasis, thrombosis, inflammation, neural regeneration, biomarkers, and drug carriers to achieve targeted drug delivery. In addition, we comprehensively report on the origin, biological functions, and applications of PMPs. The clinical transformation, high heterogeneity, future development direction, and limitations of the current research on PMPs are also discussed in depth. Evidence has revealed that PMPs play an important role in cell-cell communication, providing clues for the development of PMPs as carriers for relevant cell-targeted drugs. The development history and prospects of PMPs and their cargos are explored in this guidebook.
Collapse
Affiliation(s)
- Jingwen Guo
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Bufeng Cui
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Jie Zheng
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Chang Yu
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xuran Zheng
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Lixin Yi
- School of Pharmacy, China Medical University, Shenyang 110122, China
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Simeng Zhang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Keke Wang
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
19
|
Wang Y, Dong A, Jin M, Li S, Duan Y. TEP RNA: a new frontier for early diagnosis of NSCLC. J Cancer Res Clin Oncol 2024; 150:97. [PMID: 38372784 PMCID: PMC10876732 DOI: 10.1007/s00432-024-05620-w] [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: 12/11/2023] [Accepted: 01/10/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common type of lung cancer (LC), which is the leading cause of tumor mortality. In recent years, compared with tissue biopsy, which is the diagnostic gold standard for tumor diagnosis, Liquid biopsy (LB) is considered to be a more minimally invasive, sensitive, and safer alternative or auxiliary diagnostic method. However, the current value of LB in early diagnosis of LC is not ideal, so it is particularly important to study the changes in blood composition during the process of tumorigenesis and find more sensitive biomarkers. PURPOSE Platelets are a type of abundant blood cells that carry a large amount of RNA. In the LC regulatory network, activated platelets play an important role in the process of tumorigenesis, development, and metastasis. In order to identify predictive liquid biopsy biomarkers for the diagnosis of NSCLC, we summarized the development and function of platelets, the interaction between platelets and tumors, the value of TEP RNA in diagnosis, prognosis, and treatment of NSCLC, and the method for detecting TEP RNA of NSCLC in this article. CONCLUSION The application of platelets in the diagnosis and treatment of NSCLC remains at a nascent stage. In addition to the drawbacks of low platelet count and complex experimental processes, the diagnostic accuracy of TEP RNA-seq for cancer in different populations still needs to be improved and validated. At present, a large number of studies have confirmed significant differences in the expression of TEP RNA in platelets between NSCLC patients and healthy individuals. Continuous exploration of the diagnostic value of TEP RNA in NSCLC is of utmost importance. The integration of NSCLC platelet-related markers with other NSCLC markers can improve current tumor diagnosis and prognostic evaluation systems, providing broad prospects in tumor screening, disease monitoring, and prognosis assessment.
Collapse
Affiliation(s)
- Yuan Wang
- Clinical Laboratory, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang Medical University, Weifang, 261000, Shandong, China
- Department of Clinical Laboratory Science, Weifang Medical University, Weifang, 261000, Shandong, China
| | - Aiping Dong
- Clinical Laboratory, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang Medical University, Weifang, 261000, Shandong, China
| | - Minhan Jin
- Clinical Laboratory, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang Medical University, Weifang, 261000, Shandong, China
- Department of Clinical Laboratory Science, Weifang Medical University, Weifang, 261000, Shandong, China
| | - Shirong Li
- Clinical Laboratory, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang Medical University, Weifang, 261000, Shandong, China.
| | - Yang Duan
- Clinical Laboratory, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang Medical University, Weifang, 261000, Shandong, China.
| |
Collapse
|
20
|
Saberian M, Abak N. Hydrogel-mediated delivery of platelet-derived exosomes: Innovations in tissue engineering. Heliyon 2024; 10:e24584. [PMID: 38312628 PMCID: PMC10835177 DOI: 10.1016/j.heliyon.2024.e24584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
In this scholarly review, we conduct a thorough examination of the significant role played by platelet-derived exosomes (Plt-Exos) and hydrogels in the fields of tissue engineering and regenerative medicine. Our detailed investigation highlights the central involvement of Plt-Exos in various physiological and pathological processes, underscoring their potential contributions to diverse areas such as wound healing, neural rejuvenation, and cancer progression. Despite the promising therapeutic aspects, the notable variability in the isolation and characterization of pEVs underscores the need for a more rigorous and standardized methodology. Shifting our focus to hydrogels, they have emerged as promising biomaterials relevant to tissue engineering and regenerative medicine. Their unique characteristics, especially their chemical and physical adaptability, along with the modifiability of their biochemical properties, make hydrogels a captivating subject. These exceptional features open avenues for numerous tissue engineering applications, facilitating the delivery of essential growth factors, cytokines, and microRNAs. This analysis explores the innovative integration of Plt-Exos with hydrogels, presenting a novel paradigm in tissue engineering. Through the incorporation of Plt-Exos into hydrogels, there exists an opportunity to enhance tissue regeneration endeavors by combining the bioactive features of Plt-Exos with the restorative capabilities of hydrogel frameworks. In conclusion, the cooperative interaction between platelet-derived exosomes and hydrogels indicates a promising path in tissue engineering and regenerative medicine. Nevertheless, the successful execution of this approach requires a deep understanding of molecular dynamics, coupled with a dedication to refining isolation techniques.
Collapse
Affiliation(s)
- Mostafa Saberian
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Abak
- Hematology and Transfusion Science Department, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Zhou M, Feng Y, Zhang X, Chen J, Yao N, Fu S, Ni T, Chen Y, Xie F, Roy S, Liu J, Yang Y, He Y, Zhao Y, Yang N. Platelet-derived microparticles adoptively transfer integrin β3 to promote antitumor effect of tumor-infiltrating T cells. Oncoimmunology 2024; 13:2304963. [PMID: 38235317 PMCID: PMC10793703 DOI: 10.1080/2162402x.2024.2304963] [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: 08/23/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024] Open
Abstract
Approximately two-thirds of hepatocellular carcinoma (HCC) is considered a "cold tumor" characterized by few tumor-infiltrating T cells and an abundance of immunosuppressive cells. Cilengitide, an integrin αvβ3 inhibitor, has failed in clinical trials as a potential anticancer drug. This failure implies that integrin αvβ3 may play an important role in immune cells. However, the expression and potential role of integrin αvβ3 in T cells of HCC patients remain unknown. Here, we established two HCC models and found that cilengitide had a dual effect on the HCC microenvironment by exerting both antitumor effect and immunosuppressive effect on T cells. This may partly explain the failure of cilengitide in clinical trials. In clinical specimens, HCC-infiltrating T cells exhibited deficient expression and activation of integrin β3, which was associated with poor T-cell infiltration into tumors. Additionally, integrin β3 functioned as a positive immunomodulatory molecule to facilitate T-cell infiltration and T helper 1-type immune response in vitro. Furthermore, T cells and platelet-derived microparticles (PMPs) co-culture assay revealed that PMPs adoptively transferred integrin β3 to T cells and positively regulated T cell immune response. This process was mediated by clathrin-dependent endocytosis and macropinocytosis. Our data demonstrate that integrin β3 deficiency on HCC-infiltrating T cells may be involved in shaping the immunosuppressive tumor microenvironment. PMPs transfer integrin β3 to T cells and positively regulate T cell immune response, which may provide a new insight into immune therapy of HCC.
Collapse
Affiliation(s)
- Mimi Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yali Feng
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiaoli Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jianguo Chen
- School of Software Engineering, Sun Yat-Sen University, Zhuhai, China
| | - Naijuan Yao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Shan Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Tianzhi Ni
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yi Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Fei Xie
- Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Sahasrabda Roy
- School of International Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jinfeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yuan Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yingli He
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yingren Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Nan Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
22
|
Ning M, Hua S, Ma Y, Liu Y, Wang D, Xu K, Yu H. Microvesicles facilitate the differentiation of mesenchymal stem cells into pancreatic beta-like cells via miR-181a-5p/150-5p. Int J Biol Macromol 2024; 254:127719. [PMID: 37918601 DOI: 10.1016/j.ijbiomac.2023.127719] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/04/2023]
Abstract
Transplantation of pancreatic islet cells is a promising strategy for the long-term treatment of type 1 diabetes (T1D). The stem cell-derived beta cells showed great potential as substitute sources of transplanted pancreatic islet cells. However, the current efficiency of stem cell differentiation still cannot match the requirements for clinical transplantation. Here, we report that microvesicles (MVs) from insulin-producing INS-1 cells could induce mesenchymal stem cell (MSC) differentiation into pancreatic beta-like cells. The combination of MVs with small molecules, nicotinamide and insulin-transferrin-selenium (ITS), dramatically improved the efficiency of MSC differentiation. Notably, the function of MVs in MSC differentiation requires their entry into MSCs through giant pinocytosis. The MVs-treated or MVs combined with small molecules-treated MSCs show pancreatic beta-like cell morphology and response to glucose stimulation in insulin secretion. Using high throughput small RNA-sequencing, we found that MVs induced MSC differentiation into the beta-like cells through miR-181a-5p/150-5p. Together, our findings reveal the role of MVs or the MV-enriched miR-181a-5p/150-5p as a class of biocompatible reagents to differentiate MSCs into functional beta-like cells and demonstrate that the combined usage of MVs or miR-181a-5p/150-5p with small molecules can potentially be used in making pancreatic islet cells for future clinical purposes.
Collapse
Affiliation(s)
- Mingming Ning
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Shanshan Hua
- Department of Spine Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao 266071, China
| | - Ying Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yunpeng Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Dianliang Wang
- Stem cell and tissue engineering research laboratory, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China.
| | - Kai Xu
- Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
23
|
Wan P, Tan X, Sheng M, Xiang Y, Wang P, Yu M. Platelet Exosome-Derived miR-223-3p Regulates Pyroptosis in the Cell Model of Sepsis-Induced Acute Renal Injury by Targeting Mediates NLRP3. Crit Rev Immunol 2024; 44:53-65. [PMID: 38421705 DOI: 10.1615/critrevimmunol.2023051651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
BACKGROUND The present study investigated the roles and mechanisms of platelet-derived exosomes in sepsis-induced acute renal injury. METHODS The blood samples of septic patients and healthy controls were collected for clinical examination. The plasma levels of miR-223-3p and NLRP3 mRNA were analyzed by qRT-PCR and the serum IL-1β and creatinine levels were quantified by enzyme-linked immunosorbent assay (ELISA). C57BL/6 mice injected with LPS (lipopolysaccharide) were employed as the animal model for sepsis-induced acute renal injury. Human coronary artery endothelial cells (HCAECs) were treated with TNF-α as a cellular model for sepsis-induced endothelial damages. RESULTS The number of PMP (platelet-derived microparticles) in patients with sepsis was increased. The level of miR-223-3p in the platelet exosomes isolated from the serum sample in patients with sepsis was significantly lower than that of the healthy controls. The level of miR-223-3p was also decreased in the platelet exosomes of mouse model with sepsis-induced acute renal injury. Downregulating miR-223-3p promoted sepsis-induced acute renal injury in mice model, while the administration of miR-223-3p reduced the inflammation in endothelial cells of sepsis-induced acute renal injury. NLRP3 (NLR Family Pyrin Domain Containing 3) was identified as one target of miR-223-3p in the platelet exosomes of sepsis-induced acute kidney injury. miR-223-3p attenuated NLRP3-induced pyroptosis in endothelial cell model of sepsis-induced acute kidney injury. CONCLUSION Our data suggest that platelet exosome-derived miR-223-3p negatively regulates NLRP3-dependent inflammasome to suppress pyroptosis in endothelial cells. Decreased miR-223-3p expression promotes the inflammation in sepsis-induced acute renal injury. Targeting miR-223-3p may be developed into a therapeutic approach for sepsis-induced acute renal injury.
Collapse
Affiliation(s)
- Peng Wan
- Department of Critical Care Medicine, The First Clinical Medical College of Three Gorges University,Yichang Central People's Hospital, Yichang City, Hubei, 443000, China
| | - Xiang Tan
- Department of Critical Care Medicine, The First Clinical Medical College of Three Gorges University,Yichang Central People's Hospital, Yichang City, Hubei, 443000, China
| | - Mengting Sheng
- Department of Critical Care Medicine, The First Clinical Medical College of Three Gorges University,Yichang Central People's Hospital, Yichang City, Hubei, 443000, China
| | - Yan Xiang
- Department of Critical Care Medicine, The First Clinical Medical College of Three Gorges University,Yichang Central People's Hospital, Yichang City, Hubei, 443000, China
| | - Peng Wang
- Department of Critical Care Medicine, The First Clinical Medical College of Three Gorges University,Yichang Central People's Hospital, Yichang City, Hubei, 443000, China
| | - Min Yu
- The people's hospital of China Three Gorges University
| |
Collapse
|
24
|
Gareev I, Pavlov V, Du W, Yang B. MiRNAs and Their Role in Venous Thromboembolic Complications. Diagnostics (Basel) 2023; 13:3383. [PMID: 37958279 PMCID: PMC10650162 DOI: 10.3390/diagnostics13213383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Venous thromboembolic complications (VTCs), which include deep vein thrombosis (DVT) and pulmonary embolism (PE), have remained a pressing problem in modern clinical medicine for a long time. Despite the already wide arsenal of modern methods for diagnosing and treating this disease, VTCs rank third in the structure of causes of death among all cardiovascular diseases, behind myocardial infarction (MI) and ischemic stroke (IS). Numerous studies have confirmed the importance of understanding the molecular processes of VTCs for effective therapy and diagnosis. Significant progress has been made in VTC research in recent years, where the relative contribution of microRNAs (miRNAs) in the mechanism of thrombus formation and their consideration as therapeutic targets have been well studied. In this case, accurate, timely, and as early as possible diagnosis of VTCs is of particular importance, which will help improve both short-term and long-term prognoses of patients. This case accounts for the already well-studied circulating miRNAs as non-invasive biomarkers. This study presents currently available literature data on the role of miRNAs in VTCs, revealing their potential as therapeutic targets and diagnostic and prognostic tools for this terrible disease.
Collapse
Affiliation(s)
- Ilgiz Gareev
- Central Research Laboratory, Bashkir State Medical University, 3 Lenin Street, 450008 Ufa, Russia;
| | - Valentin Pavlov
- Department of Urology, Bashkir State Medical University, 3 Lenin Street, 450008 Ufa, Russia;
| | - Weijie Du
- Department of Pharmacology, The Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150067, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin 150081, China
| | - Baofeng Yang
- Department of Pharmacology, The Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150067, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin 150081, China
| |
Collapse
|
25
|
Feng J, Yao Y, Wang Q, Han X, Deng X, Cao Y, Chen X, Zhou M, Zhao C. Exosomes: Potential key players towards novel therapeutic options in diabetic wounds. Biomed Pharmacother 2023; 166:115297. [PMID: 37562235 DOI: 10.1016/j.biopha.2023.115297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/12/2023] Open
Abstract
Diabetic wounds are usually difficult to heal, and wounds in foot in particular are often aggravated by infection, trauma, diabetic neuropathy, peripheral vascular disease and other factors, resulting in serious foot ulcers. The pathogenesis and clinical manifestations of diabetic wounds are complicated, and there is still a lack of objective and in-depth laboratory diagnosis and classification standards. Exosomes are nanoscale vesicles containing DNA, mRNA, microRNA, cyclic RNA, metabolites, lipids, cytoplasm and cell surface proteins, etc., which are involved in intercellular communication and play a crucial role in vascular regeneration, tissue repair and inflammation regulation in the process of diabetic wound healing. Here, we discussed exosomes of different cellular origins, such as diabetic wound-related fibroblasts (DWAF), adipose stem cells (ASCs), mesenchymal stem cells (MSCs), immune cells, platelets, human amniotic epithelial cells (hAECs), epidermal stem cells (ESCs), and their various molecular components. They exhibit multiple therapeutic effects during diabetic wound healing, including promoting cell proliferation and migration associated with wound healing, regulating macrophage polarization to inhibit inflammatory responses, promoting nerve repair, and promoting vascular renewal and accelerating wound vascularization. In addition, exosomes can be designed to deliver different therapeutic loads and have the ability to deliver them to the desired target. Therefore, exosomes may become an innovative target for precision therapeutics in diabetic wounds. In this review, we summarize the latest research on the role of exosomes in the healing of diabetic wound by regulating the pathogenesis of diabetic wounds, and discuss their potential applications in the precision treatment of diabetic wounds.
Collapse
Affiliation(s)
- Jiawei Feng
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yichen Yao
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qixue Wang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaozhou Han
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Xiaofei Deng
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yemin Cao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Xinghua Chen
- Jinshan Hospital Afflicted to Fudan University, Shanghai, China.
| | - Mingmei Zhou
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Cheng Zhao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China.
| |
Collapse
|
26
|
Wu T, Jiang Y, Shi W, Wang Y, Li T. Endoplasmic reticulum stress: a novel targeted approach to repair bone defects by regulating osteogenesis and angiogenesis. J Transl Med 2023; 21:480. [PMID: 37464413 PMCID: PMC10353205 DOI: 10.1186/s12967-023-04328-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
Bone regeneration therapy is clinically important, and targeted regulation of endoplasmic reticulum (ER) stress is important in regenerative medicine. The processing of proteins in the ER controls cell fate. The accumulation of misfolded and unfolded proteins occurs in pathological states, triggering ER stress. ER stress restores homeostasis through three main mechanisms, including protein kinase-R-like ER kinase (PERK), inositol-requiring enzyme 1ɑ (IRE1ɑ) and activating transcription factor 6 (ATF6), collectively known as the unfolded protein response (UPR). However, the UPR has both adaptive and apoptotic effects. Modulation of ER stress has therapeutic potential for numerous diseases. Repair of bone defects involves both angiogenesis and bone regeneration. Here, we review the effects of ER stress on osteogenesis and angiogenesis, with emphasis on ER stress under high glucose (HG) and inflammatory conditions, and the use of ER stress inducers or inhibitors to regulate osteogenesis and angiogenesis. In addition, we highlight the ability for exosomes to regulate ER stress. Recent advances in the regulation of ER stress mediated osteogenesis and angiogenesis suggest novel therapeutic options for bone defects.
Collapse
Affiliation(s)
- Tingyu Wu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yaping Jiang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Weipeng Shi
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yingzhen Wang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Tao Li
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China.
| |
Collapse
|
27
|
Song H, Li J, Peng C, Liu D, Mei Z, Yang Z, Tian X, Zhang X, Jing Q, Yan C, Han Y. The role of CREG1 in megakaryocyte maturation and thrombocytopoiesis. Int J Biol Sci 2023; 19:3614-3627. [PMID: 37496998 PMCID: PMC10367557 DOI: 10.7150/ijbs.78660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 06/20/2023] [Indexed: 07/28/2023] Open
Abstract
Abnormal megakaryocyte maturation and platelet production lead to platelet-related diseases and impact the dynamic balance between hemostasis and bleeding. Cellular repressor of E1A-stimulated gene 1 (CREG1) is a glycoprotein that promotes tissue differentiation. However, its role in megakaryocytes remains unclear. In this study, we found that CREG1 protein is expressed in platelets and megakaryocytes and was decreased in the platelets of patients with thrombocytopenia. A cytosine arabinoside-induced thrombocytopenia mouse model was established, and the mRNA and protein expression levels of CREG1 were found to be reduced in megakaryocytes. We established megakaryocyte/platelet conditional knockout (Creg1pf4-cre) and transgenic mice (tg-Creg1). Compared to Creg1fl/fl mice, Creg1pf4-cre mice exhibited thrombocytopenia, which was mainly caused by inefficient bone marrow (BM) thrombocytopoiesis, but not by apoptosis of circulating platelets. Cultured Creg1pf4-cre-megakaryocytes exhibited impairment of the actin cytoskeleton, with less filamentous actin, significantly fewer proplatelets, and lower ploidy. CREG1 directly interacts with MEK1/2 and promotes MEK1/2 phosphorylation. Thus, our study uncovered the role of CREG1 in the regulation of megakaryocyte maturation and thrombopoiesis, and it provides a possible theoretical basis for the prevention and treatment of thrombocytopenia.
Collapse
Affiliation(s)
- HaiXu Song
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jiayin Li
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
- Northeastern University, Shenyang, China
| | - Chengfei Peng
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Dan Liu
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Zhu Mei
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Zheming Yang
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
- Northeastern University, Shenyang, China
| | - Xiaoxiang Tian
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaolin Zhang
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Quanmin Jing
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaling Han
- National Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
28
|
Chen J, Wang M, Zhang Y, Zhu F, Xu Y, Yi G, Zheng R, Wu B. Platelet extracellular vesicles: Darkness and light of autoimmune diseases. Int Rev Immunol 2023; 43:63-73. [PMID: 37350464 DOI: 10.1080/08830185.2023.2225551] [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: 01/11/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023]
Abstract
Autoimmune diseases are characterized by a breakdown of immune tolerance, leading to inflammation and irreversible end-organ tissue damage. Platelet extracellular vesicles are cellular elements that are important in blood circulation and actively participate in inflammatory and immune responses through intercellular communication and interactions between inflammatory cells, immune cells, and their secreted factors. Therefore, platelet extracellular vesicles are the "accelerator" in the pathological process of autoimmune diseases; however, this robust set of functions of platelet extracellular vesicles has also prompted new advances in therapeutic strategies for autoimmune diseases. In this review, we update fundamental mechanisms based on platelet extracellular vesicles communication function in autoimmune diseases. We also focus on the potential role of platelet extracellular vesicles for the treatment of autoimmune diseases. Some recent studies have found that antiplatelet aggregation drugs, specific biological agents can reduce the release of platelet extracellular vesicles. Platelet extracellular vesicles can also serve as vehicles to deliver drugs to targeted cells. It seems that we can try to silence or inhibit microRNA carried by platelet extracellular vesicles transcription and regulate the target cells to treat autoimmune diseases as platelet extracellular vesicles can transfer microRNA to other cells to regulate immune-inflammatory responses. Hopefully, the information presented here will provide hope for patients with autoimmune diseases.
Collapse
Affiliation(s)
- Jingru Chen
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, P.R. China
| | - Miao Wang
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
| | - Ying Zhang
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
| | - Fenglin Zhu
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
| | - Yanqiu Xu
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
| | - Guoxiang Yi
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
| | - Runxiu Zheng
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, P.R. China
| | - Bin Wu
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, P.R. China
| |
Collapse
|
29
|
Du H, Tang Q, Yang J, Yan B, Yang L, Wang M. Genome-wide DNA methylation profiling of CD4 + T lymphocytes identifies differentially methylated loci associated with adult primary refractory immune thrombocytopenia. BMC Med Genomics 2023; 16:124. [PMID: 37291547 PMCID: PMC10251572 DOI: 10.1186/s12920-023-01557-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 05/23/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND DNA methylation played a crucial role in the pathogenesis of immune thrombocytopenia (ITP). However, genome-wide DNA methylation analysis has not been applied thus far. The present study aimed to provide the first DNA methylation profiling for ITP. METHODS Peripheral blood CD4+ T lymphocytes samples were collected from 4 primary refractory ITP cases and 4 age-matched healthy controls, and DNA methylome profiling was performed using Infinium MethylationEPIC BeadChip. Differentially methylated CpG sites were further validated in another independent cohort of 10 ITP patients and 10 healthy controls using qRT-PCR. RESULTS The DNA methylome profiling identified a total of 260 differentially methylated CpG sites mapping to 72 hypermethylated and 64 hypomethylated genes. These genes were mainly enriched in the actin nucleation of the Arp2/3 complex, vesicle transport, histone H3-K36 demethylation, Th1 and Th2 cell differentiation, and Notch signaling pathway according to the GO and KEGG databases. The mRNA expression of CASP9, C1orf109, and AMD1 were significantly different. CONCLUSIONS Given the altered DNA methylation profiling of ITP, our study provides new insights into its genetic mechanism and suggests candidate biomarkers for the diagnosis and treatment of ITP.
Collapse
Affiliation(s)
- Hanzhi Du
- Department of Haematology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qinghua Tang
- Department of Osteoporosis, Xi'an Jiaotong University Affiliated HongHui Hospital, Xi'an, China
| | - Jian Yang
- Department of Clinical Research Centre, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bin Yan
- Department of Clinical Research Centre, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lihong Yang
- Department of Clinical Research Centre, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengchang Wang
- Department of Haematology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
30
|
Cacic D, Hervig T, Reikvam H. Platelets for advanced drug delivery in cancer. Expert Opin Drug Deliv 2023; 20:673-688. [PMID: 37212640 DOI: 10.1080/17425247.2023.2217378] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/19/2023] [Indexed: 05/23/2023]
Abstract
INTRODUCTION Cancer-related drug expenses are rising with the increasing cancer incidence and cost may represent a severe challenge for drug access for patients with cancer. Consequently, strategies for increasing therapeutic efficacy of already available drugs may be essential for the future health-care system. AREAS COVERED In this review, we have investigated the potential for the use of platelets as drug-delivery systems. We searched PubMed and Google Scholar to identify relevant papers written in English and published up to January 2023. Papers were included at the authors' discretion to reflect an overview of state of the art. EXPERT OPINION It is known that cancer cells interact with platelets to gain functional advantages including immune evasion and metastasis development. This platelet-cancer interaction has been the inspiration for numerous platelet-based drug delivery systems using either drug-loaded or drug-bound platelets, or platelet membrane-containing hybrid vesicles combining platelet membranes with synthetic nanocarriers. Compared to treatment with free drug or synthetic drug vectors, these strategies may improve pharmacokinetics and selective cancer cell targeting. There are multiple studies showing improved therapeutic efficacy using animal models, however, no platelet-based drug delivery systems have been tested in humans, meaning the clinical relevance of this technology remains uncertain.
Collapse
Affiliation(s)
- Daniel Cacic
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Tor Hervig
- Irish Blood Transfusion Service, Dublin, Ireland
| | - Håkon Reikvam
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
31
|
Hayashi Y, Nishimura K, Tanaka A, Inoue D. Extracellular vesicle-mediated remodeling of the bone marrow microenvironment in myeloid malignancies. Int J Hematol 2023; 117:821-829. [PMID: 37041345 DOI: 10.1007/s12185-023-03587-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 04/13/2023]
Abstract
Hematopoiesis is maintained and regulated by a bone marrow-specific microenvironment called a niche. In hematological malignancies, tumor cells induce niche remodeling, and the reconstructed niche is closely linked to disease pathogenesis. Recent studies have suggested that extracellular vesicles (EVs) secreted from tumor cells play a principal role in niche remodeling in hematological malignancies. Although EVs are emerging as potential therapeutic targets, the underlying mechanism of action remains unclear, and selective inhibition remains a challenge. This review summarizes remodeling of the bone marrow microenvironment in hematological malignancies and its contribution to pathogenesis, as well as roles of tumor-derived EVs, and provides a perspective on future research in this field.
Collapse
Affiliation(s)
- Yasutaka Hayashi
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, 6-3-7, Minatojimaminami-machi, Chuo-ku, Kobe, 650-0047, Japan.
| | - Koutarou Nishimura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, 6-3-7, Minatojimaminami-machi, Chuo-ku, Kobe, 650-0047, Japan
| | - Atsushi Tanaka
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, 6-3-7, Minatojimaminami-machi, Chuo-ku, Kobe, 650-0047, Japan
- Laboratory of Immunology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Daichi Inoue
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, 6-3-7, Minatojimaminami-machi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
32
|
Huang SJ, Zhang Y, Wang GH, Lu J, Chen PP, Zhang JX, Li XQ, Yuan BY, Liu XQ, Jiang TT, Wang MY, Liu WT, Ruan XZ, Liu BC, Ma KL. Deposition of platelet-derived microparticles in podocytes contributes to diabetic nephropathy. Int Urol Nephrol 2023; 55:355-366. [PMID: 35931920 DOI: 10.1007/s11255-022-03332-z] [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: 05/21/2021] [Accepted: 07/25/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Diabetic nephropathy (DN) is the leading cause of end-stage renal disease in the developed world. Podocyte injury is a critical cellular event involved in the progression of DN. Our previous studies demonstrated that platelet-derived microparticles (PMPs) mediated endothelial injury in diabetic rats. This study aimed to investigate whether PMPs are deposited in podocytes and to assess their potential effects on podocyte injury in DN. METHODS The deposition of PMPs in podocytes was assessed by immunofluorescent staining and electron microscopy. The changes in renal pathology and ultra-microstructure were assessed by periodic acid-Schiff staining and electron microscopy, respectively. The expression of inflammatory cytokines and extracellular matrix proteins was measured by immuno-histochemical staining and western blot. RESULTS PMPs were widely deposited in podocytes of glomeruli in diabetic patients and animal models and closely associated with DN progression. Interestingly, aspirin treatment significantly inhibited the accumulation of PMPs in the glomeruli of diabetic rats, alleviated mesangial matrix expansion and fusion of foot processes, and decreased the protein expression of inflammatory cytokines and extracellular matrix secretion. An in vitro study further confirmed the deposition of PMPs in podocytes. Moreover, PMP stimulation induced the phenotypic transition of podocytes through decreased podocin protein expression and increased protein expression of α-SMA and fibronectin, which was correlated with increased production of inflammatory cytokines. CONCLUSION Our findings demonstrated for the first time that the deposition of PMPs in podocytes contributed to the development of DN.
Collapse
Affiliation(s)
- Si Jia Huang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yang Zhang
- Renal Department, Nanjing First Hospital, Nanjing, 210006, China
| | - Gui Hua Wang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jian Lu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Pei Pei Chen
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jia Xiu Zhang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xue Qi Li
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ben Yin Yuan
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiao Qi Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ting Ting Jiang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Meng Ying Wang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Wen Tao Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiong Zhong Ruan
- John Moorhead Research Laboratory, Department of Renal Medicine, University College London (UCL) Medical School, Royal Free Campus, London, NW3 2PF, UK
| | - Bi Cheng Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Kun Ling Ma
- Department of Nephrology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
33
|
Yang S, Wang L, Wu Y, Wu A, Huang F, Tang X, Kantawong F, Anuchapreeda S, Qin D, Mei Q, Chen J, Huang X, Zhang C, Wu J. Apoptosis in megakaryocytes: Safeguard and threat for thrombopoiesis. Front Immunol 2023; 13:1025945. [PMID: 36685543 PMCID: PMC9845629 DOI: 10.3389/fimmu.2022.1025945] [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: 08/23/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023] Open
Abstract
Platelets, generated from precursor megakaryocytes (MKs), are central mediators of hemostasis and thrombosis. The process of thrombopoiesis is extremely complex, regulated by multiple factors, and related to many cellular events including apoptosis. However, the role of apoptosis in thrombopoiesis has been controversial for many years. Some researchers believe that apoptosis is an ally of thrombopoiesis and platelets production is apoptosis-dependent, while others have suggested that apoptosis is dispensable for thrombopoiesis, and is even inhibited during this process. In this review, we will focus on this conflict, discuss the relationship between megakaryocytopoiesis, thrombopoiesis and apoptosis. In addition, we also consider why such a vast number of studies draw opposite conclusions of the role of apoptosis in thrombopoiesis, and try to figure out the truth behind the mystery. This review provides more comprehensive insights into the relationship between megakaryocytopoiesis, thrombopoiesis, and apoptosis and finds some clues for the possible pathological mechanisms of platelet disorders caused by abnormal apoptosis.
Collapse
Affiliation(s)
- Shuo Yang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yuesong Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anguo Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Feihong Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Xiaoqin Tang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Fahsai Kantawong
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Dalian Qin
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qibing Mei
- School of Pharmacy, Southwest Medical University, Luzhou, China
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xinwu Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chunxiang Zhang
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Jianming Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
34
|
He Y, Wu Q. The Effect of Extracellular Vesicles on Thrombosis. J Cardiovasc Transl Res 2022:10.1007/s12265-022-10342-w. [DOI: 10.1007/s12265-022-10342-w] [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] [Received: 08/22/2022] [Accepted: 11/17/2022] [Indexed: 11/29/2022]
Abstract
Abstract
The risk of cardiovascular events caused by acute thrombosis is high, including acute myocardial infarction, acute stroke, acute pulmonary embolism, and deep vein thrombosis. In this review, we summarize the roles of extracellular vesicles of different cellular origins in various cardiovascular events associated with acute thrombosis, as described in the current literature, to facilitate the future development of a precise therapy for thrombosis caused by such vesicles. We hope that our review will indicate a new horizon in the field of cardiovascular research with regard to the treatment of acute thrombosis, especially targeting thrombosis caused by extracellular vesicles secreted by individual cells. As more emerging technologies are being developed, new diagnostic and therapeutic strategies related to EVs are expected to be identified for related diseases in the future.
Collapse
|
35
|
Badimon L, Padro T, Arderiu G, Vilahur G, Borrell-Pages M, Suades R. Extracellular vesicles in atherothrombosis: From biomarkers and precision medicine to therapeutic targets. Immunol Rev 2022; 312:6-19. [PMID: 35996799 DOI: 10.1111/imr.13127] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of global mortality. Extracellular vesicles (EVs) are small phospholipid vesicles that convey molecular bioactive cargoes and play essential roles in intercellular communication and, hence, a multifaceted role in health and disease. The present review offers a glimpse into the current state and up-to-date concepts on EV field. It also covers their association with several cardiovascular risk factors and ischemic conditions, being subclinical atherosclerosis of utmost relevance for prevention. Interestingly, we show that EVs hold promise as prognostic and diagnostic as well as predictive markers of ASCVD in the precision medicine era. We then report on the role of EVs in atherothrombosis, disentangling the mechanisms involved in the initiation, progression, and complication of atherosclerosis and showing their direct effect in the context of arterial thrombosis. Finally, their potential use for therapeutic intervention is highlighted.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Teresa Padro
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Arderiu
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Suades
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
36
|
Dai Z, Zhao T, Song N, Pan K, Yang Y, Zhu X, Chen P, Zhang J, Xia C. Platelets and platelet extracellular vesicles in drug delivery therapy: A review of the current status and future prospects. Front Pharmacol 2022; 13:1026386. [PMID: 36330089 PMCID: PMC9623298 DOI: 10.3389/fphar.2022.1026386] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/03/2022] [Indexed: 11/24/2022] Open
Abstract
Platelets are blood cells that are primarily produced by the shedding of megakaryocytes in the bone marrow. Platelets participate in a variety of physiological and pathological processes in vivo, including hemostasis, thrombosis, immune-inflammation, tumor progression, and metastasis. Platelets have been widely used for targeted drug delivery therapies for treating various inflammatory and tumor-related diseases. Compared to other drug-loaded treatments, drug-loaded platelets have better targeting, superior biocompatibility, and lower immunogenicity. Drug-loaded platelet therapies include platelet membrane coating, platelet engineering, and biomimetic platelets. Recent studies have indicated that platelet extracellular vesicles (PEVs) may have more advantages compared with traditional drug-loaded platelets. PEVs are the most abundant vesicles in the blood and exhibit many of the functional characteristics of platelets. Notably, PEVs have excellent biological efficacy, which facilitates the therapeutic benefits of targeted drug delivery. This article provides a summary of platelet and PEVs biology and discusses their relationships with diseases. In addition, we describe the preparation, drug-loaded methods, and specific advantages of platelets and PEVs targeted drug delivery therapies for treating inflammation and tumors. We summarize the hot spots analysis of scientific articles on PEVs and provide a research trend, which aims to give a unique insight into the development of PEVs research focus.
Collapse
Affiliation(s)
- Zhanqiu Dai
- Department of Spine Surgery, Zhejiang Provincial People’s Hospital, Hangzhou Medical College People’s Hospital, Hangzhou, Zhejiang, China
- Department of Orthopaedics, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Tingxiao Zhao
- Department of Spine Surgery, Zhejiang Provincial People’s Hospital, Hangzhou Medical College People’s Hospital, Hangzhou, Zhejiang, China
| | - Nan Song
- Department of Pathology, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Kaifeng Pan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Yang Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xunbin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
- *Correspondence: Pengfei Chen, ; Jun Zhang, ; Chen Xia,
| | - Jun Zhang
- Department of Spine Surgery, Zhejiang Provincial People’s Hospital, Hangzhou Medical College People’s Hospital, Hangzhou, Zhejiang, China
- *Correspondence: Pengfei Chen, ; Jun Zhang, ; Chen Xia,
| | - Chen Xia
- Department of Spine Surgery, Zhejiang Provincial People’s Hospital, Hangzhou Medical College People’s Hospital, Hangzhou, Zhejiang, China
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
- *Correspondence: Pengfei Chen, ; Jun Zhang, ; Chen Xia,
| |
Collapse
|
37
|
Bai X, Zhang H, Li Z, Chen O, He H, Jia X, Zou L. Platelet-derived extracellular vesicles encapsulate microRNA-34c-5p to ameliorate inflammatory response of coronary artery endothelial cells via PODXL-mediated P38 MAPK signaling pathway. Nutr Metab Cardiovasc Dis 2022; 32:2424-2438. [PMID: 36096977 DOI: 10.1016/j.numecd.2022.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/30/2022] [Accepted: 06/14/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND AND AIMS Low-grade chronic inflammation was reported to serve as a distinctive pathophysiologic feature of coronary artery disease (CAD), the leading cause of death around the world. Herein, the current study aimed to explore whether and how microRNA-34c-5p (miR-34c-5p), a miRNA enriched in extracellular vesicles (EVs) originated from the activated platelet (PLT-EVs), affects the inflammation of human coronary artery endothelial cells (HCAECs). METHODS AND RESULTS HCAECs were established as an in vitro cell model using oxidized low-density lipoprotein (ox-LDL). miR-34c-5p, an abundant miRNA in PLT-EVs, can be transferred to HCAECs and target PODXL by binding to its 3'UTR. Gain- and loss-of-function experiments of miR-34c-5p and podocalyxin (PODXL) were performed in ox-LDL-induced HCAECs. Subsequently, HCAECs were subjected to co-culture with PLT-EVs, followed by detection of the expression patterns of key pro-inflammatory factors. Either miR-34c-5p mimic or PLT-EVs harboring miR-34c-5p attenuated the ox-LDL-evoked inflammation in HCAECs by suppressing interleukin-1β (IL-1β), IL-6 and tumor necrosis factor-α (TNF-α). By blocking the P38 MAPK signaling pathway, miR-34c-5p-mediated depletion of PODXL contributed to protection against ox-LDL-induced inflammation. In vitro findings were further validated by findings observed in ApoE knock-out mice. Additionally, miR-34c-5p in PLT-EVs showed an athero-protective role in the murine model. CONCLUSION Altogether, our findings highlighted that miR-34c-5p in PLT-EVs could alleviate inflammation response in HCAECs by targeting PODXL and inactivation of the P38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Xuetao Bai
- Department of Anaesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, PR China
| | - Hao Zhang
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China
| | - Zhiguo Li
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China
| | - Ou Chen
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China
| | - Hengpeng He
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China
| | - Xiukun Jia
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China
| | - Lijuan Zou
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, PR China.
| |
Collapse
|
38
|
Vauclard A, Bellio M, Valet C, Borret M, Payrastre B, Severin S. Obesity: Effects on bone marrow homeostasis and platelet activation. Thromb Res 2022. [DOI: 10.1016/j.thromres.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
39
|
Liu L, Deng QJ. Role of platelet-derived extracellular vesicles in traumatic brain injury-induced coagulopathy and inflammation. Neural Regen Res 2022; 17:2102-2107. [PMID: 35259815 PMCID: PMC9083154 DOI: 10.4103/1673-5374.335825] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles are composed of fragments of exfoliated plasma membrane, organelles or nuclei and are released after cell activation, apoptosis or destruction. Platelet-derived extracellular vesicles are the most abundant type of extracellular vesicle in the blood of patients with traumatic brain injury. Accumulated laboratory and clinical evidence shows that platelet-derived extracellular vesicles play an important role in coagulopathy and inflammation after traumatic brain injury. This review discusses the recent progress of research on platelet-derived extracellular vesicles in coagulopathy and inflammation and the potential of platelet-derived extracellular vesicles as therapeutic targets for traumatic brain injury.
Collapse
Affiliation(s)
- Liang Liu
- Department of Neurosurgery, Tianjin Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Quan-Jun Deng
- Department of Neurosurgery, Tianjin Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
40
|
Sun G, Gu Q, Zheng J, Cheng H, Cheng T. Emerging roles of extracellular vesicles in normal and malignant hematopoiesis. J Clin Invest 2022; 132:160840. [PMID: 36106632 PMCID: PMC9479752 DOI: 10.1172/jci160840] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hematopoietic stem cells, regulated by their microenvironment (or “niche”), sustain the production of mature blood and immune cells. Leukemia cells remodel the microenvironment to enhance their survival, which is accompanied by the loss of support for normal hematopoiesis in hematologic malignancies. Extracellular vesicles (EVs) mediate intercellular communication in physiological and pathological conditions, and deciphering their functions in cell-cell interactions in the ecosystem can highlight potential therapeutic targets. In this Review, we illustrate the utility of EVs derived from various cell types, focusing on the biological molecules they contain and the behavioral alterations they can induce in recipient cells. We also discuss the potential for clinical application in hematologic malignancies, including EV-based therapeutic regimens, drug delivery via EVs, and the use of EVs (or their cargoes) as biomarkers.
Collapse
Affiliation(s)
- Guohuan Sun
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Quan Gu
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China
| |
Collapse
|
41
|
Al-Koussa H, AlZaim I, El-Sabban ME. Pathophysiology of Coagulation and Emerging Roles for Extracellular Vesicles in Coagulation Cascades and Disorders. J Clin Med 2022; 11:jcm11164932. [PMID: 36013171 PMCID: PMC9410115 DOI: 10.3390/jcm11164932] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
The notion of blood coagulation dates back to the ancient Greek civilization. However, the emergence of innovative scientific discoveries that started in the seventeenth century formulated the fundamentals of blood coagulation. Our understanding of key coagulation processes continues to evolve, as novel homeostatic and pathophysiological aspects of hemostasis are revealed. Hemostasis is a dynamic physiological process, which stops bleeding at the site of injury while maintaining normal blood flow within the body. Intrinsic and extrinsic coagulation pathways culminate in the homeostatic cessation of blood loss, through the sequential activation of the coagulation factors. Recently, the cell-based theory, which combines these two pathways, along with newly discovered mechanisms, emerged to holistically describe intricate in vivo coagulation mechanisms. The complexity of these mechanisms becomes evident in coagulation diseases such as hemophilia, Von Willebrand disease, thrombophilia, and vitamin K deficiency, in which excessive bleeding, thrombosis, or unnecessary clotting, drive the development and progression of diseases. Accumulating evidence implicates cell-derived and platelet-derived extracellular vesicles (EVs), which comprise microvesicles (MVs), exosomes, and apoptotic bodies, in the modulation of the coagulation cascade in hemostasis and thrombosis. As these EVs are associated with intercellular communication, molecular recycling, and metastatic niche creation, emerging evidence explores EVs as valuable diagnostic and therapeutic approaches in thrombotic and prothrombotic diseases.
Collapse
Affiliation(s)
- Houssam Al-Koussa
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Marwan E. El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Correspondence: ; Tel.: +961-01-350-000 (ext. 4765)
| |
Collapse
|
42
|
Eustes AS, Dayal S. The Role of Platelet-Derived Extracellular Vesicles in Immune-Mediated Thrombosis. Int J Mol Sci 2022; 23:7837. [PMID: 35887184 PMCID: PMC9320310 DOI: 10.3390/ijms23147837] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 12/14/2022] Open
Abstract
Platelet-derived extracellular vesicles (PEVs) play important roles in hemostasis and thrombosis. There are three major types of PEVs described based on their size and characteristics, but newer types may continue to emerge owing to the ongoing improvement in the methodologies and terms used to define various types of EVs. As the literature on EVs is growing, there are continuing attempts to standardize protocols for EV isolation and reach consensus in the field. This review provides information on mechanisms of PEV production, characteristics, cellular interaction, and their pathological role, especially in autoimmune and infectious diseases. We also highlight the mechanisms through which PEVs can activate parent cells in a feedback loop.
Collapse
Affiliation(s)
- Alicia S. Eustes
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Sanjana Dayal
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
- Iowa City VA Healthcare System, Iowa City, IA 52246, USA
| |
Collapse
|
43
|
Stone AP, Nikols E, Freire D, Machlus KR. The pathobiology of platelet and megakaryocyte extracellular vesicles: A (c)lot has changed. J Thromb Haemost 2022; 20:1550-1558. [PMID: 35506218 DOI: 10.1111/jth.15750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 11/28/2022]
Abstract
Platelet-derived extracellular vesicles (PEVs) were originally studied for their potential as regulators of coagulation, a function redundant with that of their parent cells. However, as the understanding of the diverse roles of platelets in hemostasis and disease has developed, so has the understanding of PEVs. In addition, the more recent revelation of constitutively released megakaryocyte-derived extracellular vesicles (MKEVs) in circulation provides an interesting counterpoint and avenue for investigation. In this review, we highlight the historical link of PEVs to thrombosis and hemostasis and provide critical updates. We also expand our discussion to encompass the roles that distinguish PEVs and MKEVs from their parent cells. Furthermore, the role of extracellular vesicles in disease pathology, both as biomarkers and as exacerbators, has been of great interest in recent years. We highlight some of the key roles that PEVs and MKEVs play in autoimmune blood cell disorders, liver pathology, and cardiovascular disease. We then look at the future of PEVs and MKEVs as candidates for novel therapeutics.
Collapse
Affiliation(s)
- Andrew P Stone
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Emma Nikols
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Daniela Freire
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Kellie R Machlus
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Leng Q, Ding J, Dai M, Liu L, Fang Q, Wang DW, Wu L, Wang Y. Insights Into Platelet-Derived MicroRNAs in Cardiovascular and Oncologic Diseases: Potential Predictor and Therapeutic Target. Front Cardiovasc Med 2022; 9:879351. [PMID: 35757325 PMCID: PMC9218259 DOI: 10.3389/fcvm.2022.879351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/24/2022] [Indexed: 11/28/2022] Open
Abstract
Non-communicable diseases (NCDs), represented by cardiovascular diseases and cancer, have been the leading cause of death globally. Improvements in mortality from cardiovascular (CV) diseases (decrease of 14%/100,000, United States) or cancers (increase 7.5%/100,000, United States) seem unsatisfactory during the past two decades, and so the search for innovative and accurate biomarkers of early diagnosis and prevention, and novel treatment strategies is a valuable clinical and economic endeavor. Both tumors and cardiovascular system are rich in angiological systems that maintain material exchange, signal transduction and distant regulation. This pattern determines that they are strongly influenced by circulating substances, such as glycolipid metabolism, inflammatory homeostasis and cyclic non-coding RNA and so forth. Platelets, a group of small anucleated cells, inherit many mature proteins, mRNAs, and non-coding RNAs from their parent megakaryocytes during gradual formation and manifest important roles in inflammation, angiogenesis, atherosclerosis, stroke, myocardial infarction, diabetes, cancer, and many other diseases apart from its classical function in hemostasis. MicroRNAs (miRNAs) are a class of non-coding RNAs containing ∼22 nucleotides that participate in many key cellular processes by pairing with mRNAs at partially complementary binding sites for post-transcriptional regulation of gene expression. Platelets contain fully functional miRNA processors in their microvesicles and are able to transport their miRNAs to neighboring cells and regulate their gene expression. Therefore, the importance of platelet-derived miRNAs for the human health is of increasing interest. Here, we will elaborate systematically the roles of platelet-derived miRNAs in cardiovascular disease and cancer in the hope of providing clinicians with new ideas for early diagnosis and therapeutic strategies.
Collapse
|
45
|
Extracellular Vesicles as Drivers of Immunoinflammation in Atherothrombosis. Cells 2022; 11:cells11111845. [PMID: 35681540 PMCID: PMC9180657 DOI: 10.3390/cells11111845] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023] Open
Abstract
Atherosclerotic cardiovascular disease is the leading cause of morbidity and mortality all over the world. Extracellular vesicles (EVs), small lipid-bilayer membrane vesicles released by most cellular types, exert pivotal and multifaceted roles in physiology and disease. Emerging evidence emphasizes the importance of EVs in intercellular communication processes with key effects on cell survival, endothelial homeostasis, inflammation, neoangiogenesis, and thrombosis. This review focuses on EVs as effective signaling molecules able to both derail vascular homeostasis and induce vascular dysfunction, inflammation, plaque progression, and thrombus formation as well as drive anti-inflammation, vascular repair, and atheroprotection. We provide a comprehensive and updated summary of the role of EVs in the development or regression of atherosclerotic lesions, highlighting the link between thrombosis and inflammation. Importantly, we also critically describe their potential clinical use as disease biomarkers or therapeutic agents in atherothrombosis.
Collapse
|
46
|
Zhang X, Tang J, Kou X, Huang W, Zhu Y, Jiang Y, Yang K, Li C, Hao M, Qu Y, Ma L, Chen C, Shi S, Zhou Y. Proteomic analysis of MSC-derived apoptotic vesicles identifies Fas inheritance to ameliorate haemophilia a via activating platelet functions. J Extracell Vesicles 2022; 11:e12240. [PMID: 36856683 PMCID: PMC9927920 DOI: 10.1002/jev2.12240] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/12/2022] Open
Abstract
Apoptotic vesicles (apoVs) are apoptotic cell-derived nanosized vesicles that play a crucial role in multiple pathophysiological settings. However, their detailed characteristics, specific surface markers, and biological properties are not fully elucidated. In this study, we compared mesenchymal stem cell (MSC)-derived apoVs and exosomes from three different types of MSCs including human bone marrow MSCs (hBMSCs), human adipose MSCs (hASCs), and mouse bone marrow MSCs (mBMSCs). We established a unique protein map of MSC-derived apoVs and identified the differences between apoVs and exosomes in terms of functional protein cargo and surface markers. Furthermore, we identified 13 proteins specifically enriched in apoVs compared to exosomes, which can be used as apoV-specific biomarkers. In addition, we showed that apoVs inherited apoptotic imprints such as Fas to ameliorate haemophilia A in factor VIII knockout mice via binding to the platelets' FasL to activate platelet functions, and therefore rescuing the blood clotting disorder. In summary, we systemically characterized MSC-derived apoVs and identified their therapeutic role in haemophilia A treatment through a previously unknown Fas/FasL linkage mechanism.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of ProsthodonticsPeking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081China
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Jianxia Tang
- Hunan Key Laboratory of Oral Health Research & Hunan Clinical Research Center of Oral Major Diseases and Oral HealthXiangya School of Stomatology, Xiangya Stomatological Hospital, Central South UniversityChangsha410000China
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Xiaoxing Kou
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat‐Sen University)Ministry of EducationGuangzhou510080China
| | - Weiying Huang
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Yuan Zhu
- Department of ProsthodonticsPeking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081China
| | - Yuhe Jiang
- Department of ProsthodonticsPeking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081China
| | - Kunkun Yang
- Department of ProsthodonticsPeking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081China
| | - Can Li
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Meng Hao
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Yan Qu
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Lan Ma
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and PharmacologyUniversity of Pennsylvania, School of Dental MedicinePhiladelphiaPA 19104USA
| | - Songtao Shi
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat‐Sen University)Ministry of EducationGuangzhou510080China
| | - Yongsheng Zhou
- Department of ProsthodonticsPeking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081China
| |
Collapse
|
47
|
Zhang F, Isak AN, Yang S, Song Y, Ren L, Feng C, Chen G. Smartly responsive DNA-miRNA hybrids packaged in exosomes for synergistic enhancement of cancer cell apoptosis. NANOSCALE 2022; 14:6612-6619. [PMID: 35421879 DOI: 10.1039/d1nr08539e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Endogenous and exogenous tumor-related microRNAs (miRNAs) are considered promising tumor biomarkers and tumor therapeutic agents. In this work, we propose a miRNA self-responsive drug delivery system (miR-SR DDS), which enables the association between endogenous and exogenous miRNAs, so as to achieve a smart responsive and synergistic drug delivery. The miR-SR DDS consists of DNA-miRNA hybrids of let-7a and the complementary DNA of miR-155, which was packaged in exosomes. In response to the overexpressed miR-155 in breast cancer cells, the hybrids disintegrate and release let-7a and the complementary DNA of miR-155 to inhibit the expression of HMGA1 and relieve the inhibition of SOX1, respectively. Under the dual-targeted gene regulation, results show that the growth, migration and invasion of breast cancer cells can be synergistically inhibited through the Wnt/β-catenin signaling pathway. The concept and successful practice of the miR-SR DDS can be used as a reference for the development of miRNA drugs.
Collapse
Affiliation(s)
- Fan Zhang
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Albertina N Isak
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Shiqi Yang
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Yuchen Song
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Lingjie Ren
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Chang Feng
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Guifang Chen
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| |
Collapse
|
48
|
Huang DY, Wang GM, Ke ZR, Zhou Y, Yang HH, Ma TL, Guan CX. Megakaryocytes in pulmonary diseases. Life Sci 2022; 301:120602. [DOI: 10.1016/j.lfs.2022.120602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023]
|
49
|
Luo H, Ye G, Liu Y, Huang D, Luo Q, Chen W, Qi Z. miR-150-3p enhances neuroprotective effects of neural stem cell exosomes after hypoxic-ischemic brain injury by targeting CASP2. Neurosci Lett 2022; 779:136635. [DOI: 10.1016/j.neulet.2022.136635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 12/11/2022]
|
50
|
Garcia C, Compagnon B, Poëtte M, Gratacap MP, Lapébie FX, Voisin S, Minville V, Payrastre B, Vardon-Bounes F, Ribes A. Platelet Versus Megakaryocyte: Who Is the Real Bandleader of Thromboinflammation in Sepsis? Cells 2022; 11:1507. [PMID: 35563812 PMCID: PMC9104300 DOI: 10.3390/cells11091507] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
Platelets are mainly known for their key role in hemostasis and thrombosis. However, studies over the last two decades have shown their strong implication in mechanisms associated with inflammation, thrombosis, and the immune system in various neoplastic, inflammatory, autoimmune, and infectious diseases. During sepsis, platelets amplify the recruitment and activation of innate immune cells at the site of infection and contribute to the elimination of pathogens. In certain conditions, these mechanisms can lead to thromboinflammation resulting in severe organ dysfunction. Here, we discuss the interactions of platelets with leukocytes, neutrophil extracellular traps (NETs), and endothelial cells during sepsis. The intrinsic properties of platelets that generate an inflammatory signal through the NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome are discussed. As an example of immunothrombosis, the implication of platelets in vaccine-induced immune thrombotic thrombocytopenia is documented. Finally, we discuss the role of megakaryocytes (MKs) in thromboinflammation and their adaptive responses.
Collapse
Affiliation(s)
- Cédric Garcia
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| | - Baptiste Compagnon
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Michaël Poëtte
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Marie-Pierre Gratacap
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| | - François-Xavier Lapébie
- Service de Médecine Vasculaire, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Sophie Voisin
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
| | - Vincent Minville
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Bernard Payrastre
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| | - Fanny Vardon-Bounes
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Agnès Ribes
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| |
Collapse
|