1
|
Gaceb A, Roupé L, Enström A, Almasoudi W, Carlsson R, Lindgren AG, Paul G. Pericyte Microvesicles as Plasma Biomarkers Reflecting Brain Microvascular Signaling in Patients With Acute Ischemic Stroke. Stroke 2024; 55:558-568. [PMID: 38323422 PMCID: PMC10896197 DOI: 10.1161/strokeaha.123.045720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/07/2024] [Accepted: 01/19/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Blood-based biomarkers have the potential to reflect cerebrovascular signaling after microvascular injury; yet, the detection of cell-specific signaling has proven challenging. Microvesicles retain parental cell surface antigens allowing detection of cell-specific signaling encoded in their cargo. In ischemic stroke, the progression of pathology involves changes in microvascular signaling whereby brain pericytes, perivascular cells wrapping the microcapillaries, are one of the early responders to the ischemic insult. Intercepting the pericyte signaling response peripherally by isolating pericyte-derived microvesicles may provide not only diagnostic information on microvascular injury but also enable monitoring of important pathophysiological mechanisms. METHODS Plasma samples were collected from patients with acute ischemic stroke (n=39) at 3 time points after stroke onset: 0 to 6 hours, 12 to 24 hours, and 2 to 6 days, and compared with controls (n=39). Pericyte-derived microvesicles were isolated based on cluster of differentiation 140b expression and quantified by flow cytometry. The protein content was evaluated using a proximity extension assay, and vascular signaling pathways were examined using molecular signature hallmarks and gene ontology. RESULTS In this case-control study, patients with acute ischemic stroke showed significantly increased numbers of pericyte-derived microvesicles (median, stroke versus controls) at 12 to 24 hours (1554 versus 660 microvesicles/μL; P=0.0041) and 2 to 6 days after stroke (1346 versus 660 microvesicles/μL; P=0.0237). Their proteome revealed anti-inflammatory properties mediated via downregulation of Kirsten rat sarcoma virus and IL (interleukin)-6/JAK/STAT3 signaling at 0 to 6 hours, but proangiogenic as well as proinflammatory signals at 12 to 24 hours. Between 2 and 6 days, proteins were mainly associated with vascular remodeling as indicated by activation of Hedgehog signaling in addition to proangiogenic signals. CONCLUSIONS We demonstrate that the plasma of patients with acute ischemic stroke reflects (1) an early and time-dependent increase of pericyte-derived microvesicles and (2) changes in the protein cargo of microvesicles over time indicating cell signaling specifically related to inflammation and vascular remodeling.
Collapse
Affiliation(s)
- Abderahim Gaceb
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center (A.G., A.E., R.C., G.P.), Lund University, Sweden
| | - Linnea Roupé
- Department of Neurology, Scania University Hospital, Lund, Sweden (L.R., W.A., A.G.L., G.P.)
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center (A.G., A.E., R.C., G.P.), Lund University, Sweden
| | - Wejdan Almasoudi
- Department of Neurology, Scania University Hospital, Lund, Sweden (L.R., W.A., A.G.L., G.P.)
| | - Robert Carlsson
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center (A.G., A.E., R.C., G.P.), Lund University, Sweden
| | - Arne G. Lindgren
- Department of Neurology, Scania University Hospital, Lund, Sweden (L.R., W.A., A.G.L., G.P.)
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center (A.G., A.E., R.C., G.P.), Lund University, Sweden
- Wallenberg Center for Molecular Medicine (G.P.), Lund University, Sweden
- Department of Neurology, Scania University Hospital, Lund, Sweden (L.R., W.A., A.G.L., G.P.)
| |
Collapse
|
2
|
Li S, Zheng S, Li J, Lin S, Li H, Wang P, Chen P, Ma C, Liu Y. Research progress on extracellular vesicles in the renal tubular injury of diabetic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1257430. [PMID: 37732129 PMCID: PMC10507342 DOI: 10.3389/fendo.2023.1257430] [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: 07/12/2023] [Accepted: 08/08/2023] [Indexed: 09/22/2023] Open
Abstract
Diabetic kidney disease (DKD) is a severe microvascular complication of diabetes and is a chronic progressive condition. It is also a common cause of end-stage renal disease (ESRD), which is characterized by proteinuria or a progressive decline in the glomerular filtration rate. Due to their dependence on high-energy and aerobic metabolism, renal tubules are more susceptible to the metabolic disturbances associated with DKD, leading to inflammation and fibrosis. Consequently, tubular injury has become a recent research focus, and significant advancements have been made in studying the role of extracellular vesicles in DKD-associated tubular injury. This review aimed to elucidate the mechanisms and potential applications of different types of extracellular vesicles in tubular injury in DKD to provide new insights for the prevention and treatment of DKD.
Collapse
Affiliation(s)
- Shengjie Li
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Shanshan Zheng
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jiao Li
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Nephrology, Binzhou People’s Hospital Affiliated to Shandong First Medical University, Binzhou, China
| | - Sen Lin
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Hao Li
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Ping Wang
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Nephrology Research Institute of Shandong Province, Jinan, China
| | - Ping Chen
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Nephrology Research Institute of Shandong Province, Jinan, China
| | - Chaoqun Ma
- Department of Emergency Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yipeng Liu
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Nephrology Research Institute of Shandong Province, Jinan, China
| |
Collapse
|
3
|
Mabrouk M, Guessous F, Naya A, Merhi Y, Zaid Y. The Pathophysiological Role of Platelet-Derived Extracellular Vesicles. Semin Thromb Hemost 2023; 49:279-283. [PMID: 36174608 DOI: 10.1055/s-0042-1756705] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Platelets are very abundant in the blood, where they play a role in hemostasis, inflammation, and immunity. When activated, platelets undergo a conformational change that allows the release of numerous effector molecules as well as the production of extracellular vesicles, which are circulating submicron vesicles (10 to 1,000 nm in diameter) released into the extracellular space. Extracellular vesicles are formed by the budding of platelet and they carry some of its contents, including nucleic acids, surface proteins, and organelles. While platelets cannot cross tissue barriers, platelet-derived extracellular vesicles can enter the lymph, bone marrow, and synovial fluid. This allows the transfer of diverse contents carried by these platelet-derived vesicles to cell recipients and organs inaccessible to platelets where they can perform many functions. This review highlights the importance of these platelet-derived extracellular vesicles under different physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Meryem Mabrouk
- Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.,Department of Biology, Faculty of Sciences, Immunology and Biodiversity Laboratory, Hassan II University, Casablanca, Morocco
| | - Fadila Guessous
- Research of Center, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Abdallah Naya
- Department of Biology, Faculty of Sciences, Immunology and Biodiversity Laboratory, Hassan II University, Casablanca, Morocco
| | - Yahye Merhi
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Center, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Younes Zaid
- Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.,Department of Biology, Faculty of Sciences, Immunology and Biodiversity Laboratory, Hassan II University, Casablanca, Morocco
| |
Collapse
|
4
|
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
|
5
|
Maphumulo SC, Pretorius E. Role of Circulating Microparticles in Type 2 Diabetes Mellitus: Implications for Pathological Clotting. Semin Thromb Hemost 2021; 48:188-205. [PMID: 34959250 DOI: 10.1055/s-0041-1740150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a multifactorial chronic metabolic disease characterized by chronic hyperglycemia due to insulin resistance and a deficiency in insulin secretion. The global diabetes pandemic relates primarily to T2DM, which is the most prevalent form of diabetes, accounting for over 90% of all cases. Chronic low-grade inflammation, triggered by numerous risk factors, and the chronic activation of the immune system are prominent features of T2DM. Here we highlight the role of blood cells (platelets, and red and white blood cells) and vascular endothelial cells as drivers of systemic inflammation in T2DM. In addition, we discuss the role of microparticles (MPs) in systemic inflammation and hypercoagulation. Although once seen as inert by-products of cell activation or destruction, MPs are now considered to be a disseminated storage pool of bioactive effectors of thrombosis, inflammation, and vascular function. They have been identified to circulate at elevated levels in the bloodstream of individuals with increased risk of atherothrombosis or cardiovascular disease, two significant hallmark conditions of T2DM. There is also general evidence that MPs activate blood cells, express proinflammatory and coagulant effects, interact directly with cell receptors, and transfer biological material. MPs are considered major players in the pathogenesis of many systemic inflammatory diseases and may be potentially useful biomarkers of disease activity and may not only be of prognostic value but may act as novel therapeutic targets.
Collapse
Affiliation(s)
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
6
|
Zhou H, Huang L, Liang L, Chen L, Zou C, Li Z, Li R, Jian C, Zou D. Identification of an miRNA Regulatory Network and Candidate Markers for Ischemic Stroke Related to Diabetes. Int J Gen Med 2021; 14:3213-3223. [PMID: 34262334 PMCID: PMC8274709 DOI: 10.2147/ijgm.s319503] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose Type 2 diabetes mellitus (T2DM) increases the risk of ischemic stroke and poor prognosis. This study aimed to identify molecular mechanisms that are dysregulated in T2DM-associated ischemic stroke and candidate genes that might serve as biomarkers. Methods The top 25% variance genes in the GSE21321 and GSE22255 datasets were analyzed for coexpression. The differentially expressed mRNAs (DEmRs) between patients with T2DM or ischemic stroke and controls were analyzed. Then, the union of overlapping coexpressed genes and overlapping DEmRs was analyzed. The miRNAs differentially expressed in T2DM-associated ischemic stroke were also analyzed. CIBERSORT was used to evaluate the levels of infiltration by immune cells in T2DM-associated stroke. Results Thirteen coexpression modules were identified in T2DM and 10 in ischemic stroke, and 594 module genes were shared between the two conditions. A total of 4452 mRNAs differentially expressed between T2DM patients and controls were identified, as were 2390 mRNAs differentially expressed between ischemic stroke and controls. The 771 union genes were enriched mainly in immune-related biological functions and signaling pathways. UBE2N, TGFB3, EXOSC1, and VIM were identified as candidate markers. In addition, we identified miR-576-3p as having the most regulatory roles in both T2DM and ischemic stroke. Mast cell activation was significantly down-regulated in T2DM but up-regulated in ischemic stroke. Conclusion These findings provide numerous testable hypotheses about the pathways underlying T2DM-associated ischemic stroke, which may help identify therapeutic targets.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Neurology, The People's Hospital of Guiping, Guiping, Guangxi, 537200, People's Republic of China
| | - Liujia Huang
- Department of Rehabilitation Medicine, The People's Hospital of Guiping, Guiping, Guangxi, 537200, People's Republic of China
| | - Lucong Liang
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China
| | - Liechun Chen
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China
| | - Chun Zou
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China
| | - Zhenhua Li
- Department of Emergency Medicine, The First People's Hospital of Nanning, Nanning, 530022, People's Republic of China
| | - Rongjie Li
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China
| | - Chongdong Jian
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, People's Republic of China
| | - Donghua Zou
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530022, People's Republic of China
| |
Collapse
|
7
|
Noulsri E, Lerdwana S, Palasuwan D, Palasuwan A. Cell-Derived Microparticles in Blood Products from Blood Donors Deficient in Glucose-6-Phosphate Dehydrogenase. Lab Med 2021; 52:528-535. [PMID: 33693844 DOI: 10.1093/labmed/lmab007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To quantitate the microparticles (MPs) in whole blood and blood products obtained from blood donors who are deficient in glucose-6-phosphate dehydrogenase (G6PD). METHODS The current study analyzed whole blood and blood components prepared from 49 blood donors with G6PD deficiencies and 98 with G6PD-normal results. Packed red blood cells (PRBCs), platelet concentrate (PC), and plasma were prepared according to transfusion laboratory procedures. MP concentrations were determined using a flow cytometer. RESULTS Blood components prepared from donors with G6PD deficiency were characterized by higher red blood cell-derived MP (RMP) concentration in PRBCs (25,526 vs 18,738 particles/µL) but lower concentrations of platelet-derived MPs (PMPs; in whole blood and PC), leukocyte-derived MPs (LMP; in whole blood and plasma) and total MP (in PC), compared with those from donors with G6PD-normal test results. CONCLUSIONS These results suggest that differences in G6PD status may account for variation in RMP levels during processing.
Collapse
Affiliation(s)
- Egarit Noulsri
- Research Division and Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surada Lerdwana
- Biomedical Research Incubator Unit, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Duangdao Palasuwan
- Oxidation in Red Cell Disorders and Health Task Force, Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Attakorn Palasuwan
- Oxidation in Red Cell Disorders and Health Task Force, Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
8
|
Osman A, El-Gamal H, Pasha M, Zeidan A, Korashy HM, Abdelsalam SS, Hasan M, Benameur T, Agouni A. Endoplasmic Reticulum (ER) Stress-Generated Extracellular Vesicles (Microparticles) Self-Perpetuate ER Stress and Mediate Endothelial Cell Dysfunction Independently of Cell Survival. Front Cardiovasc Med 2020; 7:584791. [PMID: 33363219 PMCID: PMC7758248 DOI: 10.3389/fcvm.2020.584791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/09/2020] [Indexed: 12/16/2022] Open
Abstract
Circulating extracellular vesicles (EVs) are recognized as biomarkers and effectors of endothelial dysfunction, the initiating step of cardiovascular abnormalities. Among these EVs, microparticles (MPs) are vesicles directly released from the cytoplasmic membrane of activated cells. MPs were shown to induce endothelial dysfunction through the activation of endoplasmic reticulum (ER) stress. However, it is not known whether ER stress can lead to MPs release from endothelial cells and what biological messages are carried by these MPs. Therefore, we aimed to assess the impact of ER stress on MPs shedding from endothelial cells, and to investigate their effects on endothelial cell function. EA.hy926 endothelial cells or human umbilical vein endothelial cells (HUVECs) were treated for 24 h with ER stress inducers, thapsigargin or dithiothreitol (DTT), in the presence or absence of 4-Phenylbutyric acid (PBA), a chemical chaperone to inhibit ER stress. Then, MPs were isolated and used to treat cells (10–20 μg/mL) for 24–48 h before assessing ER stress response, angiogenic capacity, nitric oxide (NO) release, autophagy and apoptosis. ER stress (thapsigargin or DDT)-generated MPs did not differ quantitatively from controls; however, they carried deleterious messages for endothelial function. Exposure of endothelial cells to ER stress-generated MPs increased mRNA and protein expression of key ER stress markers, indicating a vicious circle activation of ER stress. ER stress (thapsigargin)-generated MPs impaired the angiogenic capacity of HUVECs and reduced NO release, indicating an impaired endothelial function. While ER stress (thapsigargin)-generated MPs altered the release of inflammatory cytokines, they did not, however, affect autophagy or apoptosis in HUVECs. This work enhances the general understanding of the deleterious effects carried out by MPs in medical conditions where ER stress is sustainably activated such as diabetes and metabolic syndrome.
Collapse
Affiliation(s)
- Aisha Osman
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Heba El-Gamal
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Mazhar Pasha
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Asad Zeidan
- Department of Basic Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Shahenda S Abdelsalam
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Maram Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Tarek Benameur
- College of Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
9
|
Nazari M, Javandoost E, Talebi M, Movassaghpour A, Soleimani M. Platelet Microparticle Controversial Role in Cancer. Adv Pharm Bull 2020; 11:39-55. [PMID: 33747851 PMCID: PMC7961228 DOI: 10.34172/apb.2021.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
Platelet-derived microparticles (PMPs) are a group of micrometer-scale extracellular vesicles released by platelets upon activation that are responsible for the majority of microvesicles found in plasma. PMPs’ physiological properties and functions have long been investigated by researchers. In this regard, a noticeable area of studies has been devoted to evaluating the potential roles and effects of PMPs on cancer progression. Clinical and experimental evidence conflictingly implicates supportive and suppressive functions for PMPs regarding cancer. Many of these functions could be deemed as a cornerstone for future considerations of PMPs usage in cancer targeted therapy. This review discusses what is currently known about PMPs and provides insights for new and possible research directions for further grasping the intricate interplay between PMPs and cancer.
Collapse
Affiliation(s)
- Mahnaz Nazari
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsan Javandoost
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Talebi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran. Introduction
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
10
|
Osman A, Benameur T, Korashy HM, Zeidan A, Agouni A. Interplay between Endoplasmic Reticulum Stress and Large Extracellular Vesicles (Microparticles) in Endothelial Cell Dysfunction. Biomedicines 2020; 8:E409. [PMID: 33053883 PMCID: PMC7599704 DOI: 10.3390/biomedicines8100409] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 09/26/2020] [Accepted: 10/03/2020] [Indexed: 12/19/2022] Open
Abstract
Upon increased demand for protein synthesis, accumulation of misfolded and/or unfolded proteins within the endoplasmic reticulum (ER), a pro-survival response is activated termed unfolded protein response (UPR), aiming at restoring the proper function of the ER. Prolonged activation of the UPR leads, however, to ER stress, a cellular state that contributes to the pathogenesis of various chronic diseases including obesity and diabetes. ER stress response by itself can result in endothelial dysfunction, a hallmark of cardiovascular disease, through various cellular mechanisms including apoptosis, insulin resistance, inflammation and oxidative stress. Extracellular vesicles (EVs), particularly large EVs (lEVs) commonly referred to as microparticles (MPs), are membrane vesicles. They are considered as a fingerprint of their originating cells, carrying a variety of molecular components of their parent cells. lEVs are emerging as major contributors to endothelial cell dysfunction in various metabolic disease conditions. However, the mechanisms underpinning the role of lEVs in endothelial dysfunction are not fully elucidated. Recently, ER stress emerged as a bridging molecular link between lEVs and endothelial cell dysfunction. Therefore, in the current review, we summarized the roles of lEVs and ER stress in endothelial dysfunction and discussed the molecular crosstalk and relationship between ER stress and lEVs in endothelial dysfunction.
Collapse
Affiliation(s)
- Aisha Osman
- Department of Pharmaceutical Sciences, College of Pharmacy, QU health, Qatar University, Doha 2713, Qatar; (A.O.); (H.M.K.)
| | - Tarek Benameur
- Department of Biomedical Sciences, College of Medicine, King Faisal University, P.O. Box 400, Al Ahsa 31982, Saudi Arabia;
| | - Hesham M. Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU health, Qatar University, Doha 2713, Qatar; (A.O.); (H.M.K.)
| | - Asad Zeidan
- Department of Basic Medical Sciences, College of Medicine, QU health, Qatar University, Doha 2713, Qatar;
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU health, Qatar University, Doha 2713, Qatar; (A.O.); (H.M.K.)
| |
Collapse
|
11
|
Daryabor G, Atashzar MR, Kabelitz D, Meri S, Kalantar K. The Effects of Type 2 Diabetes Mellitus on Organ Metabolism and the Immune System. Front Immunol 2020; 11:1582. [PMID: 32793223 PMCID: PMC7387426 DOI: 10.3389/fimmu.2020.01582] [Citation(s) in RCA: 223] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
Metabolic abnormalities such as dyslipidemia, hyperinsulinemia, or insulin resistance and obesity play key roles in the induction and progression of type 2 diabetes mellitus (T2DM). The field of immunometabolism implies a bidirectional link between the immune system and metabolism, in which inflammation plays an essential role in the promotion of metabolic abnormalities (e.g., obesity and T2DM), and metabolic factors, in turn, regulate immune cell functions. Obesity as the main inducer of a systemic low-level inflammation is a main susceptibility factor for T2DM. Obesity-related immune cell infiltration, inflammation, and increased oxidative stress promote metabolic impairments in the insulin-sensitive tissues and finally, insulin resistance, organ failure, and premature aging occur. Hyperglycemia and the subsequent inflammation are the main causes of micro- and macroangiopathies in the circulatory system. They also promote the gut microbiota dysbiosis, increased intestinal permeability, and fatty liver disease. The impaired immune system together with metabolic imbalance also increases the susceptibility of patients to several pathogenic agents such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Thus, the need for a proper immunization protocol among such patients is granted. The focus of the current review is to explore metabolic and immunological abnormalities affecting several organs of T2DM patients and explain the mechanisms, whereby diabetic patients become more susceptible to infectious diseases.
Collapse
Affiliation(s)
- Gholamreza Daryabor
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohamad Reza Atashzar
- Department of Immunology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | | | - Seppo Meri
- Department of Bacteriology and Immunology and the Translational Immunology Research Program (TRIMM), The University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Kurosh Kalantar
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Marcoux G, Laroche A, Espinoza Romero J, Boilard E. Role of platelets and megakaryocytes in adaptive immunity. Platelets 2020; 32:340-351. [PMID: 32597341 DOI: 10.1080/09537104.2020.1786043] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system is comprised of two principal interconnected components called innate and adaptive immunity. While the innate immune system mounts a nonspecific response that provides protection against the spread of foreign pathogens, the adaptive immune system has developed to specifically recognize a given pathogen and lead to immunological memory. Platelets are small fragments produced from megakaryocytes in bone marrow and lungs. They circulate throughout the blood to monitor the integrity of the vasculature and to prevent bleeding. Given their large repertoire of immune receptors and inflammatory molecules, platelets and megakaryocytes can contribute to both innate and adaptive immunity. In adaptive immunity, platelets and megakaryocytes can process and present antigens to lymphocytes. Moreover, platelets, via FcγRIIA, rapidly respond to pathogens in an immune host when antibodies are present. This manuscript reviews the reported contributions of platelets and megakaryocytes with emphasis on antigen presentation and antibody response in adaptive immunity.
Collapse
Affiliation(s)
- Genevieve Marcoux
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Audrée Laroche
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Jenifer Espinoza Romero
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Eric Boilard
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| |
Collapse
|
13
|
Lordan R, Tsoupras A, Zabetakis I. Platelet activation and prothrombotic mediators at the nexus of inflammation and atherosclerosis: Potential role of antiplatelet agents. Blood Rev 2020; 45:100694. [PMID: 32340775 DOI: 10.1016/j.blre.2020.100694] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 03/22/2020] [Accepted: 04/07/2020] [Indexed: 12/20/2022]
Abstract
Platelets are central to inflammation-related manifestations of cardiovascular diseases (CVD) such as atherosclerosis. Platelet-activating factor (PAF), thrombin, thromboxane A2 (TxA2), and adenosine diphosphate (ADP) are some of the key agonists of platelet activation that are at the intersection between a plethora of inflammatory pathways that modulate pro-inflammatory and coagulation processes. The aim of this article is to review the role of platelets and the relationship between their structure, function, and the interactions of their constituents in systemic inflammation and atherosclerosis. Antiplatelet therapies are discussed with a view to primary prevention of CVD by the clinical reduction of platelet reactivity and inflammation. Current antiplatelet therapies are effective in reducing cardiovascular risk but increase bleeding risk. Novel therapeutic antiplatelet approaches beyond current pharmacological modalities that do not increase the risk of bleeding require further investigation. There is potential for specifically designed nutraceuticals that may become safer alternatives to pharmacological antiplatelet agents for the primary prevention of CVD but there is serious concern over their efficacy and regulation, which requires considerably more research.
Collapse
Affiliation(s)
- Ronan Lordan
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158, USA.
| | - Alexandros Tsoupras
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| | - Ioannis Zabetakis
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| |
Collapse
|
14
|
Xi XX, Sun J, Chen HC, Chen AD, Gao LP, Yin J, Jing YH. High-Fat Diet Increases Amylin Accumulation in the Hippocampus and Accelerates Brain Aging in hIAPP Transgenic Mice. Front Aging Neurosci 2019; 11:225. [PMID: 31507407 PMCID: PMC6718729 DOI: 10.3389/fnagi.2019.00225] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/08/2019] [Indexed: 12/23/2022] Open
Abstract
The accumulation of human islet amyloid polypeptide (hIAPP) in pancreatic islets under induction by a high-fat diet plays a critical role in the development of type-2 diabetes mellitus (T2DM). T2DM is a risk factor of late-onset Alzheimer’s disease (AD). Nevertheless, whether hIAPP in combination with hyperlipidemia may lead to AD-like pathological changes in the brain remains unclear. hIAPP transgenic mice were fed with a high-fat diet for 6 or 12 months to establish the T2DM model. The accumulation of amylin, the numbers of Fluoro-Jade C (FJC)-positive and β-gal positive cells, and the deposition level of Aβ42 in the hippocampi of the transgenic mice were detected by using brain sections. Cytoplasmic and membrane proteins were extracted from the hippocampi of the transgenic mice, and the ratio of membrane GLUT4 expression to cytoplasmic GLUT4 expression was measured through Western blot analysis. Changes in the cognitive functions of hIAPP transgenic mice after 12 months of feeding with a high-fat diet were evaluated. hIAPP transgenic mice fed with a high-fat diet for 6 or 12 months showed elevated blood glucose levels and insulin resistance; increased amylin accumulation, number of FJC-positive and β-gal positive cells, and Aβ42 deposition in the hippocampi; and reduced membrane GLUT4 expression levels. hIAPP transgenic mice fed with a high-fat diet for 12 months showed reductions in social cognitive ability and passive learning ability. A high-fat diet increased amylin accumulation in the hippocampi of hIAPP transgenic mice, which presented AD-like pathology and behavior characterized by neural degeneration, brain aging, Aβ42 deposition, and impaired glucose utilization and cognition.
Collapse
Affiliation(s)
- Xiao-Xia Xi
- Center of Experimental Animal, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jing Sun
- Center of Experimental Animal, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Hai-Chao Chen
- School of Basic Medical Sciences, Institute of Anatomy and Histology & Embryology, Neuroscience, Lanzhou University, Lanzhou, China
| | - An-Di Chen
- School of Basic Medical Sciences, Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou, China
| | - Li-Ping Gao
- School of Basic Medical Sciences, Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou, China
| | - Jie Yin
- School of Basic Medical Sciences, Institute of Anatomy and Histology & Embryology, Neuroscience, Lanzhou University, Lanzhou, China
| | - Yu-Hong Jing
- School of Basic Medical Sciences, Institute of Anatomy and Histology & Embryology, Neuroscience, Lanzhou University, Lanzhou, China.,Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
15
|
Microparticles as Potential Mediators of High Glucose-Induced Renal Cell Injury. Biomolecules 2019; 9:biom9080348. [PMID: 31390845 PMCID: PMC6723350 DOI: 10.3390/biom9080348] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/31/2019] [Accepted: 08/04/2019] [Indexed: 12/18/2022] Open
Abstract
Diabetic nephropathy (DN) is the most common cause of chronic kidney disease worldwide. Activation of signaling pathways such as the mammalian target of rapamycin (mTOR), extracellular signal-regulated kinases (ERK), endoplasmic reticulum (ER) stress, transforming growth factor-beta (TGF-β), and epithelial-mesenchymal transition (EMT), are thought to play a significant role in the etiology of DN. Microparticles (MPs), the small membrane vesicles containing bioactive signals shed by cells upon activation or during apoptosis, are elevated in diabetes and were identified as biomarkers in DN. However, their exact role in the pathophysiology of DN remains unclear. Here, we examined the effect of MPs shed from renal proximal tubular cells (RPTCs) exposed to high glucose conditions on naïve RPTCs in vitro. Our results showed significant increases in the levels of phosphorylated forms of 4E-binding protein 1 and ERK1/2 (the downstream targets of mTOR and ERK pathways), phosphorylated-eIF2α (an ER stress marker), alpha smooth muscle actin (an EMT marker), and phosphorylated-SMAD2 and nuclear translocation of SMAD4 (markers of TGF-β signaling). Together, our findings indicate that MPs activate key signaling pathways in RPTCs under high glucose conditions. Pharmacological interventions to inhibit shedding of MPs from RPTCs might serve as an effective strategy to prevent the progression of DN.
Collapse
|