1
|
Wurtzel JGT, Gray BD, Pak KY, Zhao X, Ma P, McKenzie SE, Tanujaya M, Rizzo V, Del Carpio-Cano F, Rao AK, Lee-Gau Chong P, Goldfinger LE. Phosphatidylserine-blocking nanoparticles inhibit thrombosis without increased bleeding in mice. J Thromb Haemost 2024:S1538-7836(24)00613-5. [PMID: 39423958 DOI: 10.1016/j.jtha.2024.10.007] [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: 07/23/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Phosphatidylserine (PS) is a procoagulant phospholipid enriched on surfaces of activated vascular cells including platelets, endothelium, monocytes, and microvesicles. As a molecular driver of thrombosis accessible to drug blockade, PS is an attractive pharmacologic target for modulating thrombogenesis, with potentially reduced bleeding risk compared to anticoagulant and antiplatelet therapies. OBJECTIVES Test antithrombotic capabilities of a liposomal formulation, Zn-dipicolylamine cyanine-3[22,22]/1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (molar ratio, 3:97), designated as DPAL, which we previously described binds selectively to PS-enriched cell surfaces, compared with effects on bleeding, in mouse models. METHODS PS-dependent DPAL binding to human and murine platelets was tested in vitro. Thrombosis and bleeding after DPAL intravenous administration were tested in C57Bl/6J mice following FeCl3 carotid arterial injury and tail tip amputation, respectively. Incorporation in hemostatic clots was investigated in the cremaster muscle laser injury model. Toxicity was tested by direct exposure to human endothelial cell cultures. RESULTS DPAL bound agonist-stimulated, PS-positive human and murine platelets, blocked by Annexin V or Ano6 deletion, which ablate PS exposure. DPAL prolonged prothrombin time, but did not prevent thrombin-induced fibrinogen receptor activation or aggregation, nor alter blood cell counts including platelets. Following arteriolar laser injury, DPAL bound wound surfaces and edges without destabilizing plugs. DPAL dose-dependently blocked FeCl3-induced arterial thrombosis but did not substantially increase bleeding, or induce endothelial cell death. CONCLUSION DPAL reduces thrombogenesis with minimal effects on bleeding in mouse models via selective binding to PS. DPAL may support novel approaches to modulate pathogenic thrombin generation with improved safety profiles in multiple contexts.
Collapse
Affiliation(s)
- Jeremy G T Wurtzel
- Cardeza Foundation for Hematologic Research, Department of Medicine, Division of Hematology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Brian D Gray
- Molecular Targeting Technologies Inc, West Chester, Pennsylvania, USA
| | - Koon Y Pak
- Molecular Targeting Technologies Inc, West Chester, Pennsylvania, USA
| | - Xuefei Zhao
- Cardeza Foundation for Hematologic Research, Department of Medicine, Division of Hematology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Peisong Ma
- Cardeza Foundation for Hematologic Research, Department of Medicine, Division of Hematology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Steven E McKenzie
- Cardeza Foundation for Hematologic Research, Department of Medicine, Division of Hematology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Michelle Tanujaya
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Fabiola Del Carpio-Cano
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - A Koneti Rao
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA; Department of Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Parkson Lee-Gau Chong
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Lawrence E Goldfinger
- Cardeza Foundation for Hematologic Research, Department of Medicine, Division of Hematology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
2
|
Kriebardis AG, Chardalias L, Damaskos C, Pouliakis A, Garmpis N, Fortis SP, Papailia A, Sideri C, Georgatzakou HT, Papageorgiou EG, Pittaras T, Tsourouflis G, Politou M, Papaconstantinou I, Dimitroulis D, Valsami S. Precision Oncology: Circulating Microvesicles as New Biomarkers in a Very Early Stage of Colorectal Cancer. Cancers (Basel) 2024; 16:1943. [PMID: 38792021 PMCID: PMC11119677 DOI: 10.3390/cancers16101943] [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: 04/20/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND The release of microvesicles (MVs) is an essential phenomenon for inter-cellular signaling in health and disease. The role of MVs in cancer is multidimensional and includes cancer cell survival, proliferation, and invasion. In this prospective study, we analyzed MV levels in colorectal cancer patients and assessed the importance of MV release in early-stage colorectal cancer and survival. METHODS This study included 98 patients and 15 controls. The characterization of MVs from human plasma was performed by flow cytometry using monoclonal antibodies. RESULTS The levels of total MVs and MUC-1-positive, tissue factor (TF)-positive, and endothelial cell-derived MVs (EMVs) were statistically significantly higher in the colon cancer patients than in the controls (p < 0.001). Furthermore, the subgroup of patients with very early-stage colorectal cancer also had statistically significant differences in the levels of the abovementioned MVs compared to the controls (p < 0.01). Highly differentiated tumors had lower levels of MUC-1-positive MVs (p < 0.02), EMVs (p < 0.002), and EMV/TF combinations (p < 0.001) versus those with tumors with low/intermediate differentiation. CONCLUSIONS Our data demonstrate that the analysis of circulating MV levels in plasma could possibly become a tool for the early diagnosis of colon cancer at a very early stage of the disease.
Collapse
Affiliation(s)
- Anastasios G. Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece; (A.G.K.); (S.P.F.); (H.T.G.); (E.G.P.)
| | - Leonidas Chardalias
- 2nd Department of Surgery, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece; (L.C.); (A.P.); (I.P.)
| | - Christos Damaskos
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 10679 Athens, Greece; (C.D.); (N.G.); (G.T.); (D.D.)
| | - Abraham Pouliakis
- Second Department of Pathology, “Attikon” University Hospital, National and Kapodistrian University of Athens, 10679 Athens, Greece;
| | - Nikolaos Garmpis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 10679 Athens, Greece; (C.D.); (N.G.); (G.T.); (D.D.)
| | - Sotirios P. Fortis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece; (A.G.K.); (S.P.F.); (H.T.G.); (E.G.P.)
| | - Aspasia Papailia
- 2nd Department of Surgery, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece; (L.C.); (A.P.); (I.P.)
| | - Christiana Sideri
- Hematology Laboratory-Blood Bank, Aretaieion Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (C.S.); (T.P.); (M.P.)
| | - Hara T. Georgatzakou
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece; (A.G.K.); (S.P.F.); (H.T.G.); (E.G.P.)
| | - Effie G. Papageorgiou
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece; (A.G.K.); (S.P.F.); (H.T.G.); (E.G.P.)
| | - Theodoros Pittaras
- Hematology Laboratory-Blood Bank, Aretaieion Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (C.S.); (T.P.); (M.P.)
| | - Gerasimos Tsourouflis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 10679 Athens, Greece; (C.D.); (N.G.); (G.T.); (D.D.)
| | - Marianna Politou
- Hematology Laboratory-Blood Bank, Aretaieion Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (C.S.); (T.P.); (M.P.)
| | - Ioannis Papaconstantinou
- 2nd Department of Surgery, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece; (L.C.); (A.P.); (I.P.)
| | - Dimitrios Dimitroulis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 10679 Athens, Greece; (C.D.); (N.G.); (G.T.); (D.D.)
| | - Serena Valsami
- Hematology Laboratory-Blood Bank, Aretaieion Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (C.S.); (T.P.); (M.P.)
| |
Collapse
|
3
|
Tamraz M, Al Ghossaini N, Temraz S. The Role of Wheatgrass in Colorectal Cancer: A Review of the Current Evidence. Int J Mol Sci 2024; 25:5166. [PMID: 38791211 PMCID: PMC11121291 DOI: 10.3390/ijms25105166] [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: 03/23/2024] [Revised: 04/23/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
The etiology of colon cancer is either genetic in nature or results from inflammatory bowel diseases such as ulcerative colitis and Crohn's disease; nevertheless, dietary habits play a crucial role in the disease. Wheatgrass is a dietary supplement that is rich in vitamins, minerals, and antioxidants which contribute to health promotion in cardiovascular diseases, liver disease, blood diseases, diabetes, and inflammatory bowel diseases, as well as in several types of cancers, such as oral squamous cell cancer, cervical cancer, and breast cancer. In colorectal cancer (CRC), the prospect that wheatgrass possesses anti-inflammatory, antioxidant, and anticancer properties, and its use as an adjunctive therapy, have been minimally investigated and evidence is still limited. In this review, we compiled the available evidence pertaining to wheatgrass and its likely impact on CRC, described the pathways of inflammation in which wheatgrass could possibly play a role, and identified future research needs on the subject.
Collapse
Affiliation(s)
- Magie Tamraz
- Department of Nutrition and Public Health, Holy Spirit University of Kaslik, Jounieh P.O. BOX 446, Mount Lebanon, Lebanon;
| | - Najib Al Ghossaini
- Department of Internal Medicine, Ain Wazein Medical Village, Chouf P.O. Box 1503-210/02, Mount Lebanon, Lebanon;
| | - Sally Temraz
- Department of Internal Medicine, Oncology/Hematology Division, American University of Beirut Medical Center, Riad El Solh, Beirut 1107 2020, Lebanon
| |
Collapse
|
4
|
Kayser A, Wolff A, Berlin P, Duehring L, Henze L, Mundkowski R, Bergmann W, Müller-Hilke B, Wagner C, Huehns M, Oehmcke-Hecht S, Maletzki C. Selective but not pan-CDK inhibition abrogates 5-FU-driven tissue factor upregulation in colon cancer. Sci Rep 2024; 14:10582. [PMID: 38719932 PMCID: PMC11078971 DOI: 10.1038/s41598-024-61076-5] [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: 09/04/2023] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
Thromboembolic events are complications in cancer patients and hypercoagulability has been linked to the tissue factor (TF) pathway, making this an attractive target. Here, we investigated the effects of chemotherapeutics and CDK inhibitors (CDKI) abemaciclib/palbociclib (CDK4/6), THZ-1 (CDK7/12/13), and dinaciclib (CDK1/2/5/9) alone and in combination regimens on TF abundance and coagulation. The human colorectal cancer (CRC) cell line HROC173 was treated with 5-FU or gemcitabine to stimulate TF expression. TF+ cells were sorted, recultured, and re-analyzed. The effect of treatment alone or in combination was assessed by functional assays. Low-dose chemotherapy induced a hypercoagulable state and significantly upregulated TF, even after reculture without treatment. Cells exhibited characteristics of epithelial-mesenchymal transition, including high expression of vimentin and mucin. Dinaciclib and THZ-1 also upregulated TF, while abemaciclib and palbociclib downregulated it. Similar results were observed in coagulation assays. The same anticoagulant activity of abemaciclib was seen after incubation with peripheral immune cells from healthy donors and CRC patients. Abemaciclib reversed 5-FU-induced TF upregulation and prolonged clotting times in second-line treatment. Effects were independent of cytotoxicity, senescence, and p27kip1 induction. TF-antibody blocking experiments confirmed the importance of TF in plasma coagulation, with Factor XII playing a minor role. Short-term abemaciclib counteracts 5-FU-induced hypercoagulation and eventually even prevents thromboembolic events.
Collapse
Affiliation(s)
- Annika Kayser
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Annabell Wolff
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, 18057, Rostock, Germany
| | - Peggy Berlin
- Department of Medicine II, Division of Gastroenterology, Rostock University Medical Center, 18057, Rostock, Germany
| | - Lara Duehring
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, 18057, Rostock, Germany
| | - Larissa Henze
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
- Department of Internal Medicine II, Asklepios Hospital Harz, Goslar, Germany
| | - Ralf Mundkowski
- Center of Pharmacology and Toxicology, Institute of Clinical Pharmacology, Rostock University Medical Center, 18057, Rostock, Germany
| | - Wendy Bergmann
- Laboratory for Clinical Immunology, Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, 18057, Rostock, Germany
| | - Brigitte Müller-Hilke
- Laboratory for Clinical Immunology, Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, 18057, Rostock, Germany
| | - Charlotte Wagner
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Maja Huehns
- Institute of Pathology, Rostock University Medical Center, 18057, Rostock, Germany
| | - Sonja Oehmcke-Hecht
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, 18057, Rostock, Germany.
| | - Claudia Maletzki
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.
| |
Collapse
|
5
|
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
|
6
|
Veilleux V, Pichaud N, Boudreau LH, Robichaud GA. Mitochondria Transfer by Platelet-Derived Microparticles Regulates Breast Cancer Bioenergetic States and Malignant Features. Mol Cancer Res 2024; 22:268-281. [PMID: 38085263 DOI: 10.1158/1541-7786.mcr-23-0329] [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: 05/05/2023] [Revised: 07/25/2023] [Accepted: 12/07/2023] [Indexed: 03/02/2024]
Abstract
An increasing number of studies show that platelets as well as platelet-derived microparticles (PMP) play significant roles in cancer malignancy and disease progression. Particularly, PMPs have the capacity to interact and internalize within target cells resulting in the transfer of their bioactive cargo, which can modulate the signaling and activation processes of recipient cells. We recently identified a new subpopulation of these vesicles (termed mitoMPs), which contain functional mitochondria. Given the predominant role of mitochondria in cancer cell metabolism and disease progression, we set out to investigate the impact of mitoMPs on breast cancer metabolic reprograming and phenotypic processes leading to malignancy. Interestingly, we observed that recipient cell permeability to PMP internalization varied among the breast cancer cell types evaluated in our study. Specifically, cells permissive to mitoMPs acquire mitochondrial-dependent functions, which stimulate increased cellular oxygen consumption rates and intracellular ATP levels. In addition, cancer cells co-incubated with PMPs display enhanced malignant features in terms of migration and invasion. Most importantly, the cancer aggressive processes and notable metabolic plasticity induced by PMPs were highly dependent on the functional status of the mitoMP-packaged mitochondria. These findings characterize a new mechanism by which breast cancer cells acquire foreign mitochondria resulting in the gain of metabolic processes and malignant features. A better understanding of these mechanisms may provide therapeutic opportunities through PMP blockade to deprive cancer cells from resources vital in disease progression. IMPLICATIONS We show that the transfer of foreign mitochondria by microparticles modulates recipient cancer cell metabolic plasticity, leading to greater malignant processes.
Collapse
Affiliation(s)
- Vanessa Veilleux
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | - Nicolas Pichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
| | - Luc H Boudreau
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
| | - Gilles A Robichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| |
Collapse
|
7
|
Krishnan ST, Winkler D, Creek D, Anderson D, Kirana C, Maddern GJ, Fenix K, Hauben E, Rudd D, Voelcker NH. Staging of colorectal cancer using lipid biomarkers and machine learning. Metabolomics 2023; 19:84. [PMID: 37731020 PMCID: PMC10511619 DOI: 10.1007/s11306-023-02049-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/07/2023] [Indexed: 09/22/2023]
Abstract
INTRODUCTION Colorectal cancer (CRC) is the third most commonly diagnosed cancer worldwide. Alteration in lipid metabolism and chemokine expression are considered hallmark characteristics of malignant progression and metastasis of CRC. Validated diagnostic and prognostic biomarkers are urgently needed to define molecular heterogeneous CRC clinical stages and subtypes, as liver dominant metastasis has poor survival outcomes. OBJECTIVES The aim of this study was to integrate lipid changes, concentrations of chemokines, such as platelet factor 4 and interleukin 8, and gene marker status measured in plasma samples, with clinical features from patients at different CRC stages or who had progressed to stage-IV colorectal liver metastasis (CLM). METHODS High-resolution liquid chromatography-mass spectrometry (HR-LC-MS) was used to determine the levels of candidate lipid biomarkers in each CRC patient's preoperative plasma samples and combined with chemokine, gene and clinical data. Machine learning models were then trained using known clinical outcomes to select biomarker combinations that best classify CRC stage and group. RESULTS Bayesian neural net and multilinear regression-machine learning identified candidate biomarkers that classify CRC (stages I-III), CLM patients and control subjects (cancer-free or patients with polyps/diverticulitis), showing that integrating specific lipid signatures and chemokines (platelet factor-4 and interluken-8; IL-8) can improve prognostic accuracy. Gene marker status could contribute to disease prediction, but requires ubiquitous testing in clinical cohorts. CONCLUSION Our findings demonstrate that correlating multiple disease related features with lipid changes could improve CRC prognosis. The identified signatures could be used as reference biomarkers to predict CRC prognosis and classify stages, and monitor therapeutic intervention.
Collapse
Affiliation(s)
- Sanduru Thamarai Krishnan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- Department of Chemistry, University of Reading, Whiteknights, Reading, RG6 6DX, UK
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - David Winkler
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, 3086, Australia
- School of Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2QL, UK
| | - Darren Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- Monash Proteomics and Metabolomics Facility, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Dovile Anderson
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- Monash Proteomics and Metabolomics Facility, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Chandra Kirana
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA, 5011, Australia
| | - Guy J Maddern
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA, 5011, Australia
| | - Kevin Fenix
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA, 5011, Australia
| | - Ehud Hauben
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA, 5011, Australia
| | - David Rudd
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
| | - Nicolas Hans Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, VIC, 3168, Australia.
| |
Collapse
|
8
|
Zhang Y, Zhang X, Xu X, Guo X, Xu S, Ma S, Chen J, Qi X. Association of C-type lectin-like receptor 2 and galectin-1 with portal vein system thrombosis in HBV-related liver cirrhosis. Front Med (Lausanne) 2023; 10:1228636. [PMID: 37720512 PMCID: PMC10501130 DOI: 10.3389/fmed.2023.1228636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Background and aims Hepatitis B virus (HBV) infection is the most common cause of liver cirrhosis. Portal venous system thrombosis (PVST) is a major complication of liver cirrhosis. Recently, it has been shown that C-type lectin-like receptor 2 (CLEC-2) and galectin-1 participate in the activation and aggregation of platelets, thereby promoting the development of thrombosis. This cross-sectional study aims to evaluate the association of serum CLEC-2 and galectin-1 levels with PVST in patients with HBV-related liver cirrhosis. Methods Overall, 65 patients with HBV-related liver cirrhosis were included, of whom 23 had PVST and 42 did not have. Serum CLEC-2 and galectin-1 levels were measured using enzyme-linked immunosorbent assay kits. PVST was assessed by contrast-enhanced computed tomography and/or magnetic resonance imaging scans. Subgroup analyses were conducted according to the degree and location of PVST. Results Patients with PVST had significantly higher serum CLEC-2 (p = 0.006) and galectin-1 (p = 0.009) levels than those without. Patients with partial/complete PVST or fibrotic cord (p = 0.007; p = 0.002), but not those with mural PVST (p = 0.199; p = 0.797), had significantly higher serum CLEC-2 and galectin-1 levels than those without PVST. Patients with superior mesenteric vein thrombosis had significantly higher serum CLEC-2 (p = 0.013) and galectin-1 (p = 0.025) levels than those without PVST. Patients with main portal vein thrombosis had higher serum CLEC-2 (p = 0.020) and galectin-1 (p = 0.066) levels than those without PVST, but the difference in serum galectin-1 level was not significant between them. Conclusion Serum CLEC-2 and galectin-1 levels may be associated with the presence of PVST in HBV-related cirrhotic patients, but this association should be dependent upon the degree of PVST.
Collapse
Affiliation(s)
- Yiyan Zhang
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China
- Postgraduate College, China Medical University, Shenyang, China
| | - Xintong Zhang
- Chinese People’s Liberation Army General Hospital, Chinese People’s Liberation Army Medical School, Beijing, China
| | - Xiangbo Xu
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China
- Postgraduate College, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaozhong Guo
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China
| | - Shixue Xu
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China
- Postgraduate College, China Medical University, Shenyang, China
| | - Shaoze Ma
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China
- Postgraduate College, Dalian Medical University, Dalian, China
| | - Jihong Chen
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China
- Postgraduate College, Dalian Medical University, Dalian, China
| | - Xingshun Qi
- Liver Cirrhosis Study Group, Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
9
|
Tong H, Li K, Zhou M, Wu R, Yang H, Peng Z, Zhao Q, Luo KQ. Coculture of cancer cells with platelets increases their survival and metastasis by activating the TGFβ/Smad/PAI-1 and PI3K/AKT pathways. Int J Biol Sci 2023; 19:4259-4277. [PMID: 37705745 PMCID: PMC10496510 DOI: 10.7150/ijbs.85986] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/18/2023] [Indexed: 09/15/2023] Open
Abstract
When cancer cells enter the bloodstream, they can interact with platelets to acquire stronger survival and metastatic abilities. To elucidate the underlying mechanisms, we cocultured metastatic melanoma and triple-negative breast cancer cells with species-homologous platelets. We found that cocultured cancer cells displayed higher viabilities in circulation, stronger capacities for cell migration, invasion, and colony formation in vitro, and more tumorigenesis and metastasis in mice. RNA sequencing analysis revealed that the level of serpin family E member 1 (SERPINE1) was significantly upregulated in cocultured cancer cells. Knockdown of SERPINE1 reversed the coculture-elevated survival and metastatic phenotypes of cancer cells. Mechanistic studies indicated that coculture with platelets activated the TGFβ/Smad pathway to induce SERPINE1 expression in cancer cells, which encodes plasminogen activator inhibitor 1 (PAI-1). PAI-1 then activated PI3K to increase the phosphorylation of AKTThr308 and Bad to elevate Bcl-2, which enhanced cell survival in circulation. Moreover, higher levels of PAI-1 were detected in metastatic tumors from melanoma and triple-negative breast cancer patients than in normal tissues, and high levels of PAI-1 were associated with a shorter overall survival time and worse disease progression in breast cancer. PAI-1 may act as a potential biomarker for detecting and treating metastatic tumor cells.
Collapse
Affiliation(s)
- Haibo Tong
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Koukou Li
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Muya Zhou
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Renfei Wu
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Hongmei Yang
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Zheng Peng
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Qi Zhao
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau
| |
Collapse
|
10
|
Bekendam RH, Ravid K. Mechanisms of platelet activation in cancer-associated thrombosis: a focus on myeloproliferative neoplasms. Front Cell Dev Biol 2023; 11:1207395. [PMID: 37457287 PMCID: PMC10342211 DOI: 10.3389/fcell.2023.1207395] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023] Open
Abstract
Platelets are anucleate blood cells that play key roles in thrombosis and hemostasis. Platelets are also effector cells in malignancy and are known to home into the microenvironment of cancers. As such, these cells provide central links between the hemostatic system, inflammation and cancer progression. Activation of platelets by cancers has been postulated to contribute to metastasis and progression of local tumor invasion. Similarly, cancer-activated platelets can increase the risk of development of both arterial and venous thrombosis; a major contributor to cancer-associated morbidity. Platelet granules secretion within the tumor environment or the plasma provide a rich source of potential biomarkers for prediction of thrombotic risk or tumor progression. In the case of myeloproliferative neoplasms (MPNs), which are characterized by clonal expansion of myeloid precursors and abnormal function and number of erythrocytes, leukocytes and platelets, patients suffer from thrombotic and hemorrhagic complications. The mechanisms driving this are likely multifactorial but remain poorly understood. Several mouse models developed to recapitulate MPN phenotype with one of the driving mutations, in JAK2 (JAK2V617F) or in calreticulin (CALR) or myeloproliferative leukemia virus oncogene receptor (MPL), have been studied for their thrombotic phenotype. Variability and discrepancies were identified within different disease models of MPN, emphasizing the complexity of increased risk of clotting and bleeding in these pathologies. Here, we review recent literature on the role of platelets in cancer-associated arterial and venous thrombosis and use MPN as case study to illustrate recent advances in experimental models of thrombosis in a malignant phenotype. We address major mechanisms of tumor-platelet communication leading to thrombosis and focus on the role of altered platelets in promoting thrombosis in MPN experimental models and patients with MPN. Recent identification of platelet-derived biomarkers of MPN-associated thrombosis is also reviewed, with potential therapeutic implications.
Collapse
Affiliation(s)
- Roelof H. Bekendam
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Katya Ravid
- Department of Medicine and Biochemistry, Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
11
|
Ferlizza E, Romaniello D, Borrelli F, Pagano F, Girone C, Gelfo V, Kuhre RS, Morselli A, Mazzeschi M, Sgarzi M, Filippini DM, D'Uva G, Lauriola M. Extracellular Vesicles and Epidermal Growth Factor Receptor Activation: Interplay of Drivers in Cancer Progression. Cancers (Basel) 2023; 15:cancers15112970. [PMID: 37296932 DOI: 10.3390/cancers15112970] [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: 03/24/2023] [Revised: 05/12/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Extracellular vesicles (EVs) are of great interest to study the cellular mechanisms of cancer development and to diagnose and monitor cancer progression. EVs are a highly heterogeneous population of cell derived particles, which include microvesicles (MVs) and exosomes (EXOs). EVs deliver intercellular messages transferring proteins, lipids, nucleic acids, and metabolites with implications for tumour progression, invasiveness, and metastasis. Epidermal Growth Factor Receptor (EGFR) is a major driver of cancer. Tumour cells with activated EGFR could produce EVs disseminating EGFR itself or its ligands. This review provides an overview of EVs (mainly EXOs and MVs) and their cargo, with a subsequent focus on their production and effects related to EGFR activation. In particular, in vitro studies performed in EGFR-dependent solid tumours and/or cell cultures will be explored, thus shedding light on the interplay between EGFR and EVs production in promoting cancer progression, metastases, and resistance to therapies. Finally, an overview of liquid biopsy approaches involving EGFR and EVs in the blood/plasma of EGFR-dependent tumour patients will also be discussed to evaluate their possible application as candidate biomarkers.
Collapse
Affiliation(s)
- Enea Ferlizza
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Donatella Romaniello
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Francesco Borrelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Federica Pagano
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Cinzia Girone
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Valerio Gelfo
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Rikke Sofie Kuhre
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Alessandra Morselli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Martina Mazzeschi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Michela Sgarzi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Daria Maria Filippini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Gabriele D'Uva
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Mattia Lauriola
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
12
|
Perez GI, Bernard MP, Vocelle D, Zarea AA, Saleh NA, Gagea MA, Schneider D, Bauzon M, Hermiston T, Kanada M. Phosphatidylserine-Exposing Annexin A1-Positive Extracellular Vesicles: Potential Cancer Biomarkers. Vaccines (Basel) 2023; 11:639. [PMID: 36992223 PMCID: PMC10059271 DOI: 10.3390/vaccines11030639] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 02/11/2023] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
Under physiological conditions, phosphatidylserine (PS) predominantly localizes to the cytosolic leaflet of the plasma membrane of cells. During apoptosis, PS is exposed on the cell surface and serves as an "eat-me" signal for macrophages to prevent releasing self-immunogenic cellular components from dying cells which could potentially lead to autoimmunity. However, increasing evidence indicates that viable cells can also expose PS on their surface. Interestingly, tumor cell-derived extracellular vesicles (EVs) externalize PS. Recent studies have proposed PS-exposing EVs as a potential biomarker for the early detection of cancer and other diseases. However, there are confounding results regarding subtypes of PS-positive EVs, and knowledge of PS exposure on the EV surface requires further elucidation. In this study, we enriched small EVs (sEVs) and medium/large EVs (m/lEVs) from conditioned media of breast cancer cells (MDA-MB-231, MDA-MB-468) and non-cancerous cells (keratinocytes, fibroblasts). Since several PS-binding molecules are available to date, we compared recombinant proteins of annexin A5 and the carboxylated glutamic acid domain of Protein S (GlaS), also specific for PS, to detect PS-exposing EVs. Firstly, PS externalization in each EV fraction was analyzed using a bead-based EV assay, which combines EV capture using microbeads and analysis of PS-exposing EVs by flow cytometry. The bulk EV assay showed higher PS externalization in m/lEVs derived from MDA-MB-468 cells but not from MDA-MB-231 cells, while higher binding of GlaS was also observed in m/lEVs from fibroblasts. Second, using single EV flow cytometry, PS externalization was also analyzed on individual sEVs and m/lEVs. Significantly higher PS externalization was detected in m/lEVs (annexin A1+) derived from cancer cells compared to m/lEVs (annexin A1+) from non-cancerous cells. These results emphasize the significance of PS-exposing m/lEVs (annexin A1+) as an undervalued EV subtype for early cancer detection and provide a better understanding of PS externalization in disease-associated EV subtypes.
Collapse
Affiliation(s)
- Gloria I. Perez
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA (M.P.B.)
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Matthew P. Bernard
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA (M.P.B.)
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Daniel Vocelle
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Ahmed A. Zarea
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA (M.P.B.)
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA
- College of Natural Science, Michigan State University, East Lansing, MI 48824, USA
| | - Najla A. Saleh
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA (M.P.B.)
| | - Matthew A. Gagea
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA (M.P.B.)
- Lyman Briggs College, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | | - Masamitsu Kanada
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA (M.P.B.)
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
13
|
Setua S, Thangaraju K, Dzieciatkowska M, Wilkerson RB, Nemkov T, Lamb DR, Tagaya Y, Boyer T, Rowden T, Doctor A, D'Alessandro A, Buehler PW. Coagulation potential and the integrated omics of extracellular vesicles from COVID-19 positive patient plasma. Sci Rep 2022; 12:22191. [PMID: 36564503 PMCID: PMC9780627 DOI: 10.1038/s41598-022-26473-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Extracellular vesicles (EVs) participate in cell-to-cell communication and contribute toward homeostasis under physiological conditions. But EVs can also contribute toward a wide array of pathophysiology like cancer, sepsis, sickle cell disease, and thrombotic disorders. COVID-19 infected patients are at an increased risk of aberrant coagulation, consistent with elevated circulating levels of ultra-high molecular weight VWF multimers, D-dimer and procoagulant EVs. The role of EVs in COVID-19 related hemostasis may depend on cells of origin, vesicular cargo and size, however this is not well defined. We hypothesized that the procoagulant potential of EV isolates from COVID-19 (+) patient plasmas could be defined by thrombin generation assays. Here we isolated small EVs (SEVs) and large EVs (LEVs) from hospitalized COVID-19 (+) patient (n = 21) and healthy donor (n = 20) plasmas. EVs were characterized by flow cytometry, Transmission electron microscopy, nanoparticle tracking analysis, plasma thrombin generation and a multi-omics approach to define coagulation potential. These data were consistent with differences in EV metabolite, lipid, and protein content when compared to healthy donor plasma isolated SEVs and LEVs. Taken together, the effect of EVs on plasma procoagulant potential as defined by thrombin generation and supported by multi-omics is enhanced in COVID-19. Further, we observe that this effect is driven both by EV size and phosphatidyl serine.
Collapse
Affiliation(s)
- Saini Setua
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kiruphagaran Thangaraju
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver-Anschutz Medical Campus, 12801 East 17th Ave., Aurora, CO, 80045, USA
| | - Rebecca B Wilkerson
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver-Anschutz Medical Campus, 12801 East 17th Ave., Aurora, CO, 80045, USA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver-Anschutz Medical Campus, 12801 East 17th Ave., Aurora, CO, 80045, USA
| | - Derek R Lamb
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yutaka Tagaya
- Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tori Boyer
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tobi Rowden
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Allan Doctor
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver-Anschutz Medical Campus, 12801 East 17th Ave., Aurora, CO, 80045, USA.
| | - Paul W Buehler
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
14
|
Muñoz-Hernández R, Rojas Á, Gato S, Gallego J, Gil-Gómez A, Castro MJ, Ampuero J, Romero-Gómez M. Extracellular Vesicles as Biomarkers in Liver Disease. Int J Mol Sci 2022; 23:ijms232416217. [PMID: 36555854 PMCID: PMC9786586 DOI: 10.3390/ijms232416217] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane-derived vesicles released by a variety of cell types, including hepatocytes, hepatic stellate cells, and immune cells in normal and pathological conditions. Depending on their biogenesis, there is a complex repertoire of EVs that differ in size and origin. EVs can carry lipids, proteins, coding and non-coding RNAs, and mitochondrial DNA causing alterations to the recipient cells, functioning as intercellular mediators of cell-cell communication (auto-, para-, juxta-, or even endocrine). Nevertheless, many questions remain unanswered in relation to the function of EVs under physiological and pathological conditions. The development and optimization of methods for EV isolation are crucial for characterizing their biological functions, as well as their potential as a treatment option in the clinic. In this manuscript, we will comprehensively review the results from different studies that investigated the role of hepatic EVs during liver diseases, including non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, alcoholic liver disease, fibrosis, and hepatocellular carcinoma. In general, the identification of patients with early-stage liver disease leads to better therapeutic interventions and optimal management. Although more light needs to be shed on the mechanisms of EVs, their use for early diagnosis, follow-up, and prognosis has come into the focus of research as a high-potential source of 'liquid biopsies', since they can be found in almost all biological fluids. The use of EVs as new targets or nanovectors in drug delivery systems for liver disease therapy is also summarized.
Collapse
Affiliation(s)
- Rocío Muñoz-Hernández
- SeLiver Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain
- CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (R.M.-H.); (M.R.-G.)
| | - Ángela Rojas
- SeLiver Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain
- CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sheila Gato
- SeLiver Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain
- CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Gallego
- SeLiver Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain
| | - Antonio Gil-Gómez
- SeLiver Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain
- CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María José Castro
- Servicio de Citometría y Separación Celular, Instituto de Biomedicina de Sevilla Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain
| | - Javier Ampuero
- SeLiver Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain
- CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
- UCM Digestive Diseases, Virgen del Rocío University Hospital, 41013 Seville, Spain
| | - Manuel Romero-Gómez
- SeLiver Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain
- CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
- UCM Digestive Diseases, Virgen del Rocío University Hospital, 41013 Seville, Spain
- Correspondence: (R.M.-H.); (M.R.-G.)
| |
Collapse
|
15
|
Sun H, Du Y, Kumar R, Buchkovich N, He P. Increased circulating microparticles contribute to severe infection and adverse outcomes of COVID-19 in patients with diabetes. Am J Physiol Heart Circ Physiol 2022; 323:H1176-H1193. [PMID: 36269646 PMCID: PMC9678425 DOI: 10.1152/ajpheart.00409.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Patients with diabetes infected with COVID-19 have greater mortality than those without comorbidities, but the underlying mechanisms remain unknown. This study aims to identify the mechanistic interactions between diabetes and severe COVID-19. Microparticles (MPs), the cell membrane-derived vesicles released on cell activation, are largely increased in patients with diabetes. To date, many mechanisms have been postulated for increased severity of COVID-19 in patients with underlying conditions, but the contributions of excessive MPs in patients with diabetes have been overlooked. This study characterizes plasma MPs from normal human subjects and patients with type 2 diabetes in terms of amount, cell origins, surface adhesive properties, ACE2 expression, spike protein binding capacity, and their roles in SARS-CoV-2 infection. Results showed that over 90% of plasma MPs express ACE2 that binds the spike protein of SARS-CoV-2. MPs in patients with diabetes increase 13-fold in quantity and 11-fold in adhesiveness when compared with normal subjects. Perfusion of human plasma with pseudo-typed SARS-CoV-2 virus or spike protein-bound MPs into human endothelial cell-formed microvessels-on-a chip demonstrated that MPs from patients with diabetes, not normal subjects, interact with endothelium and carry SARS-CoV-2 into cells through endocytosis, providing additional virus entry pathways and enhanced infection. Results also showed a large percentage of platelet-derived tissue factor-bearing MPs in diabetic plasma, which could contribute to thrombotic complications with SARS-CoV-2 infection. This study reveals a dual role of diabetic MPs in promoting SARS-CoV-2 entry and propagating vascular inflammation. These findings provide novel mechanistic insight into the high prevalence of COVID-19 in patients with diabetes and their propensity to develop severe vascular complications.NEW & NOTEWORTHY This study provides the first evidence that over 90% of human plasma microparticles express ACE2 that binds SARS-CoV-2 S protein with high affinity. Thus, the highly elevated adhesive circulating microparticles identified in patients with diabetes not only have greater SARS-CoV-2 binding capacity but also enable additional viral entry through virus-bound microparticle-endothelium interactions and enhanced infection. These findings reveal a novel mechanistic insight into the adverse outcomes of COVID-19 in patients with diabetes.
Collapse
Affiliation(s)
- Haoyu Sun
- 1Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Yong Du
- 1Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Rinki Kumar
- 2Department of Microbiology and Immunology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Nicholas Buchkovich
- 2Department of Microbiology and Immunology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Pingnian He
- 1Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| |
Collapse
|
16
|
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: 18] [Impact Index Per Article: 9.0] [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
|
17
|
Tang X, Li D, Gu Y, Zhao Y, Li A, Qi F, Liu J. Natural cell based biomimetic cellular transformers for targeted therapy of digestive system cancer. Theranostics 2022; 12:7080-7107. [PMID: 36276645 PMCID: PMC9576611 DOI: 10.7150/thno.75937] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/29/2022] [Indexed: 11/22/2022] Open
Abstract
Digestive system cancer is the most common cause of cancer death in the world. Although cancer treatment options are increasingly diversified, the mortality rate of malignant cancer of the digestive system remains high. Therefore, it is necessary to explore effective cancer treatment methods. Recently, biomimetic nanoparticle delivery systems based on natural cells that organically integrate the low immunogenicity, high biocompatibility, cancer targeting, and controllable, versatile functionality of smart nanocarrier design with natural cells have been expected to break through the bottleneck of tumor targeted therapy. In this review, we focus on the dynamic changes and complex cellular communications that occur in vivo in natural cells based vehicles. Recent studies on the development of advanced targeted drug delivery systems using the dynamic behaviors such as specific surface protein affinity, morphological changes, and phenotypic polarization of natural cells are summarized. In addition to drug delivery mediated by dynamic behavior, functional "delivery" based on the natural cell themselves is also involved. Aiming to make the best use of the functions of cells, providing clues for the development of advanced drug delivery platforms.
Collapse
Affiliation(s)
- Xiaomeng Tang
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Dan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yongwei Gu
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yunan Zhao
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Aixue Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Fu Qi
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pharmacy, Shanghai Proton and Heavy Ion Center, Shanghai 201315, China
| |
Collapse
|
18
|
Chen Y, Liu J, Su Y, Zhao H, Zhao Y, Wen M, Lu S, Cao X, Zhang W, Liu L, Wu J. Annexin V - and tissue factor + microparticles as biomarkers for predicting deep vein thrombosis in patients after joint arthroplasty. Clin Chim Acta 2022; 536:169-179. [PMID: 36191610 DOI: 10.1016/j.cca.2022.09.011] [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: 02/21/2021] [Revised: 08/29/2022] [Accepted: 09/08/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Venous thromboembolism (VTE) is a common and severe complication of joint arthroplasty. Microparticles (MPs) containing phosphatidylserine (PS) and tissue factor (TF) can trigger coagulation in VTE. This study aims to measure and compare MP levels in joint arthroplasty patients with and without VTE. METHODS This prospective cohort study enrolled 181 patients who underwent joint arthroplasty. Ultrasound examination was used to diagnose VTE on preoperative day 0 and postoperative day 6. MPs were analysed using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and flow cytometry. The levels of platelet-derived microparticles (PMPs), endothelial cell-derived microparticles (EMPs), granulocyte-derived microparticles (GMPs), red cell-derived microparticles (RMPs), monocyte-derived microparticles (MMPs), Annexin V+ MPs (AV+ MPs), and tissue factor+ MPs (TF+ MPs) derived from five kinds of MPs were measured on day 0 (before surgery), 1, 2, 3, 4, 5, and 6 after surgery. RESULTS The levels of AV-TF+ EMPs and AV-TF+ MMPs were significantly increased in patients with VTE on postoperative day 5 compared to those without VTE (P=0.031 and P=0.031, respectively). CONCLUSION AV-TF+ MPs may indicate the development of VTE and serve as predictive markers in joint arthroplasty patients.
Collapse
Affiliation(s)
- Yuying Chen
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China
| | - Jian Liu
- Adult reconstruction department, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Yu Su
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Huiru Zhao
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Yujing Zhao
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Meng Wen
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Shan Lu
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Xiangyu Cao
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China
| | - Wenjie Zhang
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China
| | - Lei Liu
- Department of Clinical Laboratory, Liyuan Hospital of Tongji Medical College of Huazhong University of Science and Technology, Hubei, P.R.China
| | - Jun Wu
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China; Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China.
| |
Collapse
|
19
|
Li JC, Zou XM, Yang SF, Jin JQ, Zhu L, Li CJ, Yang H, Zhang AG, Zhao TQ, Chen CY. Neutrophil extracellular traps participate in the development of cancer-associated thrombosis in patients with gastric cancer. World J Gastroenterol 2022; 28:3132-3149. [PMID: 36051331 PMCID: PMC9331535 DOI: 10.3748/wjg.v28.i26.3132] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/20/2022] [Accepted: 03/16/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The development of venous thromboembolism (VTE) is associated with high mortality among gastric cancer (GC) patients. Neutrophil extracellular traps (NETs) have been reported to correlate with the prothrombotic state in some diseases, but are rarely reported in GC patients.
AIM To investigate the effect of NETs on the development of cancer-associated thrombosis in GC patients.
METHODS The levels of NETs in blood and tissue samples of patients were analyzed by ELISA, flow cytometry, and immunofluorescence staining. NET generation and hypercoagulation of platelets and endothelial cells (ECs) in vitro were observed by immunofluorescence staining. NET procoagulant activity (PCA) was determined by fibrin formation and thrombin–antithrombin complex (TAT) assays. Thrombosis in vivo was measured in a murine model induced by flow stenosis in the inferior vena cava (IVC).
RESULTS NETs were likely to form in blood and tissue samples of GC patients compared with healthy individuals. In vitro studies showed that GC cells and their conditioned medium, but not gastric mucosal epithelial cells, stimulated NET release from neutrophils. In addition, NETs induced a hypercoagulable state of platelets by upregulating the expression of phosphatidylserine and P-selectin on the cells. Furthermore, NETs stimulated the adhesion of normal platelets on glass surfaces. Similarly, NETs triggered the conversion of ECs to hypercoagulable phenotypes by downregulating the expression of their intercellular tight junctions but upregulating that of tissue factor. Treatment of normal platelets or ECs with NETs augmented the level of plasma fibrin formation and the TAT complex. In the models of IVC stenosis, tumor-bearing mice showed a stronger ability to form thrombi, and NETs abundantly accumulated in the thrombi of tumor-bearing mice compared with control mice. Notably, the combination of deoxyribonuclease I, activated protein C, and sivelestat markedly abolished the PCA of NETs.
CONCLUSION GC-induced NETs strongly increased the risk of VTE development both in vitro and in vivo. NETs are potential therapeutic targets in the prevention and treatment of VTE in GC patients.
Collapse
Affiliation(s)
- Jia-Cheng Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Xiao-Ming Zou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Shi-Feng Yang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Jia-Qi Jin
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Lei Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Chang-Jian Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Hao Yang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - An-Ge Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Tian-Qi Zhao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Chong-Yan Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
20
|
Chronic Immune Platelet Activation Is Followed by Platelet Refractoriness and Impaired Contractility. Int J Mol Sci 2022; 23:ijms23137336. [PMID: 35806341 PMCID: PMC9266422 DOI: 10.3390/ijms23137336] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Autoimmune diseases, including systemic lupus erythematosus (SLE), have a high risk of thrombotic and hemorrhagic complications associated with altered platelet functionality. We studied platelets from the blood of SLE patients and their reactivity. The surface expression of phosphatidylserine, P-selectin, and active integrin αIIbβ3 were measured using flow cytometry before and after platelet stimulation. Soluble P-selectin was measured in plasma. The kinetics of platelet-driven clot contraction was studied, as well as scanning and transmission electron microscopy of unstimulated platelets. Elevated levels of membrane-associated phosphatidylserine and platelet-attached and soluble P-selectin correlated directly with the titers of IgG, anti-dsDNA-antibodies, and circulating immune complexes. Morphologically, platelets in SLE lost their resting discoid shape, formed membrane protrusions and aggregates, and had a rough plasma membrane. The signs of platelet activation were associated paradoxically with reduced reactivity to a physiological stimulus and impaired contractility that revealed platelet exhaustion and refractoriness. Platelet activation has multiple pro-coagulant effects, and the inability to fully contract (retract) blood clots can be either a hemorrhagic or pro-thrombotic mechanism related to altered clot permeability, sensitivity of clots to fibrinolysis, obstructiveness, and embologenicity. Therefore, chronic immune platelet activation followed by secondary platelet dysfunction comprise an understudied pathogenic mechanism that supports hemostatic disorders in autoimmune diseases, such as SLE.
Collapse
|
21
|
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: 6.0] [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
|
22
|
Igami K, Uchiumi T, Shiota M, Ueda S, Tsukahara S, Akimoto M, Eto M, Kang D. Extracellular vesicles expressing CEACAM proteins in the urine of bladder cancer patients. Cancer Sci 2022; 113:3120-3133. [PMID: 35611462 PMCID: PMC9459299 DOI: 10.1111/cas.15438] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/14/2022] [Accepted: 05/18/2022] [Indexed: 12/26/2022] Open
Abstract
Early detection and long‐term monitoring are important for urothelial carcinoma of the bladder (UCB). Urine cytology and existing markers have insufficient diagnostic performance. Here, we examined medium‐sized extracellular vesicles (EVs) in urine to identify specific markers for UCB and evaluated their usefulness as diagnostic material. To identify specific markers in urinary EVs derived from UCB, we undertook shotgun proteomics using urine from four UCB patients and four healthy subjects. Next, 29 healthy specimens, 18 noncancer specimens, and 33 UCB specimens, all from men, were analyzed for urinary EVs by flow cytometry to evaluate the diagnostic performance of UCB‐specific EVs. Nanoparticle‐tracking analysis indicated that the size of EVs extracted from urine was mostly <400 nm. By shotgun proteomics, we detected several proteins characteristic of UCB and found that carcinoembryonic antigen‐related adhesion molecule (CEACAM) proteins were increased in patients. Flow cytometric analysis revealed that the degree of expression of CEACAM1, CEACAM5, and CEACAM6 proteins on the surface of EVs varied among patients. Extracellular vesicles expressing CEACAM proteins also expressed mucin 1, suggesting that they were derived from tumorigenic uroepithelial cells. The number of EVs expressing CEACAM1, 5, and 6 proteins was significantly increased in UCB (mean ± SD, 8.6 ± 13%) compared to non‐UCB (0.69 ± 0.46) and healthy (0.46 ± 0.34) by flow cytometry. The results of receiver operating characteristic (ROC) analysis showed a good score of area under the ROC curve of 0.907. We identified EVs that specifically express CEACAM proteins in urine and have potential for diagnostic applications. These EVs are potential targets in a new liquid biopsy test for UCB patients.
Collapse
Affiliation(s)
- Ko Igami
- Business Management Division, Clinical Laboratory Business Segment, LSI Medience Corporation, Tokyo, Japan.,Kyushu Pro Search Limited Liability Partnership, Fukuoka, Japan.,Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan.,Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Saori Ueda
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
| | - Shigehiro Tsukahara
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan.,Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaru Akimoto
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Bourguignon A, Tasneem S, Hayward CP. Screening and diagnosis of inherited platelet disorders. Crit Rev Clin Lab Sci 2022; 59:405-444. [PMID: 35341454 DOI: 10.1080/10408363.2022.2049199] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inherited platelet disorders are important conditions that often manifest with bleeding. These disorders have heterogeneous underlying pathologies. Some are syndromic disorders with non-blood phenotypic features, and others are associated with an increased predisposition to developing myelodysplasia and leukemia. Platelet disorders can present with thrombocytopenia, defects in platelet function, or both. As the underlying pathogenesis of inherited thrombocytopenias and platelet function disorders are quite diverse, their evaluation requires a thorough clinical assessment and specialized diagnostic tests, that often challenge diagnostic laboratories. At present, many of the commonly encountered, non-syndromic platelet disorders do not have a defined molecular cause. Nonetheless, significant progress has been made over the past few decades to improve the diagnostic evaluation of inherited platelet disorders, from the assessment of the bleeding history to improved standardization of light transmission aggregometry, which remains a "gold standard" test of platelet function. Some platelet disorder test findings are highly predictive of a bleeding disorder and some show association to symptoms of prolonged bleeding, surgical bleeding, and wound healing problems. Multiple assays can be required to diagnose common and rare platelet disorders, each requiring control of preanalytical, analytical, and post-analytical variables. The laboratory investigations of platelet disorders include evaluations of platelet counts, size, and morphology by light microscopy; assessments for aggregation defects; tests for dense granule deficiency; analyses of granule constituents and their release; platelet protein analysis by immunofluorescent staining or flow cytometry; tests of platelet procoagulant function; evaluations of platelet ultrastructure; high-throughput sequencing and other molecular diagnostic tests. The focus of this article is to review current methods for the diagnostic assessment of platelet function, with a focus on contemporary, best diagnostic laboratory practices, and relationships between clinical and laboratory findings.
Collapse
Affiliation(s)
- Alex Bourguignon
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Subia Tasneem
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Catherine P Hayward
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada.,Department of Medicine, McMaster University, Hamilton, Canada
| |
Collapse
|
24
|
Blood Circulating CD133+ Extracellular Vesicles Predict Clinical Outcomes in Patients with Metastatic Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14051357. [PMID: 35267665 PMCID: PMC8909146 DOI: 10.3390/cancers14051357] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/28/2022] [Accepted: 03/05/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary In this study, we explored the prognostic and predictive value of blood circulating EVs expressing selected surface proteins in patients with metastatic colorectal cancer (mCRC). A recently patented flow cytometry protocol was used for the identification and subtyping of blood circulating EVs in a cohort of patients with stage IV colorectal cancer (n = 54) and in a cohort of healthy controls (n = 48). We observed an increased blood concentration of tumor-induced blood circulating EVs in the mCRC cohort as compared to healthy controls. Additionally, we show an intriguing link between circulating CD133+ EVs and poor clinical outcomes in patients with mCRC. This study provides novel insights about the potential impact of EVs as a relevant source of candidate biomarkers in mCRC. Abstract Colorectal cancer (CRC) is one of the most incident and lethal malignancies worldwide. Recent treatment advances prolonged survival in patients with metastatic colorectal cancer (mCRC). However, there are still few biomarkers to guide clinical management and treatment selection in mCRC. In this study, we applied an optimized flow cytometry protocol for EV identification, enumeration, and subtyping in blood samples of 54 patients with mCRC and 48 age and sex-matched healthy controls (HCs). The overall survival (OS) and overall response rate (ORR) were evaluated in mCRC patients enrolled and treated with a first line fluoropyrimidine-based regimen. Our findings show that patients with mCRC presented considerably higher blood concentrations of total EVs, as well as CD133+ and EPCAM+ EVs compared to HCs. Overall survival analysis revealed that increased blood concentrations of total EVs and CD133+ EVs before treatment were significantly associated with shorter OS in mCRC patients (p = 0.001; and p = 0.0001, respectively). In addition, we observed a correlation between high blood levels of CD133+ EVs at baseline and reduced ORR to first-line systemic therapy (p = 0.045). These findings may open exciting perspectives into the application of novel blood-based EV biomarkers for improved risk stratification and optimized treatment strategies in mCRC.
Collapse
|
25
|
Palacios-Acedo AL, Langiu M, Crescence L, Mège D, Dubois C, Panicot-Dubois L. Platelet and Cancer-Cell Interactions Modulate Cancer-Associated Thrombosis Risk in Different Cancer Types. Cancers (Basel) 2022; 14:730. [PMID: 35159000 PMCID: PMC8833365 DOI: 10.3390/cancers14030730] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 02/01/2023] Open
Abstract
The first cause of death in cancer patients, after tumoral progression itself, is thrombo-embolic disease. This cancer-associated hypercoagulability state is known as Trousseau's syndrome, and the risk for developing thrombotic events differs according to cancer type and stage, as well as within patients. Massive platelet activation by tumor cells is the key mediator of thrombus formation in Trousseau's syndrome. In this literature review, we aimed to compare the interactions between cancer cells and platelets in three different cancer types, with low, medium and high thrombotic risk. We chose oral squamous cell carcinoma for the low-thrombotic-risk, colorectal adenocarcinoma for the medium-thrombotic-risk, and pancreatic carcinoma for the high-thrombotic-risk cancer type. We showcase that understanding these interactions is of the highest importance to find new biomarkers and therapeutic targets for cancer-associated thrombosis.
Collapse
Affiliation(s)
- Ana-Luisa Palacios-Acedo
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
| | - Mélanie Langiu
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
| | - Lydie Crescence
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
- Marseille University, PIVMI (Plateforme d’Imagerie Vasculaire et de Microscopie Intravitale), C2VN (Center for CardioVascular and Nutrition Research), 13385 Marseille, France
| | - Diane Mège
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
- Department of Digestive Surgery, La Timone University Hospital, 13005 Marseille, France
| | - Christophe Dubois
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
- Marseille University, PIVMI (Plateforme d’Imagerie Vasculaire et de Microscopie Intravitale), C2VN (Center for CardioVascular and Nutrition Research), 13385 Marseille, France
| | - Laurence Panicot-Dubois
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
- Marseille University, PIVMI (Plateforme d’Imagerie Vasculaire et de Microscopie Intravitale), C2VN (Center for CardioVascular and Nutrition Research), 13385 Marseille, France
| |
Collapse
|
26
|
Wahab S, Alshahrani MY, Ahmad MF, Abbas H. Current trends and future perspectives of nanomedicine for the management of colon cancer. Eur J Pharmacol 2021; 910:174464. [PMID: 34474029 DOI: 10.1016/j.ejphar.2021.174464] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023]
Abstract
Colon cancer (CC) kills countless people every year throughout the globe. It persists as one of the highly lethal diseases to be treated because the overall survival rate for CC is meagre. Early diagnosis and efficient treatments are two of the biggest hurdles in the fight against cancer. In the present work, we will review thriving strategies for CC targeted drug delivery and critically explain the most recent progressions on emerging novel nanotechnology-based drug delivery systems. Nanotechnology-based animal and human clinical trial studies targeting CC are discussed. Advancements in nanotechnology-based drug delivery systems intended to enhance cellular uptake, improved pharmacokinetics and effectiveness of anticancer drugs have facilitated the powerful targeting of specific agents for CC therapy. This review provides insight into current progress and future opportunities for nanomedicines as potential curative targets for CC treatment. This information could be used as a platform for the future expansion of multi-functional nano constructs for CC's advanced detection and functional drug delivery.
Collapse
Affiliation(s)
- Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Hashim Abbas
- Queens Medical Center, Nottingham University Hospitals, NHS, Nottingham, UK
| |
Collapse
|
27
|
Shim Y, Kwon I, Park Y, Lee HW, Kim J, Kim YD, Nam HS, Park S, Heo JH. Characterization of Ferric Chloride-Induced Arterial Thrombosis Model of Mice and the Role of Red Blood Cells in Thrombosis Acceleration. Yonsei Med J 2021; 62:1032-1041. [PMID: 34672137 PMCID: PMC8542466 DOI: 10.3349/ymj.2021.62.11.1032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/06/2021] [Accepted: 08/19/2021] [Indexed: 11/27/2022] Open
Abstract
PURPOSE The ferric chloride (FeCl3)-induced thrombosis model is widely used for thrombosis research. However, it lacks standardization with uncertainty in the exact mechanism of thrombosis. This study aimed to characterize thrombus formation in a mouse model. MATERIALS AND METHODS We investigated thrombus formation and stability using various FeCl3 concentrations (10%, 20%, 30%, 40%, and 50%, w/v) in carotid arteries of the Institute of Cancer Research (ICR) and C57BL/6N mice using the FeCl3-induced thrombosis model. We also investigated thrombus histopathology using immunohistochemistry and electron microscopy. RESULTS Higher FeCl3 concentrations induced dose-dependent, faster, larger, and more stable thrombus formation in both strains of mice. However, the ICR mice showed better dose-responses in thrombus formation and stability compared to the C57BL/6N mice. Thrombi were fibrin- and platelet-rich without significant changes across FeCl3 concentrations. However, the content of red blood cells (RBCs) increased with increasing FeCl3 concentrations (p for trend <0.001) and inversely correlated with time to occlusion (r=-0.65, p<0.001). While platelets and fibrin were evenly distributed over the thrombus, RBCs were predominantly located near the FeCl3 treatment area. Transmission electron microscopy showed that RBCs attached to and were surrounded by aggregates of degranulated platelets, suggesting their potential role in platelet activation. CONCLUSION Faster and larger thrombus formation is induced in a dose-dependent manner by a wide range of FeCl3 concentrations, but the stable thrombus formation requires higher FeCl3 concentrations. Mouse strain affects thrombus formation and stability. RBCs and their interaction with platelets play a key role in the acceleration of FeCl3-induced thrombosis.
Collapse
Affiliation(s)
- Yeseul Shim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Il Kwon
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Youngseon Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Heow Won Lee
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Jayoung Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Young Dae Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Hyo Suk Nam
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Sungha Park
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hoe Heo
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
28
|
Zhang S, Cao Y, Du J, Liu H, Chen X, Li M, Xiang M, Wang C, Wu X, Liu L, Wang C, Wu Y, Li Z, Fang S, Shi J, Wang L. Neutrophil extracellular traps contribute to tissue plasminogen activator resistance in acute ischemic stroke. FASEB J 2021; 35:e21835. [PMID: 34449927 DOI: 10.1096/fj.202100471rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 01/08/2023]
Abstract
Circulating neutrophil extracellular traps (NETs) resistant to t-PA have not been studied completely although NETs in thrombi may contribute to tissue plasminogen activator (t-PA) resistance. This research intended to elucidate whether circulating NETs are associated with t-PA resistance and the underlying mechanism. The levels of NETs were detected in the circulating neutrophils, ischemic brain tissue of acute ischemic stroke (AIS) patients, and transient middle cerebral artery occlusion (tMCAO) models. NET formation in blood, thrombi, and ischemic brain tissue of mice were analyzed by immunofluorescence. Exposed phosphatidylserine (PS) was assessed using flow cytometry and confocal microscopy. Procoagulant activity (PCA) was evaluated using fibrin formation assays, thrombin, and purified coagulation complex. The plasma levels of NETs in AIS patients were significantly higher than those in healthy individuals. After thrombolysis, a significant increase was noted in NET markers in no-improvement patients, while the changes in improvement patients were not significant. Importantly, NETs were decorated with von Willebrand factor (vWF) and plasminogen activator inhibitor-1 (PAI-1) in the blood and thrombi, which could reverse the fibrinolytic effects. In addition, NETs activated platelets (PLTs) and endothelial cells (ECs), stimulating a procoagulant phenotype and facilitating vWF and PAI-1 release. DNase I, activated protein C (APC), and sivelestat markedly inhibited these effects. Furthermore, targeting NETs protected mice from tMCAO-induced cerebral ischemia, possibly by regulating vWF and PAI-1. In summary, NETs may contribute to t-PA resistance in AIS through activation of PLTs and ECs. Strategies against NETs may present a promising therapeutic approach to improve the thrombolysis efficiency of t-PA in AIS patients.
Collapse
Affiliation(s)
- Shuoqi Zhang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Yuze Cao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingwen Du
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Huan Liu
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiaojing Chen
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Mengdi Li
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Mengqi Xiang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chengyue Wang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiaoming Wu
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Langjiao Liu
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chunli Wang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yinsong Wu
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhuxin Li
- Department of Acupuncture and Moxibustion, College of Acupuncture and Moxibustion, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shaohong Fang
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Jialan Shi
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China.,Department of Medicine, Brigham and Women's Hospital, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
29
|
Papadopoulos V, Tsapakidis K, Markou A, Kokkalis A, Aidarinis C, Kotsakis A. New prophylaxis strategies to reduce the risk of thromboembolism in cancer. Expert Rev Anticancer Ther 2021; 21:1135-1144. [PMID: 34139938 DOI: 10.1080/14737140.2021.1941889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION : Patients with cancer are at risk of thrombotic events, mainly deep vein thrombosis and/or pulmonary embolism. The thrombosis risk is generally 4-6 times higher than in a healthy population and depends on factors related to patient characteristics, tumor factors, and treatment-related factors. The decision-making for prophylactic anticoagulation is individualized according to the relative risks and benefits. The VTE risk has been quantified using different assessment scores. In recent years, an effort has been made to establish "risk assessment models" specifically for patients undergoing chemotherapy. AREAS COVERED This article reviews current data and ongoing research on predictive factors involved in cancer-related thrombosis and it is highlighted the currently suggested strategies for prophylaxis. Several trials that compared the two treatment options, direct factor Xa inhibitor or LMWH, with placebo and not each other are discussed. In this article, was analyzed the safety and efficacy features that led several international organizations such as ASCO, NCCN, and others, to issue guidelines for the prophylaxis and treatment of patients at high risk of thrombosis by using LMWH, fondaparinux and DOACs. EXPERT OPINION ASCO, NCCN, and other international organizations recommend thromboprophylaxis in high risk patients. However, further investigation is needed to define better biomarkers for more accurate identification of cancer patients that will benefit from anticoagulant treatment.
Collapse
Affiliation(s)
- Vasileios Papadopoulos
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece
| | - Konstantinos Tsapakidis
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece
| | - Alexandra Markou
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece
| | - Alexandros Kokkalis
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece
| | | | - Athanasios Kotsakis
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece.,Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Thessaly, Greece
| |
Collapse
|
30
|
Alterations of the Platelet Proteome in Lung Cancer: Accelerated F13A1 and ER Processing as New Actors in Hypercoagulability. Cancers (Basel) 2021; 13:cancers13092260. [PMID: 34066760 PMCID: PMC8125802 DOI: 10.3390/cancers13092260] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The risk of venous thromboembolism in cancer is nine times higher than in the general population and the second leading cause of death in these patients. Tissue factor and downstream plasmatic coagulation cascade are largely responsible for the risk of thrombosis in cancer. In recent years, it has been increasingly recognised that platelets also play a central role in tumour growth and cancer-associated thrombosis. The underlying molecular mechanisms are largely unknown. In order to comprehensively investigate the biochemical changes in platelets from cancers with high risk of thrombosis, we examined the platelet proteome of brain and lung cancer patients in comparison to sex and age-matched healthy controls. However, we only found alterations in lung cancer, where some of these platelet proteins directly promote thrombosis. One example is the increased amount of the enzyme protein disulfide isomerase, which is clinically investigated as an antithrombotic drug target of the plant-based flavonol quercetin. Abstract In order to comprehensively expose cancer-related biochemical changes, we compared the platelet proteome of two types of cancer with a high risk of thrombosis (22 patients with brain cancer, 19 with lung cancer) to 41 matched healthy controls using unbiased two-dimensional differential in-gel electrophoresis. The examined platelet proteome was unchanged in patients with brain cancer, but considerably affected in lung cancer with 15 significantly altered proteins. Amongst these, the endoplasmic reticulum (ER) proteins calreticulin (CALR), endoplasmic reticulum chaperone BiP (HSPA5) and protein disulfide-isomerase (P4HB) were significantly elevated. Accelerated conversion of the fibrin stabilising factor XIII was detected in platelets of patients with lung cancer by elevated levels of a coagulation factor XIII (F13A1) 55 kDa fragment. A significant correlation of this F13A1 cleavage product with plasma levels of the plasmin–α-2-antiplasmin complex and D-dimer suggests its enhanced degradation by the fibrinolytic system. Protein association network analysis showed that lung cancer-related proteins were involved in platelet degranulation and upregulated ER protein processing. As a possible outcome, plasma FVIII, an immediate end product for ER-mediated glycosylation, correlated significantly with the ER-executing chaperones CALR and HSPA5. These new data on the differential behaviour of platelets in various cancers revealed F13A1 and ER chaperones as potential novel diagnostic and therapeutic targets in lung cancer patients.
Collapse
|
31
|
Podoplelova NA, Nechipurenko DY, Ignatova AA, Sveshnikova AN, Panteleev MA. Procoagulant Platelets: Mechanisms of Generation and Action. Hamostaseologie 2021; 41:146-153. [PMID: 33860522 DOI: 10.1055/a-1401-2706] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
During the past decades, it has been increasingly recognized that the major function of accelerating membrane-dependent reactions of blood coagulation is predominantly implemented by a subset of activated platelets. These procoagulant platelets (also called collagen- and thrombin-activated or COAT, coated, necrotic, although there could be subtle differences between these definitions) are uniquely characterized by both procoagulant activity and, at the same time, inactivated integrins and profibrinolytic properties. The mechanisms of their generation both in vitro and in situ have been increasingly becoming clear, suggesting unique and multidirectional roles in hemostasis and thrombosis. In this mini-review, we shall highlight the existing concepts and challenges in this field.
Collapse
Affiliation(s)
- N A Podoplelova
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russia
| | - D Y Nechipurenko
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russia.,Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| | - A A Ignatova
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russia
| | - A N Sveshnikova
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russia.,Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| | - M A Panteleev
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russia.,Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
32
|
Abstract
Circulating microparticles in human plasma may play a significant role in thrombogenesis because they carry the initiator of blood coagulation, tissue factor. Microparticles in blood are derived from diverse cell types, including erythrocytes, endothelial cells and platelets. Thrombin generation is an important part of the coagulation system and might be influenced by the presence of microparticles in the circulation. With this study, we determined the contribution of microparticles to increased thrombin generation in plasma samples received for thrombophilia workup and compare that with normal plasma. Microparticles were isolated from 50 plasma samples with increased thrombin generation and 20 plasma samples with normal thrombin generation, using filtration. Thrombin generation assay were performed by adding a low concentration of tissue factor-containing phospholipids and a fluorescence substrate for thrombin formation to plasma samples and measuring fluorescence at 1-min intervals over a period of 90 min on all samples (with and without the presence of microparticles). The peak thrombin, velocity-index and area under the curve were calculated. Microparticles contribute to the different parameters in samples with increased thrombin generation as follows: 50 ± 19% for peak thrombin, 58 ± 24% for velocity-index and 35 ± 13% for area under the curve. Microparticles did not contribute to thrombin generation in plasma samples with normal thrombin generation. Microparticles play a significant role in coagulation and contribute largely to increased thrombin generation in plasma; however, microparticles do not contribute to coagulation in the plasma of participants with normal thrombin generation.
Collapse
|
33
|
Dhami SPS, Patmore S, O'Sullivan JM. Advances in the Management of Cancer-Associated Thrombosis. Semin Thromb Hemost 2021; 47:139-149. [PMID: 33636745 DOI: 10.1055/s-0041-1722863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The association between cancer and venous thromboembolism (VTE) has been established for more than 150 years. Nevertheless, cancer-associated thrombosis still remains a major clinical challenge and is associated with significant morbidity and mortality for patients with cancer. The clinical presentation of cancer-associated thrombosis can be distinct from that of a patient without an underlying malignancy. Moreover, specific cancer types, including pancreatic cancer and hematological malignancies, as well as advanced stage disease can confer a significant thrombotic risk. This risk is further augmented by specific anticancer treatment modalities. The pathophysiology of cancer-associated thrombosis is complex and multifactorial. However, understanding the biological mechanisms underpinning VTE risk may provide insight into novel targeted prophylaxis in cancer patients. Over the last decade, low-molecular-weight heparin has been the preferred anticoagulant agent for patients with cancer-associated thrombosis due to improved efficacy compared with vitamin K antagonists. However, the advent of direct oral anticoagulants (DOACs) has added to the repertoire of ammunition now at the disposal of clinicians to aid in the management of cancer-associated thrombosis. Several randomized controlled trials have now been published, demonstrating DOAC as a noninferior alternative for both the treatment and prevention of cancer-associated thrombosis. Notwithstanding this, limitations for their widespread use remain, with the potential for increased bleeding risk, drug interactions, and poor DOAC metabolism. This review discusses the evidence base for the incidence and risk factors associated with VTE in cancer, development, and refinement of risk prediction models and novel advances in the therapeutic management of cancer-associated thrombosis.
Collapse
Affiliation(s)
- Sukhraj Pal Singh Dhami
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Sean Patmore
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Jamie M O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
34
|
Böhm JK, Schäfer N, Maegele M, Stümpges B, Bauerfeind U, Caspers M. Plasmatic and cell-based enhancement by microparticles originated from platelets and endothelial cells under simulated in vitro conditions of a dilutional coagulopathy. Scand J Trauma Resusc Emerg Med 2021; 29:38. [PMID: 33622398 PMCID: PMC7901091 DOI: 10.1186/s13049-021-00847-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 02/05/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aggressive fluid management and other external factors may lead to hypothermia, acidosis and hemodilution (defined as Lethal Triad, LT) contributing to a trauma-induced coagulopathy (TIC) that worsens patients' outcomes. Procoagulant microparticles (MP) are crucial players at the interface of cellular and plasmatic coagulation. However, their functions remain largely unexplored. This study aimed to characterize effects of MP subtypes and concentrations on functional coagulation under in vitro simulated conditions. METHODS Blood from eleven volunteers were collected to simulate in vitro conditions of hemodilution (HD) and LT, respectively. HD was induced by replacing a blood volume of 33% by crystalloids and for LT, samples were further processed by reducing the temperature to 32 °C and lowering the pH to 6.8. MP were obtained either from platelet concentrates (platelet-derived MP, PDMP) or from cell culture (ECV304 cells for endothelial-derived MP, EDMP) by targeted stimulation. After introducing MP to in vitro conditions, we measured their concentration-dependent effects (1.000, 10.000 and 15.000 MP/μl blood) on coagulation compared to whole blood (WB). For each condition, coagulation was characterized by flow cytometric platelet activation and by quantification of fibrin clot propagation using Thrombodynamics® technology. RESULTS MP originated from platelets and endothelial cells affected blood coagulation in a concentration-dependent manner. Particularly, high PDMP quantities (10.000 and 15.000 PDMP/μl blood) significantly induced platelet activation and fibrin clot growth and size in HD conditions. In LT conditions as well, only high PDMP concentration induced platelet activation, clot growth and size. In contrast, EDMP did not induce platelet activation, but resulted in enhanced formation of spontaneous clots, irrespective of simulated condition. With increasing EDMP concentration, the time until the onset of spontaneous clotting decreased in both HD and LT conditions. DISCUSSION The study demonstrates an essential role of MP within the coagulation process under simulated coagulopathic conditions. PDMP affected platelets promoting clot formation likely by providing a surface enlargement. EDMP presumably affected clotting factors of the plasmatic coagulation resulting in an increased formation of spontaneous clots. CONCLUSION Under simulated conditions of a dilutional coagulopathy, MP from different cellular origin indicate a divergent but both procoagulant mechanism within the coagulation process.
Collapse
Affiliation(s)
- Julia K Böhm
- The Institute for Research in Operative Medicine, Faculty of Health, Department of Medicine, Witten/Herdecke University, Ostmerheimer Straße 200, 51109, Cologne, Germany
| | - Nadine Schäfer
- The Institute for Research in Operative Medicine, Faculty of Health, Department of Medicine, Witten/Herdecke University, Ostmerheimer Straße 200, 51109, Cologne, Germany
| | - Marc Maegele
- The Institute for Research in Operative Medicine, Faculty of Health, Department of Medicine, Witten/Herdecke University, Ostmerheimer Straße 200, 51109, Cologne, Germany
- Department of Traumatology, Orthopaedic Surgery and Sports Traumatology, Cologne-Merheim Medical Centre (CMMC), Witten/Herdecke University, Campus Cologne-Merheim, Ostmerheimer Str. 200, D-51109, Cologne, Germany
| | - Birgit Stümpges
- Department of Haematology and Transfusion Medicine, Cologne-Merheim Medical Centre (CMMC), Witten/Herdecke University, Campus Cologne-Merheim, Ostmerheimer Str. 200, D-51109, Cologne, Germany
| | - Ursula Bauerfeind
- Department of Haematology and Transfusion Medicine, Cologne-Merheim Medical Centre (CMMC), Witten/Herdecke University, Campus Cologne-Merheim, Ostmerheimer Str. 200, D-51109, Cologne, Germany
| | - Michael Caspers
- The Institute for Research in Operative Medicine, Faculty of Health, Department of Medicine, Witten/Herdecke University, Ostmerheimer Straße 200, 51109, Cologne, Germany.
- Department of Traumatology, Orthopaedic Surgery and Sports Traumatology, Cologne-Merheim Medical Centre (CMMC), Witten/Herdecke University, Campus Cologne-Merheim, Ostmerheimer Str. 200, D-51109, Cologne, Germany.
| |
Collapse
|
35
|
Tohidi-Esfahani I, Lee CSM, Liang HPH, Chen VMY. Procoagulant platelets: Laboratory detection and clinical significance. Int J Lab Hematol 2021; 42 Suppl 1:59-67. [PMID: 32543068 DOI: 10.1111/ijlh.13197] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
Platelets play a critical role in both haemostasis and thrombosis, and it is now evident that not all platelets behave the same when they are called to action. A functionally distinct subpopulation of platelets forms in response to maximal agonist stimulation: the procoagulant platelet. This platelet subpopulation is defined by its ability to expose phosphatidylserine on its surface, allowing for coagulation factor complexes to form and generate bursts of thrombin and fibrin to stabilize platelet clots. Reduced levels of procoagulant platelets have been linked to bleeding in Scott's syndrome and haemophilia A patients, and elevated levels have been demonstrated in many thrombotic disorders, including identifying patients at higher risk for stroke recurrence. One obstacle for incorporating an assay for measuring procoagulant platelets into clinical management algorithms is the lack of consensus on the exact definition and markers for this subpopulation. This review will outline the biological characteristics of procoagulant platelets and the laboratory assays currently used to identify them in research settings. It will summarize the findings of clinical research demonstrating the relevance of measuring the procoagulant platelet levels in patients and will discuss how an appropriate assay can be used to elucidate the mechanism behind the formation of this subpopulation, facilitating novel drug discovery to improve upon current outcomes in cardiovascular and other thrombotic disorders.
Collapse
Affiliation(s)
- Ibrahim Tohidi-Esfahani
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Christine S M Lee
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Hai Po H Liang
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Vivien M Y Chen
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
36
|
Human placental mesenchymal stem cells improve stroke outcomes via extracellular vesicles-mediated preservation of cerebral blood flow. EBioMedicine 2020; 63:103161. [PMID: 33348090 PMCID: PMC7753936 DOI: 10.1016/j.ebiom.2020.103161] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Background Besides long-term trans-differentiation into neural cells, benefits of stem cell therapy (SCT) in ischemic stroke may include secretion of protective factors, which partly reflects extracellular vesicle (EVs) released by stem cell. However, the mechanism(s) by which stem cells/EVs limit stroke injury have yet to be fully defined. Methods We evaluated the protection effect of human placenta mesenchymal stem cells (hPMSC) as a potential form of SCT in experimental ischemic stroke ‘transient middle cerebral artery occusion (MCAO)/reperfusion’ mice model. Findings We found for the first time that intraperitoneal administration of hPMSCs or intravenous hPMSC-derived EVs, given at the time of reperfusion, significantly protected the ipsilateral hemisphere from ischemic injury. This protection was associated with significant restoration of normal blood flow to the post-MCAO brain. More importantly, EVs derived from hPMSC promote paracrine-based protection of SCT in the MCAO model in a cholesterol/lipid-dependent manner. Interpretation Together, our results demonstrated beneficial effects of hPMSC/EVs in experimental stroke models which could permit the rapid “translation” of these cells into clinical trials in the near-term.
Collapse
|
37
|
West AL, Michaelson LV, Miles EA, Haslam RP, Lillycrop KA, Georgescu R, Han L, Napier JA, Calder PC, Burdge GC. Lipidomic Analysis of Plasma from Healthy Men and Women Shows Phospholipid Class and Molecular Species Differences between Sexes. Lipids 2020; 56:229-242. [PMID: 33284478 PMCID: PMC8048887 DOI: 10.1002/lipd.12293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/14/2020] [Accepted: 10/29/2020] [Indexed: 12/19/2022]
Abstract
The phospholipid composition of lipoproteins is determined by the specificity of hepatic phospholipid biosynthesis. Plasma phospholipid 20:4n‐6 and 22:6n‐3 concentrations are higher in women than in men. We used this sex difference in a lipidomics analysis of the impact of endocrine factors on the phospholipid class and molecular species composition of fasting plasma from young men and women. Diester species predominated in all lipid classes measured. 20/54 Phosphatidylcholine (PtdCho) species were alkyl ester, 15/48 phosphatidylethanolamine (PtdEtn) species were alkyl ester, and 12/48 PtdEtn species were alkenyl ester. There were no significant differences between sexes in the proportions of alkyl PtdCho species. The proportion of alkyl ester PtdEtn species was greater in women than men, while the proportion of alkenyl ester PtdEtn species was greater in men than women. None of the phosphatidylinositol (PtdIns) or phosphatidylserine (PtdSer) molecular species contained ether‐linked fatty acids. The proportion of PtdCho16:0_22:6, and the proportions of PtdEtn O‐16:0_20:4 and PtdEtn O‐18:2_20:4 were greater in women than men. There were no sex differences in PtdIns and PtdSer molecular species compositions. These findings show that plasma phospholipids can be modified by sex. Such differences in lipoprotein phospholipid composition could contribute to sexual dimorphism in patterns of health and disease.
Collapse
Affiliation(s)
- Annette L West
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - Louise V Michaelson
- Department of Plant Sciences, Rothamsted Research, West Common, Harpenden, AL5 2JQ, UK
| | - Elizabeth A Miles
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - Richard P Haslam
- Department of Plant Sciences, Rothamsted Research, West Common, Harpenden, AL5 2JQ, UK
| | - Karen A Lillycrop
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, University Road, Southampton, SO17 1BJ, UK
| | - Ramona Georgescu
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - Lihua Han
- Department of Plant Sciences, Rothamsted Research, West Common, Harpenden, AL5 2JQ, UK
| | - Johnathan A Napier
- Department of Plant Sciences, Rothamsted Research, West Common, Harpenden, AL5 2JQ, UK
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - Graham C Burdge
- School of Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| |
Collapse
|
38
|
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
|
39
|
Avisar A, Cohen M, Brenner B, Bronshtein T, Machluf M, Bar-Sela G, Aharon A. Extracellular Vesicles Reflect the Efficacy of Wheatgrass Juice Supplement in Colon Cancer Patients During Adjuvant Chemotherapy. Front Oncol 2020; 10:1659. [PMID: 32984039 PMCID: PMC7479215 DOI: 10.3389/fonc.2020.01659] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction Colorectal cancer (CC) is the third most common type of cancer, accounting for 10% of all cancer cases. Adjuvant chemotherapy is recommended in stages II–III CC. Wheatgrass juice (WGJ) from wheat seeds has high nutritional values, may induce synergistic benefits to chemotherapy and may attenuate chemotherapy-related side effects. Extracellular vesicles (EVs) are subcellular membrane blebs. EVs include exosomes (generated in the endosome, in size <150 nm) and microvesicles (shed from the plasma cell membrane) provide information on their parental cells and play a role in intercellular communication. We aimed to elucidate the effects of chemotherapy administration with supportive treatment of WGJ on CC patients’ EVs characteristics. Methods EVs were isolated from the blood samples of 15 healthy controls (HCs) and 50 CC patients post-surgery, treated by chemotherapy, with or without additional daily WGJ. Blood samples were taken before, during, and at the end of chemotherapy. EVs were characterized by size, concentration, membrane antigens and cytokine content using nanoparticle-tracking analysis, western blot, flow cytometry, and protein array methods. Results EVs were found to be similar by size and concentration with reduced levels of exosome markers (CD81) on samples at the end of combined treatment (chemotherapy and WGJ). Higher levels of endothelial EVs, which may indicate impairment of the vascular endothelial cells during treatment, were found in CC patients treated by chemotherapy only compared to those with chemotherapy and daily WGJ. Also, EVs thrombogenicity was lower in patients added WGJ compared to patients who had only chemotherapy (levels of tissue factor p = 0.029 and endothelial protein C receptor p = 0.005). Following treatments, levels of vascular endothelial growth factor receptors (VEGFR-1) and the majority of growth-factors/pro-inflammatory cytokines were higher in EVs of patients treated by chemotherapy only than in EVs obtained from patients with the combined treatment. Conclusion Daily consumption of WGJ during chemotherapy may reduce vascular damage and chemotherapy-related thrombogenicity, growth factors and cytokines, as reflected by the characteristics of patient’s EVs.
Collapse
Affiliation(s)
- Adva Avisar
- The Graduate Studies Authority, University of Haifa, Haifa, Israel
| | - Miri Cohen
- School of Social Work, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| | - Benjamin Brenner
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tomer Bronshtein
- The Lab for Cancer Drug Delivery & Cell Based Technologies, The Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Marcelle Machluf
- The Lab for Cancer Drug Delivery & Cell Based Technologies, The Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Gil Bar-Sela
- Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Cancer Center, Emek Medical Center, Afula, Israel
| | - Anat Aharon
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Hematology Research Laboratory, Hematology and Bone Marrow Transplantation, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
40
|
Campos J, Brill A. The role of bone marrow-derived cells in venous thromboembolism. Int J Biochem Cell Biol 2020; 128:105850. [PMID: 32950686 PMCID: PMC7607213 DOI: 10.1016/j.biocel.2020.105850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/03/2020] [Accepted: 09/14/2020] [Indexed: 12/22/2022]
Abstract
Venous thrombosis is a life-threatening condition with high morbidity and mortality. Abnormal functioning of different cells in the blood is an integral part of its pathogenesis. In this review, we describe the contribution of bone marrow-derived cells to the development of this debilitating disease. We present both epidemiological and clinical data demonstrating involvement of various cell types in venous thrombosis, and discuss potential mechanisms underlying these effects. Modern concepts including recently discovered new paradigms in thrombosis, such as neutrophil extracellular traps, mast cells, and polyphosphate, are summarized.
Collapse
Affiliation(s)
- Joana Campos
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Alexander Brill
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| |
Collapse
|
41
|
Menck K, Sivaloganathan S, Bleckmann A, Binder C. Microvesicles in Cancer: Small Size, Large Potential. Int J Mol Sci 2020; 21:E5373. [PMID: 32731639 PMCID: PMC7432491 DOI: 10.3390/ijms21155373] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EV) are secreted by all cell types in a tumor and its microenvironment (TME), playing an essential role in intercellular communication and the establishment of a TME favorable for tumor invasion and metastasis. They encompass a variety of vesicle populations, among them the well-known endosomal-derived small exosomes (Exo), but also larger vesicles (diameter > 100 nm) that are shed directly from the plasma membrane, the so-called microvesicles (MV). Increasing evidence suggests that MV, although biologically different, share the tumor-promoting features of Exo in the TME. Due to their larger size, they can be readily harvested from patients' blood and characterized by routine methods such as conventional flow cytometry, exploiting the plethora of molecules expressed on their surface. In this review, we summarize the current knowledge about the biology and the composition of MV, as well as their role within the TME. We highlight not only the challenges and potential of MV as novel biomarkers for cancer, but also discuss their possible use for therapeutic intervention.
Collapse
Affiliation(s)
- Kerstin Menck
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.S.); (A.B.)
| | - Suganja Sivaloganathan
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.S.); (A.B.)
| | - Annalen Bleckmann
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.S.); (A.B.)
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Claudia Binder
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
42
|
Sheikh Hosseini M, Parhizkar Roudsari P, Gilany K, Goodarzi P, Payab M, Tayanloo-Beik A, Larijani B, Arjmand B. Cellular Dust as a Novel Hope for Regenerative Cancer Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1288:139-160. [DOI: 10.1007/5584_2020_537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
43
|
Berckmans RJ, Lacroix R, Hau CM, Sturk A, Nieuwland R. Extracellular vesicles and coagulation in blood from healthy humans revisited. J Extracell Vesicles 2019; 8:1688936. [PMID: 31762964 PMCID: PMC6853244 DOI: 10.1080/20013078.2019.1688936] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/25/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background: In 2001, we studied the presence and coagulant properties of “microparticles” in the blood of healthy humans. Since then, multiple improvements in detection, isolation and functional characterization of the now called “extracellular vesicles” (EVs) have been made, and shortcomings were identified. Aim: To revisit the presence and function of EVs in blood from healthy humans. Methods: Blood was collected from 20 healthy donors. EV-containing plasma was prepared according to new guidelines, and plasma was diluted to prevent swarm detection. Single EVs were measured by flow cytometry with known sensitivity of fluorescence and light scatter. The haemostatic properties of EVs were measured by thrombin-, fibrin-, and plasmin generation. Plasma concentrations of thrombin-antithrombin complexes and prothrombin fragment 1 + 2 were measured to assess the coagulation status in vivo. Results: Compared to 2001, the total concentrations of detected EVs increased from 190- to 264-fold. In contrast to 2001, however, EVs are non-coagulant which we show can be attributed to improvements in blood collection and plasma preparation. No relation is present between the plasma concentrations of EVs and either TAT or F1 + 2. Finally, we show that EVs support plasmin generation. Discussion: Improvements in blood collection, plasma preparation and detection of EVs reveal that results from earlier studies have to be interpreted with care. Compared to 2001, higher concentrations of EVs are detected in blood of healthy humans which promote fibrinolysis rather than coagulation.
Collapse
Affiliation(s)
- René J Berckmans
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Vesicle Observation Centre, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Romaric Lacroix
- Department of Hematology and Vascular Biology, CHU la Conception, APHM, Marseille, France.,Aix-Marseille University, C2VN, INSERM, Faculty of Pharmacy, Marseille, France
| | - Chi M Hau
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Vesicle Observation Centre, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Auguste Sturk
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Vesicle Observation Centre, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Vesicle Observation Centre, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
44
|
Microparticles from tumors exposed to radiation promote immune evasion in part by PD-L1. Oncogene 2019; 39:187-203. [PMID: 31467431 PMCID: PMC6937213 DOI: 10.1038/s41388-019-0971-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 07/26/2019] [Accepted: 08/02/2019] [Indexed: 12/21/2022]
Abstract
Radiotherapy induces immune-related responses in cancer patients by various mechanisms. Here, we investigate the immunomodulatory role of tumor-derived microparticles (TMPs)—extracellular vesicles shed from tumor cells—following radiotherapy. We demonstrate that breast carcinoma cells exposed to radiation shed TMPs containing elevated levels of immune-modulating proteins, one of which is programmed death-ligand 1 (PD-L1). These TMPs inhibit cytotoxic T lymphocyte (CTL) activity both in vitro and in vivo, and thus promote tumor growth. Evidently, adoptive transfer of CTLs pre-cultured with TMPs from irradiated breast carcinoma cells increases tumor growth rates in mice recipients in comparison with control mice receiving CTLs pre-cultured with TMPs from untreated tumor cells. In addition, blocking the PD-1-PD-L1 axis, either genetically or pharmacologically, partially alleviates TMP-mediated inhibition of CTL activity, suggesting that the immunomodulatory effects of TMPs in response to radiotherapy is mediated, in part, by PD-L1. Overall, our findings provide mechanistic insights into the tumor immune surveillance state in response to radiotherapy and suggest a therapeutic synergy between radiotherapy and immune checkpoint inhibitors.
Collapse
|
45
|
Zhang Y, Wang C, Yu M, Zhao X, Du J, Li Y, Jing H, Dong Z, Kou J, Bi Y, Novakovic VA, Zhou J, Shi J. Neutrophil extracellular traps induced by activated platelets contribute to procoagulant activity in patients with colorectal cancer. Thromb Res 2019; 180:87-97. [PMID: 31271975 DOI: 10.1016/j.thromres.2019.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 06/03/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022]
Abstract
Patients with colorectal cancer (CRC) are at increased risk of venous thrombosis, but the precise mechanisms of thrombogenesis in CRC remain largely unknown. We aimed to identify the novel role of neutrophil extracellular traps (NETs) in the induction of procoagulant activity (PCA) in CRC, and to evaluate its interactions with platelets and endothelial cells (ECs). In this study, we first showed that the levels of NETs in the peripheral blood of CRC patients were increased in parallel with cancer progression and reached significance in stage II patients compared to healthy subjects. In addition, neutrophils from CRC patients were more prone to produce NETs, resulting in shortened coagulation time, significantly increased thrombin-antithrombin (TAT) complexes and fibrin fibrils compared to healthy controls. Furthermore, platelets from CRC patients stimulated healthy neutrophils to extrude NETs, which could be inhibited by the depletion of HMGB1. Conversely, NETs from CRC patients could also induce the exposure of PS on platelets, leading to markedly enhanced PCA. Importantly, ECs were also converted to a procoagulant phenotype when exposed to NETs from CRC patients. The PCA of NETs-activated platelets or ECs could be inhibited either by the cleavage of NETs with DNase1 or the blockage of histone with activated protein C (APC). Our results reveal the complex interactions between neutrophils, platelets and ECs and their potential role in the hypercoagulable state in CRC. We propose that NETs may provide new therapeutic targets to combat the thrombotic consequences of CRC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Chunxu Wang
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Muxin Yu
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Xinyi Zhao
- Department of Cardiology of the Second Hospital, Harbin Medical University, 148 Baojian Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Jingwen Du
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Yueyue Li
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Haijiao Jing
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Zengxiang Dong
- Department of Cardiology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Junjie Kou
- Department of Cardiology of the Second Hospital, Harbin Medical University, 148 Baojian Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Yayan Bi
- Department of Cardiology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Valerie A Novakovic
- Departments of Research and Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, 1400 VFW Parkway, West Roxbury, Boston, MA, USA
| | - Jin Zhou
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China.
| | - Jialan Shi
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China; Departments of Research and Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, 1400 VFW Parkway, West Roxbury, Boston, MA, USA.
| |
Collapse
|
46
|
Silachev DN, Goryunov KV, Shpilyuk MA, Beznoschenko OS, Morozova NY, Kraevaya EE, Popkov VA, Pevzner IB, Zorova LD, Evtushenko EA, Starodubtseva NL, Kononikhin AS, Bugrova AE, Evtushenko EG, Plotnikov EY, Zorov DB, Sukhikh GT. Effect of MSCs and MSC-Derived Extracellular Vesicles on Human Blood Coagulation. Cells 2019; 8:cells8030258. [PMID: 30893822 PMCID: PMC6468445 DOI: 10.3390/cells8030258] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/09/2019] [Accepted: 03/15/2019] [Indexed: 01/10/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a potent therapeutic tool for the treatment of a number of pathologies, including immune pathologies. However, unwelcome effects of MSCs on blood coagulation have been reported, motivating us to explore the thrombotic properties of human MSCs from the umbilical cord. We revealed strong procoagulant effects of MSCs on human blood and platelet-free plasma using rotational thromboelastometry and thrombodynamic tests. A similar potentiation of clotting was demonstrated for MSC-derived extracellular vesicles (EVs). To offer approaches to avoid unwanted effects, we studied the impact of a heparin supplement on MSC procoagulative properties. However, MSCs still retained procoagulant activity toward blood from children receiving a therapeutic dose of unfractionated heparin. An analysis of the mechanisms responsible for the procoagulant effect of MSCs/EVs revealed the presence of tissue factor and other proteins involved in coagulation-associated pathways. Also, we found that some MSCs and EVs were positive for annexin V, which implies the presence of phosphatidylserine on their surfaces, which can potentiate clot formation. Thus, we revealed procoagulant activity of MSCs/EVs associated with the presence of phosphatidylserine and tissue factor, which requires further analysis to avoid adverse effects of MSC therapy in patients with a risk of thrombosis.
Collapse
Affiliation(s)
- Denis N. Silachev
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Kirill V. Goryunov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Margarita A. Shpilyuk
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Olga S. Beznoschenko
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Natalya Y. Morozova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Elizaveta E. Kraevaya
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Vasily A. Popkov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Irina B. Pevzner
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Ljubava D. Zorova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | | | - Natalia L. Starodubtseva
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Moscow Institute of Physics and Technology, Moscow 141701, Russia
| | - Alexey S. Kononikhin
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Moscow Institute of Physics and Technology, Moscow 141701, Russia
| | - Anna E. Bugrova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | | | - Egor Y. Plotnikov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P. & D.B.Z.)
| | - Dmitry B. Zorov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P. & D.B.Z.)
| | - Gennady T. Sukhikh
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Department of Obstetrics, Gynecology, Perinatology and Reproductology, Institute of Professional Education, First Moscow State Medical University Named after I.M. Sechenov, Moscow 119992, Russia
| |
Collapse
|
47
|
Feng Q, Stork CJ, Xu S, Yuan D, Xia X, LaPenna KB, Guo G, Sun H, Xu L, Siedlecki CA, Brundage KM, Sheaffer N, Schell TD, He P. Increased circulating microparticles in streptozotocin-induced diabetes propagate inflammation contributing to microvascular dysfunction. J Physiol 2019; 597:781-798. [PMID: 30548258 PMCID: PMC6355626 DOI: 10.1113/jp277312] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 12/11/2018] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS Circulating microparticles (MPs) are elevated in many cardiovascular diseases and have been considered as biomarkers of disease prognosis; however, current knowledge of MP functions has been mainly derived from in vitro studies and their precise impact on vascular inflammation and disease progression remains obscure. Using a diabetic rat model, we identified a >130-fold increase in MPs in plasma of diabetic rats compared to normal rats, the majority of which circulated as aggregates, expressing multiple cell markers and largely externalized phosphatidylserine; vascular images illustrate MP biogenesis and their manifestations in microvessels of diabetic rats. Using combined single microvessel perfusion and systemic cross-transfusion approaches, we delineated how diabetic MPs propagate inflammation in the vasculature and transform normal microvessels into an inflammatory phenotype observed in the microvessels of diabetic rats. Our observations derived from animal studies resembling conditions in diabetic patients, providing a mechanistic insight into MP-mediated pathogenesis of diabetes-associated multi-organ microvascular dysfunction. ABSTRACT In various cardiovascular diseases, microparticles (MPs), the membrane-derived vesicles released during cell activation, are markedly increased in the circulation. These MPs have been recognized to play diverse roles in the regulation of cellular functions. However, current knowledge of MP function has been largely derived from in vitro studies. The precise impact of disease-induced MPs on vascular inflammation and disease progression remains obscure. In this study we investigated the biogenesis, profile and functional roles of circulating MPs using a streptozotocin-induced diabetic rat model with well-characterized microvascular functions. Our study revealed a >130-fold increase in MPs in the plasma of diabetic rats compared to normal rats. The majority of these MPs originate from platelets, leukocytes and endothelial cells (ECs), and circulate as aggregates. Diabetic MPs show greater externalized phosphatidylserine (PS) than normal MPs. When diabetic plasma or isolated diabetic MPs were perfused into normal microvessels or systemically transfused into normal rats, MPs immediately adhered to endothelium and subsequently mediated leukocyte adhesion. These microvessels then exhibited augmented permeability responses to inflammatory mediators, replicating the microvascular manifestations observed in diabetic rats. These effects were abrogated when MPs were removed from diabetic plasma or when diabetic MPs were pre-coated with a lipid-binding protein, annexin V, suggesting externalized PS to be key in mediating MP interactions with endothelium and leukocytes. Our study demonstrated that the elevated MPs in diabetic plasma are actively involved in the propagation of vascular inflammation through their adhesive surfaces, providing mechanistic insight into the pathogenesis of multi-organ vascular dysfunction that commonly occurs in diabetic patients.
Collapse
Affiliation(s)
- Qilong Feng
- Department of Cellular and Molecular Physiology, College of MedicinePenn State UniversityHersheyPA17033USA
- Department of Physiology and Pharmacology, School of MedicineWest Virginia UniversityMorgantownWV26506USA
- Department of PhysiologyShanxi Medical UniversityTaiyuanShanxiChina030001
| | - Christian J. Stork
- Department of Physiology and Pharmacology, School of MedicineWest Virginia UniversityMorgantownWV26506USA
| | - Sulei Xu
- Department of Cellular and Molecular Physiology, College of MedicinePenn State UniversityHersheyPA17033USA
- Department of Physiology and Pharmacology, School of MedicineWest Virginia UniversityMorgantownWV26506USA
| | - Dong Yuan
- Department of Physiology and Pharmacology, School of MedicineWest Virginia UniversityMorgantownWV26506USA
| | - Xinghai Xia
- Department of Cellular and Molecular Physiology, College of MedicinePenn State UniversityHersheyPA17033USA
| | - Kyle B. LaPenna
- Department of Cellular and Molecular Physiology, College of MedicinePenn State UniversityHersheyPA17033USA
| | - Ge Guo
- Department of Physiology and Pharmacology, School of MedicineWest Virginia UniversityMorgantownWV26506USA
| | - Haoyu Sun
- Department of Cellular and Molecular Physiology, College of MedicinePenn State UniversityHersheyPA17033USA
| | - Li‐Chong Xu
- Department of Surgery, College of MedicinePenn State UniversityHersheyPA17033USA
| | | | - Kathleen M. Brundage
- Department of Microbiology, Immunology and Cell Biology, School of MedicineWest Virginia UniversityMorgantownWV26506USA
| | - Nate Sheaffer
- Flow Cytometry Core, College of MedicinePenn State UniversityHersheyPA17033USA
| | - Todd D. Schell
- Flow Cytometry Core, College of MedicinePenn State UniversityHersheyPA17033USA
- Department of Microbiology and Immunology, College of MedicinePenn State UniversityHersheyPA17033USA
| | - Pingnian He
- Department of Cellular and Molecular Physiology, College of MedicinePenn State UniversityHersheyPA17033USA
- Department of Physiology and Pharmacology, School of MedicineWest Virginia UniversityMorgantownWV26506USA
| |
Collapse
|
48
|
Thrombin Generation and Cancer: Contributors and Consequences. Cancers (Basel) 2019; 11:cancers11010100. [PMID: 30654498 PMCID: PMC6356447 DOI: 10.3390/cancers11010100] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
The high occurrence of cancer-associated thrombosis is associated with elevated thrombin generation. Tumour cells increase the potential for thrombin generation both directly, through the expression and release of procoagulant factors, and indirectly, through signals that activate other cell types (including platelets, leukocytes and erythrocytes). Furthermore, cancer treatments can worsen these effects. Coagulation factors, including tissue factor, and inhibitors of coagulation are altered and extracellular vesicles (EVs), which can promote and support thrombin generation, are released by tumour and other cells. Some phosphatidylserine-expressing platelet subsets and platelet-derived EVs provide the surface required for the assembly of coagulation factors essential for thrombin generation in vivo. This review will explore the causes of increased thrombin production in cancer, and the availability and utility of tests and biomarkers. Increased thrombin production not only increases blood coagulation, but also promotes tumour growth and metastasis and as a consequence, thrombin and its contributors present opportunities for treatment of cancer-associated thrombosis and cancer itself.
Collapse
|
49
|
Li M, Guo Q, Hu W. Incidence, risk factors, and outcomes of venous thromboembolism after oncologic surgery: A systematic review and meta-analysis. Thromb Res 2018; 173:48-56. [PMID: 30471508 DOI: 10.1016/j.thromres.2018.11.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 11/06/2018] [Accepted: 11/14/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND The risk and prognosis of VTE associated with oncologic surgery need to be quantified to guide patient management. We aimed to examine the availability of data and to report the incidence of venous thromboembolism (VTE) in cancer patients after surgery, as well as the clinical outcomes of VTE following oncologic surgery. METHODS We searched multiple databases for terms related to VTE after oncologic surgery from inception to November 1, 2017. A random-effects meta-analysis was done to calculate the pooled incidence of VTE. RESULTS Of the 8611 citations identified, 136 studies including 1,481,659 patients met the eligibility criteria. The overall incidence of VTE was estimated to be 2.3% (95% CI 2.1-2.5). Bone and soft tissue cancer (10.6%, 95% CI 2.9-18.2) and lung cancer (8.1%, 95% CI 3.7-12.6) were associated with the highest and second highest risk of postoperative VTE, respectively. Age (standardized mean difference [SMD] = 0.46, 95% CI 0.40-0.53; I2 = 93.8%), radiation (OR 1.29, 95% CI 1.03-1.62; I2 = 34.6%), transfusion (OR 1.96, 95% CI 1.48-2.59; I2 = 57.0%), and operative time (SMD = 1.12, 95% CI 1.07-1.16; I2 = 100%) were possible risk factors for postoperative VTE. Patients with VTE versus those without had increased odds of all-cause fatal events (11.15, 95% CI 4.07-30.56; I2 = 92.0%). CONCLUSIONS The risk of VTE after oncologic surgery remains high, and this risk varied according to the cancer type, study region, surgical location, and thromboprophylactic strategy. VTE is associated with increased mortality at the early stage of cancer surgery.
Collapse
Affiliation(s)
- Mao Li
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Qiang Guo
- Department of Vascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Weiming Hu
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China.
| |
Collapse
|
50
|
Cancer-Associated Thrombosis in Cirrhotic Patients with Hepatocellular Carcinoma. Cancers (Basel) 2018; 10:cancers10110450. [PMID: 30453547 PMCID: PMC6266984 DOI: 10.3390/cancers10110450] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/02/2018] [Accepted: 11/13/2018] [Indexed: 12/24/2022] Open
Abstract
It is common knowledge that cancer patients are more prone to develop venous thromboembolic complications (VTE). It is therefore not surprising that patients with hepatocellular carcinoma (HCC) present with a significant risk of VTE, with the portal vein being the most frequent site (PVT). However, patients with HCC are peculiar as both cancer and liver cirrhosis are conditions that can perturb the hemostatic balance towards a prothrombotic state. Because HCC-related hypercoagulability is not clarified at all, the aim of the present review is to summarize the currently available knowledge on epidemiology and pathogenesis of non-malignant thrombotic complications in patients with liver cirrhosis and HCC. They are at increased risk to develop both PVT and non-splanchnic VTE, indicating that both local and systemic factors can foster the development of site-specific thrombosis. Recent studies have suggested multiple and often interrelated mechanisms through which HCC can tip the hemostatic balance of liver cirrhosis towards hypercoagulability. Described mechanisms include increased fibrinogen concentration/polymerization, thrombocytosis, and release of tissue factor-expressing extracellular vesicles. Currently, there are no specific guidelines on the use of thromboprophylaxis in this unique population. There is the urgent need of prospective studies assessing which patients have the highest prothrombotic profile and would therefore benefit from early thromboprophylaxis.
Collapse
|