1
|
Wang QH, Cheng S, Han CY, Yang S, Gao SR, Yin WZ, Song WZ. Tailoring cell-inspired biomaterials to fuel cancer therapy. Mater Today Bio 2025; 30:101381. [PMID: 39742146 PMCID: PMC11683242 DOI: 10.1016/j.mtbio.2024.101381] [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: 11/04/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
Cancer stands as a predominant cause of mortality across the globe. Traditional cancer treatments, including surgery, radiotherapy, and chemotherapy, are effective yet face challenges like normal tissue damage, complications, and drug resistance. Biomaterials, with their advantages of high efficacy, targeting, and spatiotemporal controllability, have been widely used in cancer treatment. However, the biocompatibility limitations of traditional synthetic materials have restricted their clinical translation and application. Natural cell-inspired biomaterials inherently possess the targeting abilities of cells, biocompatibility, and immune evasion capabilities. Therefore, cell-inspired biomaterials can be used alone or in combination with other drugs or treatment strategies for cancer therapy. In this review, we first introduce the timeline of key milestones in cell-inspired biomaterials for cancer therapy. Then, we describe the abnormalities in cancer including biophysics, cellular biology, and molecular biology aspects. Afterwards, we summarize the design strategies of cell-inspired antitumor biomaterials. Subsequently, we elaborate on the application of antitumor biomaterials inspired by various cell types. Finally, we explore the current challenges and prospects of cell-inspired antitumor materials. This review aims to provide new opportunities and references for the development of antitumor cell-inspired biomaterials.
Collapse
Affiliation(s)
- Qi-Hui Wang
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Shi Cheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
| | - Chun-Yu Han
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Shuang Yang
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Sheng-Rui Gao
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Hospital of Jilin University, Changchun, 130061, PR China
| | - Wan-Zhong Yin
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Hospital of Jilin University, Changchun, 130061, PR China
| | - Wen-Zhi Song
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| |
Collapse
|
2
|
Xin X, Koenen RR. Assessing platelet-derived extracellular vesicles for potential as therapeutic targets in cardiovascular diseases. Expert Opin Ther Targets 2025. [PMID: 39817690 DOI: 10.1080/14728222.2025.2454617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/17/2024] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
INTRODUCTION Cardiovascular disease (CVD) is the leading cause of death worldwide. Platelet-derived extracellular vesicles (PEV) have attracted extensive attention in cardiovascular disease research in recent years because their cargo is involved in a variety of pathophysiological processes, such as thrombosis, immune response, promotion or inhibition of inflammatory response, promotion of angiogenesis as well as cell proliferation and migration. AREAS COVERED This review explores the role of PEV in various cardiovascular diseases (such as atherosclerosis, myocardial infarction, ischemia-reperfusion injury, and heart failure), with relation to its molecular cargo (nucleic acids, bioactive lipids, proteins) and aims to provide new insights in the pathophysiologic role of PEV, and methods for preventing and treating cardiovascular diseases based on PEV. EXPERT OPINION Studies have shown that the cargo of PEV may be dysregulated during cardiovascular disease and delivery to tissues can result in detrimental pathophysiologic effects. Counteracting this process might have the potential to inhibit inflammation, promote angiogenesis, and inhibit cardiomyocyte death. In addition, PEV have potential as biocompatible and autologous drug carriers. Therefore, better research on the mechanisms how PEV act during cardiovascular disease and could be implemented as a therapeutic will provide new perspectives for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Xin Xin
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
3
|
Xie X, Xiang J, Zhao H, Tong B, Zhang L, Kang X, Kong S, Wang T, Cao W. Integrative Quantitative Analysis of Platelet Proteome and Site-Specific Glycoproteome Reveals Diagnostic Potential of Platelet Glycoproteins for Liver Cancer. Anal Chem 2025. [PMID: 39813102 DOI: 10.1021/acs.analchem.4c03855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
The role of peripheral blood platelets as indicators of cancer progression is increasingly recognized, and the significance of abnormal glycosylation in platelet function and related disorders is gaining attention. However, the potential of platelets as a source of protein site-specific glycosylation for cancer diagnosis remains underexplored. In this study, we proposed a general pipeline that integrates quantitative proteomics with site-specific glycoproteomics, allowing for an in-depth investigation of the platelet glycoproteome. With this pipeline, we generated a data set comprising 3,466 proteins with qualitative information, 3,199 proteins with quantitative information, 3,419 site-specific glycans with qualitative information and 3,377 site-specific glycans with quantitative information from peripheral blood platelets of hepatocellular carcinoma (HCC) patients, metastatic liver cancer (mLC) patients, and healthy controls. The integrated analysis revealed significant changes in platelet protein N-glycosylation in liver cancer patients. Further systems biology analysis and lectin pull-down-coupled ELISA assays in independent clinical samples confirmed two N-glycoproteins with specific glycan types, complement C3 (C3) with oligomannose modification and integrin β-3 (ITGB3) with sialylation, as potential biomarkers distinguishing liver cancer patients from healthy individuals, without differentiating between HCC and mLC patient group. These findings highlight the potential of platelet protein glycosylation as biomarkers.
Collapse
Affiliation(s)
- Xiaofeng Xie
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, China
| | - Jianfeng Xiang
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Huanhuan Zhao
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, China
| | - Bingrun Tong
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lei Zhang
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, China
| | - Xiaonan Kang
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Siyuan Kong
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, China
| | - Tao Wang
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Weiqian Cao
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, China
| |
Collapse
|
4
|
Weiss L, Macleod H, Maguire PB. Platelet-derived extracellular vesicles in cardiovascular disease and treatment - from maintaining homeostasis to targeted drug delivery. Curr Opin Hematol 2025; 32:4-13. [PMID: 39377239 PMCID: PMC11620325 DOI: 10.1097/moh.0000000000000845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
PURPOSE OF REVIEW Cardiovascular disease (CVD) remains a major global health burden. Rising incidences necessitate improved understanding of the pathophysiological processes underlying disease progression to foster the development of novel therapeutic strategies. Besides their well recognized role in CVD, platelet-derived extracellular vesicles (PEVs) mediate inter-organ cross talk and contribute to various inflammatory diseases. RECENT FINDINGS PEVs are readily accessible diagnostic biomarkers that mirror pathophysiological disease progression but also may confer cardioprotective properties. Monitoring the effects of modulation of PEV signatures through pharmacotherapies has also provided novel insights into treatment efficacy. Furthermore, exploiting their inherent ability to infiltrate thrombi, atherosclerotic plaques and solid tumours, PEVs as well as platelet-membrane coated nanoparticles are emerging as novel effective and targeted treatment options for CVD and cancer. SUMMARY Collectively, in-depth characterization of PEVs in various diseases ultimately enhances their use as diagnostic or prognostic biomarkers and potential therapeutic targets, making them clinically relevant candidates to positively impact patient outcomes.
Collapse
Affiliation(s)
- Luisa Weiss
- Conway SPHERE Research Group, Conway Institute
- School of Biomolecular and Biomedical Science
- AI for Healthcare Hub, Institute for Discovery, O’Brien Centre of Science, University College Dublin, Dublin, Ireland
| | - Hayley Macleod
- Conway SPHERE Research Group, Conway Institute
- School of Biomolecular and Biomedical Science
| | - Patricia B. Maguire
- Conway SPHERE Research Group, Conway Institute
- School of Biomolecular and Biomedical Science
- AI for Healthcare Hub, Institute for Discovery, O’Brien Centre of Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
5
|
Yao W, Zhao K, Li X. Platelet stimulation-regulated expression of ILK and ITGB3 contributes to intrahepatic cholangiocarcinoma progression through FAK/PI3K/AKT pathway activation. Cell Mol Life Sci 2024; 82:19. [PMID: 39725790 DOI: 10.1007/s00018-024-05526-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Intrahepatic cholangiocarcinoma (iCCA) is a highly lethal hepatobiliary malignancy with an increasing incidence annually. Extensive research has elucidated the existence of a reciprocal interaction between platelets and cancer cells, which promotes tumor proliferation and metastasis. This study aims to investigate the function and mechanism underlying iCCA progression driven by the interplay between platelets and tumor cells, aiming to provide novel therapeutic strategies for iCCA. METHODS The associations between platelets and cancer development were investigated by analyzing the peripheral blood platelet count, degree of platelet activation and infiltration in the microenvironment of patients with iCCA. By co-culturing tumor cells with platelets, the influence of platelet stimulation on the epithelial-mesenchymal transition (EMT), proliferation, and metastasis of iCCA cells was assessed through in vitro and in vivo experiments. Quantitative proteomic profiling was conducted to identify key downstream targets that were altered in tumor cells following platelet stimulation. The RNA interference technique was utilized to investigate the impacts of gene silencing on the malignant biological behaviors of tumor cells. RESULTS Compared with healthy adults, patients with iCCA presented significantly higher levels of peripheral blood platelet counts, platelet activation and infiltration degrees, which were also found to be correlated with patient prognosis. Platelet stimulation greatly facilitated the EMT of iCCA cells, leading to enhanced proliferative and metastatic capabilities. Mechanistically, proteomic profiling identified a total of 67 up-regulated and 40 down-regulated proteins in iCCA cells co-cultured with platelets. Among these proteins, two elevated targets ILK and ITGB3, were further demonstrated to be partially responsible for platelet-induced iCCA progression, which might depend on their regulatory effects on FAK/PI3K/AKT signaling transduction. CONCLUSIONS Our data revealed that platelet-related indices were abnormally ascendant in iCCA patients compared to healthy adults. Co-culturing with platelets enhanced the progression of EMT, and the motility and viability of iCCA cells in vitro and in vivo. Proteomic profiling discovered that platelets promoted the development of iCCA through FAK/PI3K/AKT pathway by means of elevating the expression of ILK and ITGB3, indicating that both proteins are promising therapeutic targets for iCCA with the guidance of platelet-related indices.
Collapse
Affiliation(s)
- Wei Yao
- Department of Oncology Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Kai Zhao
- Department of Biliary and Pancreatic Surgery, Cancer Research Center Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Xiangyu Li
- Department of Thoracic Surgery Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
6
|
Tavukcuoglu Z, Butt U, de Faria AVS, Oesterreicher J, Holnthoner W, Laitinen S, Palviainen M, Siljander PRM. Platelet-derived extracellular vesicles induced through different activation pathways drive melanoma progression by functional and transcriptional changes. Cell Commun Signal 2024; 22:601. [PMID: 39695652 DOI: 10.1186/s12964-024-01973-4] [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: 10/24/2024] [Accepted: 11/30/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Beyond their conventional roles in hemostasis and wound healing, platelets have been shown to facilitate hematogenous metastasis by interacting with cancer cells. Depending on the activation route, platelets also generate different platelet-derived extracellular vesicles (PEVs) that may educate cancer cells in the circulation or within the tumor microenvironment. We engaged different platelet-activating receptors, including glycoprotein VI and C-type lectin-like receptor 2, to generate a spectrum of PEV types. This allowed us to investigate the differential capacity of PEVs to alter cancer hallmark functions such as proliferation, invasion, and pro-angiogenic potential using melanoma as a model. Additionally, we analyzed changes in the cell transcriptomes and cancer EV profiles. METHODS Two human melanoma cell lines (MV3 and A2058) with differential metastatic potential were studied in the 3D spheroid cultures. Human platelets were activated with collagen related peptide (CRP), fucoidan from Fucus vesiculosus (FFV), thrombin & collagen co-stimulus and Ca2+ ionophore, and PEVs were isolated by size-exclusion chromatography followed by ultrafiltration. Spheroids or cells were treated with PEVs and used in functional assays of proliferation, invasion, and endothelial tube formation as well as for the analysis of cancer EV production and their tetraspanin profiles. Differentially expressed genes and enriched signaling pathways in the PEV-treated spheroids were analyzed at 6 h and 24 h by RNA sequencing. RESULTS Among the studied PEVs, those generated by CRP and FFV exhibited the most pronounced effects on altering cancer hallmark functions. Specifically, CRP and FFV PEVs increased proliferation in both MV3 and A2058 spheroids. Distinct tetraspanin signatures of melanoma EVs were induced by all PEV types. While the PI3K-Akt and MAPK signaling pathways were activated by both CRP and FFV PEVs, they differently upregulated the immunomodulatory TGF-β and type-I interferon signaling pathways, respectively. CONCLUSIONS Our study revealed both shared and distinct, cancer-promoting functions of PEVs, which contributed to the transcriptome and metastatic capabilities of the melanoma spheroids. Inhibiting the platelet receptors that modulate the PEVs' cancer-promoting properties may open up new strategies for identifying promising treatment targets for cancer therapy.
Collapse
Affiliation(s)
- Zeynep Tavukcuoglu
- EV group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland
| | - Umar Butt
- EV group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland
| | - Alessandra V Sousa de Faria
- EV group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | - Wolfgang Holnthoner
- AUVA Research Centre, Ludwig Boltzmann Institute for Traumatology, Vienna, Austria
| | - Saara Laitinen
- Finnish Red Cross Blood Service (FRCBS), Helsinki, Finland
| | - Mari Palviainen
- EV group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland
- EV Core, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Pia R-M Siljander
- EV group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland.
- EV Core, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
7
|
Benariba MA, Hannachi K, Zhu S, Zhang Y, Wang X, Zhou N. A liposome-based assay for cancer biomarker detection: exploring the correlation between platelet-derived microvesicles and NSCLC-associated miRNAs. NANOSCALE 2024; 16:22037-22046. [PMID: 39527124 DOI: 10.1039/d4nr03704a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Advances in molecular biology have enabled the identification of numerous cancer biomarkers, offering the potential to improve the diagnosis and prognosis of cancer. In non-small cell lung cancer (NSCLC), the role of platelet-derived microvesicles (PMVs) in cancer progression has received limited attention. While previous studies have focused on the increase of extracellular vesicles in plasma and their interaction with cancer, the expression of microRNAs (miRNAs) delivered through PMVs following platelet activation has remained largely unexplored. This study fills this knowledge gap by investigating miRNA expression in PMVs isolated from healthy donors and NSCLC patients following calcium treatment, a known platelet activator. A significant correlation was found between PMV levels and the expression of specific miRNAs; specifically, miRNA-21 expression increased 7.89 ± 0.44-fold in NSCLC patients and 7.12 ± 0.49-fold in healthy donors after calcium treatment. These findings highlight the potential of PMVs and their miRNA cargo to serve as specific biomarkers for NSCLC, offering valuable insights into cancer diagnosis and prognosis. To facilitate the sensitive detection of these miRNAs, a novel carboxyfluorescein (CF)-loaded liposome-based assay was developed. This assay demonstrated enhanced sensitivity, achieving a detection limit of 1.03 pg mL-1, when combined with a calcium platelet-activation approach. This research has the potential to lead to the development of innovative diagnostic tools and therapeutic strategies, ultimately improving outcomes for patients with NSCLC and other cancers.
Collapse
Affiliation(s)
- Mohamed Aimene Benariba
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
- Bioengineering Laboratory, Ecole Nationale Supérieure de Biotechnologie, Ville Universitaire Ali Mendjeli, BP E66 25100, Constantine, Algeria
| | - Kanza Hannachi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Sha Zhu
- Department of Urology, the Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, Wuxi 214002, China.
| | - Yuting Zhang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Xiaoli Wang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Nandi Zhou
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
8
|
Fan Z, Gan Y, Hu Y. The potential utilization of platelet-derived extracellular vesicles in clinical treatment. Platelets 2024; 35:2397592. [PMID: 39287127 DOI: 10.1080/09537104.2024.2397592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/09/2024] [Accepted: 08/02/2024] [Indexed: 09/19/2024]
Abstract
Platelet-derived extracellular vesicles (PEVs) are released by platelets in the blood circulation, which carry a rich bio-molecular cargo influential in intercellular communications. PEVs can enter the lymph, bone marrow, and synovial fluid as nano-sized particles, while platelets cannot cross tissue barriers. Considering the advantages of PEVs such as low immunogenicity, high regulation of signal transduction, and easy obtainment, PEVs may be promising therapeutic tools for medical applications. The exceptional functional roles played by PEVs explain the recent interest in exploring new cell-free therapies that could address needs in angiogenesis, regenerative medicine, and targeted drug delivery. The review takes a critical look at the main advances of PEVs in the treatment of diseases by presenting the latest knowledge from the performed studies, in order to enhance the further translation of the PEVs research into feasible therapeutic applications.
Collapse
Affiliation(s)
- Zhijia Fan
- Department of Laboratory Medicine, Beijing Chaoyang Hospital, Beijing Center for Clinical Laboratories, The Third Clinical Medical College of Capital Medical University, Beijing, PR China
| | - Yixiao Gan
- Department of Transfusion Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Yanwei Hu
- Department of Laboratory Medicine, Beijing Chaoyang Hospital, Beijing Center for Clinical Laboratories, The Third Clinical Medical College of Capital Medical University, Beijing, PR China
| |
Collapse
|
9
|
Duret T, Elmallah M, Rollin J, Gatault P, Jiang LH, Roger S. Role of purinoreceptors in the release of extracellular vesicles and consequences on immune response and cancer progression. Biomed J 2024:100805. [PMID: 39510381 DOI: 10.1016/j.bj.2024.100805] [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: 09/04/2024] [Revised: 10/24/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024] Open
Abstract
Cell-to-cell communication is a major process for accommodating cell functioning to changes in the environments and to preserve tissue and organism homeostasis. It is achieved by different mechanisms characterized by the origin of the message, the molecular nature of the messenger, its speed of action and its reach. Purinergic signalling is a powerful mechanism initiated by extracellular nucleotides, such as ATP, acting on plasma membrane purinoreceptors. Purinergic signalling is tightly controlled in time and space by the action of ectonucleotidases. Recent studies have highlighted the critical role of purinergic signalling in controlling the generation, release and fate of extracellular vesicles and, in this way, mediating long-distance responses. Most of these discoveries have been made in immune and cancer cells. This review is aimed at establishing the current knowledge on the way which purinoreceptors control extracellular vesicle-mediated communications and consequences for recipient cells.
Collapse
Affiliation(s)
- Thomat Duret
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France; Fédération Hospitalo-Universitaire Survival optimization in organ Transplantation (FHU SUPORT), Tours, France
| | - Mohammed Elmallah
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France
| | - Jérôme Rollin
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France; Service d'Hématologie-Hémostase, CHRU de Tours, Tours, France
| | - Philippe Gatault
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France; Service de Néphrologie, Hypertension, Dialyse et Transplantation Rénale, CHRU Tours, Tours, France; Fédération Hospitalo-Universitaire Survival optimization in organ Transplantation (FHU SUPORT), Tours, France
| | - Lin-Hua Jiang
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France; School of Basic Medical Sciences, Xinxiang Medical University, China; School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Sébastien Roger
- Université de Tours, Inserm UMR1327 ISCHEMIA « Membrane Signalling and Inflammation in Reperfusion Injuries », Tours, France; Fédération Hospitalo-Universitaire Survival optimization in organ Transplantation (FHU SUPORT), Tours, France.
| |
Collapse
|
10
|
Das K, Rao LVM. Coagulation protease-induced extracellular vesicles: their potential effects on coagulation and inflammation. J Thromb Haemost 2024; 22:2976-2990. [PMID: 39127325 PMCID: PMC11726980 DOI: 10.1016/j.jtha.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 08/12/2024]
Abstract
Coagulation proteases, in addition to playing an essential role in blood coagulation, often influence diverse cellular functions by inducing specific signaling pathways via the activation of protease-activated receptors (PARs). PAR activation-induced cellular effects are known to be cell-specific as PARs are expressed selectively in specific cell types. However, a growing body of evidence indicates that coagulation protease-induced PAR activation in a specific cell type could affect cellular responses in other cell types via communicating through extracellular vesicles (EVs) as coagulation protease-induced PAR signaling could promote the release of EVs in various cell types. EVs are membrane-enclosed nanosized vesicles that facilitate intercellular communication by transferring bioactive molecules, such as proteins, lipids, messenger RNAs, and microRNAs, etc., from donor cells to recipient cells. Our recent findings established that factor (F)VIIa promotes the release of EVs from vascular endothelium via endothelial cell protein C receptor-dependent activation of PAR1-mediated biased signaling. FVIIa-released EVs exhibit procoagulant activity and cytoprotective responses in both in vitro and in vivo model systems. This review discusses how FVIIa and other coagulation proteases trigger the release of EVs. The review specifically discusses how FVIIa-released EVs are enriched with phosphatidylserine and anti-inflammatory microRNAs and the impact of FVIIa-released EVs on hemostasis in therapeutic settings. The review also briefly highlights the therapeutic potential of FVIIa-released EVs in treating bleeding and inflammatory disorders, such as hemophilic arthropathy.
Collapse
Affiliation(s)
- Kaushik Das
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas at Tyler School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, Texas, USA.
| |
Collapse
|
11
|
Wen K, Lin Z, Tan H, Han M. Correlations between coagulation abnormalities and inflammatory markers in trauma-induced coagulopathy. Front Physiol 2024; 15:1474707. [PMID: 39539951 PMCID: PMC11557354 DOI: 10.3389/fphys.2024.1474707] [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: 08/02/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction In multiple trauma patients, the occurrence of trauma-induced coagulopathy (TIC) is closely associated with tissue damage and coagulation function abnormalities in the pathophysiological process. Methods This study established a multiple trauma and shock model in Sprague-Dawley (SD) rats and comprehensively utilized histological staining and radiographic imaging techniques to observe injuries in the intestine, liver, skeletal muscles, and bones. Monitoring activated partial thromboplastin time (APTT), platelet (PLT) count, respiratory rate, blood pressure, and other physiological indicators revealed time-dependent alterations in coagulation function and physiological indicators. Enzyme-linked immunosorbent assay (ELISA) measurements of inflammatory factors Tumor Necrosis Factor-alpha (TNF-α), Interleukin-6 (IL-6) and vascular endothelial injury marker (Syndecan-1) were also conducted. Results Experimental results demonstrated significant changes in tissue structure after multiple traumas, although widespread necrosis or hemorrhagic lesions were not observed. There were time-dependent alterations in coagulation function and physiological indicators. ELISA measurements showed a strong positive correlation between the significant decrease in PLT count and the increase in TNF-α and IL-6 concentrations. Discussion The study provides crucial information for the early diagnosis and treatment of TIC. The findings suggest that structured monitoring of coagulation and inflammatory indicators can help in understanding the pathophysiological changes and aid in the management of TIC in multiple trauma patients.
Collapse
Affiliation(s)
- Ke Wen
- Department of Hand and Microsurgery, Taihe Hospital, Affiliated Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | - Zhexuan Lin
- Bio-Analytical Laboratory of Shantou University Medical College, Shantou, China
| | - Haizhu Tan
- Department of Preventive Medicine, Shantou University Medical College, Shantou, China
| | - Ming Han
- Emergency Department of Shenzhen University General Hospital, Shenzhen, China
| |
Collapse
|
12
|
Duttaroy AK. Functional Foods in Preventing Human Blood Platelet Hyperactivity-Mediated Diseases-An Updated Review. Nutrients 2024; 16:3717. [PMID: 39519549 PMCID: PMC11547462 DOI: 10.3390/nu16213717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Backgrounds/Objectives: Abnormal platelet functions are associated with human morbidity and mortality. Platelets have emerged as critical regulators of numerous physiological and pathological processes beyond their established roles in hemostasis and thrombosis. Maintaining physiological platelet function is essential to hemostasis and preventing platelet-associated diseases such as cardiovascular disease, cancer metastasis, immune disorders, hypertension, diabetes, sickle cell disease, inflammatory bowel disease, sepsis, rheumatoid arthritis, myeloproliferative disease, and Alzheimer's disease. Platelets become hyperactive in obesity, diabetes, a sedentary lifestyle, hypertension, pollution, and smokers. Platelets, upon activation, can trawl leukocytes and progenitor cells to the vascular sites. Platelets release various proinflammatory, anti-inflammatory, and angiogenic factors and shed microparticles in the circulation, thus promoting pathological reactions. These platelet-released factors also maintain sustained activation, further impacting these disease processes. Although the mechanisms are unknown, multiple stimuli induce platelet hyperreactivity but involve the early pathways of platelet activation. The exact mechanisms of how hyperactive platelets contribute to these diseases are still unclear, and antiplatelet strategies are inevitable for preventing these diseases. Reducing platelet function during the early stages could significantly impact these diseases. However, while this is potentially a worthwhile intervention, using antiplatelet drugs to limit platelet function in apparently healthy individuals without cardiovascular disease is not recommended due to the increased risk of internal bleeding, resistance, and other side effects. The challenge for therapeutic intervention in these diseases is identifying factors that preferentially block specific targets involved in platelets' complex contribution to these diseases while leaving their hemostatic function at least partially intact. Since antiplatelet drugs such as aspirin are not recommended as primary preventives, it is essential to use alternative safe platelet inhibitors without side effects. METHODS A systematic search of the PUBMED database from 2000 to 2023 was conducted using the selected keywords: "functional foods", "polyphenols", "fatty acids", "herbs", fruits and vegetables", "cardioprotective agents", "plant", "platelet aggregation", "platelet activation", "clinical and non-clinical trial", "randomized", and "controlled". RESULTS Potent natural antiplatelet factors have been described, including omega-3 fatty acids, polyphenols, and other phytochemicals. Antiplatelet bioactive compounds in food that can prevent platelet hyperactivity and thus may prevent several platelet-mediated diseases, including cardiovascular disease. CONCLUSIONS This narrative review describes the work during 2000-2023 in developing functional foods from natural sources with antiplatelet effects.
Collapse
Affiliation(s)
- Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0313 Oslo, Norway
| |
Collapse
|
13
|
He X, Xiang Y, Lin C, Shen W. Development and validation of an inflammation-nutrition indices-based nomogram for predicting early recurrence in patients with stage IB lung adenocarcinoma. Sci Rep 2024; 14:25111. [PMID: 39443648 PMCID: PMC11500178 DOI: 10.1038/s41598-024-76230-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
To explore the inflammation-nutrition indices and related clinical factors affecting early recurrence in patients with stage IB LUAD. A retrospective analysis was conducted on clinical and pathological data of patients diagnosed with stage IB LUAD who underwent radical surgery in our hospital from January 2016 to January 2021. Using R software, patients were randomly divided into training (n = 140) and validation (n = 59) cohorts in a 7:3 ratio. Univariate and multivariate Cox regression analyses were performed to identify risk factors for RFS and construct a predictive model. The performance of the model was evaluated using the area under the receiver operating characteristic curve (AUC), concordance index (C-index), and calibration curve. Clinical utility of the model was assessed using decision curve analysis (DCA). Multivariate Cox regression analysis revealed that vascular invasion, visceral pleural invasion, predominant pattern, preoperative NLR > 2.33, preoperative PLR > 127.62, and preoperative PNI ≤ 48.3 were independent risk factors for RFS. The C-index of the nomogram model constructed based on these independent risk factors was 0.825 (95% CI: 0.762-0.881) in the training cohort and 0.772 (95% CI: 0.667-0.876) in the validation cohort. The ROC curves showed AUCs of 0.902, 0.881, and 0.877 for 1-year, 2-year, and 3-year RFS in the training cohort and AUCs of 0.782, 0.825, and 0.732 in the validation cohort respectively. Calibration curve and decision curve analysis indicated good clinical value of the model. The nomogram model based on inflammation-nutrition indices has predictive value for early recurrence in patients with stage IB LUAD.
Collapse
Affiliation(s)
- Xianneng He
- Department of Thoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, 315000, China
- Health Science Center, Ningbo University, Ningbo, 315000, China
| | - Yishun Xiang
- Department of Thoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, 315000, China
- Health Science Center, Ningbo University, Ningbo, 315000, China
| | - Chengbin Lin
- Department of Thoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, 315000, China
| | - Weiyu Shen
- Department of Thoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, 315000, China.
- Health Science Center, Ningbo University, Ningbo, 315000, China.
| |
Collapse
|
14
|
Palviainen M, Puutio J, Østergaard RH, Eble JA, Maaninka K, Butt U, Ndika J, Kari OK, Kamali‐Moghaddam M, Kjaer‐Sorensen K, Oxvig C, Aransay AM, Falcon‐Perez JM, Federico A, Greco D, Laitinen S, Hayashi Y, Siljander PR. Beyond basic characterization and omics: Immunomodulatory roles of platelet-derived extracellular vesicles unveiled by functional testing. J Extracell Vesicles 2024; 13:e12513. [PMID: 39330919 PMCID: PMC11428872 DOI: 10.1002/jev2.12513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
Renowned for their role in haemostasis and thrombosis, platelets are also increasingly recognized for their contribution in innate immunity, immunothrombosis and inflammatory diseases. Platelets express a wide range of receptors, which allows them to reach a variety of activation endpoints and grants them immunomodulatory functions. Activated platelets release extracellular vesicles (PEVs), whose formation and molecular cargo has been shown to depend on receptor-mediated activation and environmental cues. This study compared the immunomodulatory profiles of PEVs generated via activation of platelets by different receptors, glycoprotein VI, C-type lectin-like receptor 2 and combining all thrombin-collagen receptors. Functional assays in vivo in zebrafish and in vitro in human macrophages highlighted distinct homing and secretory responses triggered by the PEVs. In contrast, omics analyses of protein and miRNA cargo combined with physicochemical particle characterization found only subtle differences between the activated PEV types, which were insufficient to predict their different immunomodulatory functions. In contrast, constitutively released PEVs, formed in the absence of an exogenous activator, displayed a distinct immunomodulatory profile from the receptor-induced PEVs. Our findings underscore that PEVs are tunable through receptor-mediated activation. To truly comprehend their role(s) in mediating platelet functions among immune cells, conducting functional assays is imperative.
Collapse
Affiliation(s)
- Mari Palviainen
- EV Group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
- EV Core, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | - Johanna Puutio
- EV Group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | | | - Johannes A. Eble
- Institute of Physiological Chemistry and PathobiochemistryUniversity of MünsterMünsterGermany
| | - Katariina Maaninka
- EV Group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Umar Butt
- EV Group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Joseph Ndika
- Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Otto K. Kari
- Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Masood Kamali‐Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life LaboratoryUppsala UniversityUppsalaSweden
| | | | - Claus Oxvig
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Ana M. Aransay
- Genome Analysis Platform, Center for Cooperative Research in BiosciencesBasque Research and Technology Alliance (BRTA)MendaroSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd)MadridSpain
| | - Juan M. Falcon‐Perez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd)MadridSpain
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)DerioSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
| | - Antonio Federico
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE); Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Division of Pharmaceutical Biosciences, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE); Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Division of Pharmaceutical Biosciences, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Saara Laitinen
- Research and DevelopmentFinnish Red Cross Blood Service (FRCBS)HelsinkiFinland
| | - Yuya Hayashi
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- Interdisciplinary Nanoscience Center (iNANO)Aarhus UniversityAarhusDenmark
| | - Pia R.‐M. Siljander
- EV Group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
- EV Core, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
15
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
16
|
Dong J, Jiang W, Zhang W, Guo T, Yang Y, Jiang X, Zheng L, Du T. Exploring the J-shaped relationship between HALP score and mortality in cancer patients: A NHANES 1999-2018 cohort study. Front Oncol 2024; 14:1388610. [PMID: 39301556 PMCID: PMC11410770 DOI: 10.3389/fonc.2024.1388610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Background The recent hemoglobin, albumin, lymphocyte, and platelet (HALP) scores, combined with various clinically available indicators, can comprehensively evaluate the nutritional and immune status of patients. Some observational studies have found a positive correlation between HALP score and cancer prognosis, but the clinical application of HALP score has raised concerns due to the presence of confounding factors. The aim of this study is to explore the relationship between HALP score and long-term mortality in cancer patients. Methods We extracted 3832 cancer patients with complete baseline information from the National Health and Nutrition Examination Survey (NHANES). The COX regressions and restricted cubic spline (RCS) curves were used to explore the nonlinear relationship between HALP score and long-term mortality risk in cancer patients. Kaplan-Meier (K-M) curve was conducted to evaluate the impact of HALP score on long-term mortality risk. Additionally, subgroup analysis was conducted to verify the stability of the above results. Results We divided participants into 4 groups based on HALP score, and the COX regression results showed that risk of long-term mortality tended to be lower in cancer patients with high HALP scores. Meanwhile, the RCS curves showed that there was a nonlinear association. The results remained stable in subgroup analyses and in breast cancer, colorectal cancer, cervix and uterus cancer, melanoma, prostate cancer and skin cancer. Conclusions HALP score were independently associated with the risk of long-term mortality in cancer patients, and there is also a non-linear association. This will provide new perspectives on clinical and nutritional interventions for cancer patients.
Collapse
Affiliation(s)
- Jiaxing Dong
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wanju Jiang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenjia Zhang
- Department of Respiratory Medicine, Shanghai Tenth Peoples Hospital, Tongji University, Shanghai, China
| | - Taohua Guo
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yucheng Yang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaohua Jiang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liang Zheng
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tao Du
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
17
|
Liu H, Mao H, Ouyang X, Lu R, Li L. Intercellular Mitochondrial Transfer: The Novel Therapeutic Mechanism for Diseases. Traffic 2024; 25:e12951. [PMID: 39238078 DOI: 10.1111/tra.12951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 09/07/2024]
Abstract
Mitochondria, the dynamic organelles responsible for energy production and cellular metabolism, have the metabolic function of extracting energy from nutrients and synthesizing crucial metabolites. Nevertheless, recent research unveils that intercellular mitochondrial transfer by tunneling nanotubes, tumor microtubes, gap junction intercellular communication, extracellular vesicles, endocytosis and cell fusion may regulate mitochondrial function within recipient cells, potentially contributing to disease treatment, such as nonalcoholic steatohepatitis, glioblastoma, ischemic stroke, bladder cancer and neurodegenerative diseases. This review introduces the principal approaches to intercellular mitochondrial transfer and examines its role in various diseases. Furthermore, we provide a comprehensive overview of the inhibitors and activators of intercellular mitochondrial transfer, offering a unique perspective to illustrate the relationship between intercellular mitochondrial transfer and diseases.
Collapse
Affiliation(s)
- Huimei Liu
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Hui Mao
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Xueqian Ouyang
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Ruirui Lu
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
18
|
Song MY, Zhao L, Huang WJ, Cui MM, Liu YX, Wang RT, Zhang X. Preoperative platelet distribution width predicts bone metastasis in patients with breast cancer. BMC Cancer 2024; 24:1066. [PMID: 39210343 PMCID: PMC11360324 DOI: 10.1186/s12885-024-12837-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
PURPOSE Bone metastases occur in 50-70% of patients with breast cancer (BC) and result in high mortality. Platelet distribution width (PDW), a commonly used parameter of activated platelets, has been associated with a poor prognosis in BC. We aim to investigate the prognostic role of PDW for bone metastasis in BC patients. METHODS 515 patients who received BC surgery in the Harbin Medical University Cancer Hospital from July 1, 2016, to December 31, 2017, were reviewed. Patients' characteristics and platelet indices upon enrollment in this study were collected. The Kaplan-Meier method was used to estimate the 5-year bone metastasis incidence. The univariate and multivariate Cox regression analyses were utilized to identify risk factors associated with bone metastasis. RESULTS The patients with bone metastases exhibited lower PDW levels than the patients without bone metastases. Moreover, decreased PDW was significantly correlated with histologic type, multifocal disease, and lymph node status. In addition, the patients with reduced PDW levels were more likely to develop bone metastasis. Multivariate analysis showed that PDW was an independent predictor for bone metastasis. CONCLUSION PDW is an independent predictor of bone metastasis in BC. Further research is warranted.
Collapse
Affiliation(s)
- Mei-Yue Song
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Lin Zhao
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, NO.150 Haping ST, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Wen-Juan Huang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, NO.150 Haping ST, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Ming-Ming Cui
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, NO.150 Haping ST, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Yu-Xi Liu
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, NO.150 Haping ST, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Rui-Tao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, NO.150 Haping ST, Nangang District, Harbin, 150081, Heilongjiang, China.
| | - Xin Zhang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, NO.150 Haping ST, Nangang District, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
19
|
Zhuang T, Wang S, Yu X, He X, Guo H, Ou C. Current status and future perspectives of platelet-derived extracellular vesicles in cancer diagnosis and treatment. Biomark Res 2024; 12:88. [PMID: 39183323 PMCID: PMC11346179 DOI: 10.1186/s40364-024-00639-0] [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: 06/29/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
Platelets are a significant component of the cell population in the tumour microenvironment (TME). Platelets influence other immune cells and perform cross-talk with tumour cells, playing an important role in tumour development. Extracellular vesicles (EVs) are small membrane vesicles released from the cells into the TME. They can transfer biological information, including proteins, nucleic acids, and metabolites, from secretory cells to target receptor cells. This process affects the progression of various human diseases, particularly cancer. In recent years, several studies have demonstrated that platelet-derived extracellular vesicles (PEVs) can help regulate the malignant biological behaviours of tumours, including malignant proliferation, resistance to cell death, invasion and metastasis, metabolic reprogramming, immunity, and angiogenesis. Consequently, PEVs have been identified as key regulators of tumour progression. Therefore, targeting PEVs is a potential strategy for tumour treatment. Furthermore, the extensive use of nanomaterials in medical research has indicated that engineered PEVs are ideal delivery systems for therapeutic drugs. Recent studies have demonstrated that PEV engineering technologies play a pivotal role in the treatment of tumours by combining photothermal therapy, immunotherapy, and chemotherapy. In addition, aberrant changes in PEVs are closely associated with the clinicopathological features of patients with tumours, which may serve as liquid biopsy markers for early diagnosis, monitoring disease progression, and the prognostic assessment of patients with tumours. A comprehensive investigation into the role and potential mechanisms of PEVs in tumourigenesis may provide novel diagnostic biomarkers and potential therapeutic strategies for treating human tumours.
Collapse
Affiliation(s)
- Tongtao Zhuang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shenrong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoqian Yu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hongbin Guo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
20
|
Miao L, Yang Y, Cheng M, Chen L, Han C. Ginsenoside Rb prevents the metastasis of hepatocarcinoma by blocking the platelet-tumor cell interaction. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03387-y. [PMID: 39172150 DOI: 10.1007/s00210-024-03387-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/15/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND The interaction between platelets and tumor cells is a crucial step in the progression of tumor metastasis. Blocking platelet-tumor cell interaction is a potential target against metastasis. Ginsenoside Rb (G-Rb) exhibits potential anti-tumor pharmacological properties and may offer a therapeutic option for cancer. PURPOSE This study aimed to investigate anti-metastatic effects of G-Rb through regulating the crosstalk of platelets with tumor cells. METHODS In order to explore anti-metastatic effects of G-Rb in vitro, HepG2 cell and platelets were co-cultured to mimic the interaction of platelets with tumor cells. Wound healing and Transwell assays were used to assess the effect of G-Rb on cell migration and invasion. The expression of epithelial-mesenchymal transition (EMT)-related markers was determined by RT-qPCR and western blot assays. The aggregation and activation of platelets were detected by flow cytometry. Moreover, a lung metastasis model of mice was established to evaluate inhibitory effects of G-Rb in vivo. Metastatic nodules on the lung surface were counted and sections of lung tissues were stained by H&E. RESULTS G-Rb effectively suppressed tumor metastasis in the co-culture of platelets with HepG2 cell. First, G-Rb treatment significantly inhibited the migration and invasion of HepG2 cells induced by platelets. Second, the expressions of EMT-related markers, including N-cadherin, Snail, and MMP9, were decreased by the treatment of G-Rb in the presence of platelets. Meanwhile, G-Rb also suppressed platelet hyperactivity by regulating the adhesion to tumor cells, activation, TCIPA, and TGF-β1 secretion of platelets in vitro. In addition, the results of in vivo experiments proved G-Rb administration not only significantly decreased lung metastasis but also attenuated platelets aberrant aggregation and activation in vivo. CONCLUSION Our findings showed that G-Rb inhibited tumor metastasis and platelet activation through mediating platelet-tumor cell interaction, indicating the potential values of G-Rb in tumor metastasis therapy.
Collapse
Affiliation(s)
- Longxing Miao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People's Republic of China
| | - Yijun Yang
- Department of Pharmacy, Shandong Medical College, Jinan, 250002, People's Republic of China
| | - Mengtao Cheng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
| | - Lijing Chen
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China
- The Second Affiliated Hospital of Shandong, University of Traditional Chinese Medicine, Jinan, 250000, People's Republic of China
| | - Chunchao Han
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People's Republic of China.
| |
Collapse
|
21
|
Muttiah B, Ng SL, Lokanathan Y, Ng MH, Law JX. Beyond Blood Clotting: The Many Roles of Platelet-Derived Extracellular Vesicles. Biomedicines 2024; 12:1850. [PMID: 39200314 PMCID: PMC11351396 DOI: 10.3390/biomedicines12081850] [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: 05/30/2024] [Revised: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Platelet-derived extracellular vesicles (pEVs) are emerging as pivotal players in numerous physiological and pathological processes, extending beyond their traditional roles in hemostasis and thrombosis. As one of the most abundant vesicle types in human blood, pEVs transport a diverse array of bioactive molecules, including growth factors, cytokines, and clotting factors, facilitating crucial intercellular communication, immune regulation, and tissue healing. The unique ability of pEVs to traverse tissue barriers and their biocompatibility position them as promising candidates for targeted drug delivery and regenerative medicine applications. Recent studies have underscored their involvement in cancer progression, viral infections, wound healing, osteoarthritis, sepsis, cardiovascular diseases, rheumatoid arthritis, and atherothrombosis. For instance, pEVs promote tumor progression and metastasis, enhance tissue repair, and contribute to thrombo-inflammation in diseases such as COVID-19. Despite their potential, challenges remain, including the need for standardized isolation techniques and a comprehensive understanding of their mechanisms of action. Current research efforts are focused on leveraging pEVs for innovative anti-cancer treatments, advanced drug delivery systems, regenerative therapies, and as biomarkers for disease diagnosis and monitoring. This review highlights the necessity of overcoming technical hurdles, refining isolation methods, and establishing standardized protocols to fully unlock the therapeutic potential of pEVs. By understanding the diverse functions and applications of pEVs, we can advance their use in clinical settings, ultimately revolutionizing treatment strategies across various medical fields and improving patient outcomes.
Collapse
Affiliation(s)
- Barathan Muttiah
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Sook Luan Ng
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Min Hwei Ng
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| |
Collapse
|
22
|
Yuan X, Liu P, Xu L, Liang L, Dong Q, Fan T, Yue W, Qu M, Pei X, Xie X. miR-1915-3p regulates megakaryocytic and erythroid differentiation by targeting SOCS4. Thromb J 2024; 22:74. [PMID: 39123189 PMCID: PMC11316338 DOI: 10.1186/s12959-024-00615-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/13/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Proper control of the lineage bias of megakaryocytic and erythroid progenitor cells (MEPs) is of significant importance, the disorder of which will lead to abnormalities in the number and function of platelets and erythrocytes. Unfortunately, the signaling pathways regulating MEP differentiation largely remain to be elucidated. This study aimed to analyze the role and the underlying molecular mechanism of miR-1915-3p in megakaryocytic and erythroid differentiation. METHODS We utilized miRNA mimics and miRNA sponge to alter the expression of miR-1915-3p in megakaryocytic and/or erythroid potential cells; siRNA and overexpression plasmid to change the expression of SOCS4, a potential target of miR-1915-3p. The expression of relevant surface markers was detected by flow cytometry. We scanned for miR-1915-3p target genes by mRNA expression profiling and bioinformatic analysis, and confirmed the targeting by dual-luciferase reporter assay, western blot and gain- and lost-of-function studies. One-way ANOVA and t-test were used to analyze the statistical significance. RESULTS In this study, overexpression or knockdown of miR-1915-3p inhibited or promoted erythroid differentiation, respectively. Accordingly, we scanned for miR-1915-3p target genes and confirmed that SOCS4 is one of the direct targets of miR-1915-3p. An attentive examination of the endogenous expression of SOCS4 during megakaryocytic and erythroid differentiation suggested the involvement of SOCS4 in erythroid/megakaryocytic lineage determination. SOCS4 knockdown lessened erythroid surface markers expression, as well as improved megakaryocytic differentiation, similar to the effects of miR-1915-3p overexpression. While SOCS4 overexpression resulted in reversed effects. SOCS4 overexpression in miR-1915-3p upregulated cells rescued the effect of miR-1915-3p. CONCLUSIONS miR-1915-3p acts as a negative regulator of erythropoiesis, and positively in thrombopoiesis. SOCS4 is one of the key mediators of miR-1915-3p during the differentiation of MEPs.
Collapse
Affiliation(s)
- Xin Yuan
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Pengcong Liu
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Lei Xu
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Liqing Liang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Qian Dong
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Tao Fan
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Wen Yue
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Mingyi Qu
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.
| | - Xuetao Pei
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.
| | - Xiaoyan Xie
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.
| |
Collapse
|
23
|
Yang K, Tang J, Li H, Zhang H, Ding J, Li Z, Luo J. LncRNAs in Kawasaki disease and Henoch-Schönlein purpura: mechanisms and clinical applications. Mol Cell Biochem 2024; 479:1969-1984. [PMID: 37639198 DOI: 10.1007/s11010-023-04832-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023]
Abstract
Kawasaki disease (KD) and Henoch-Schönlein purpura (HSP) are the two most predominant types of childhood vasculitis. In childhood vasculitis, factors such as lack of sensitive diagnostic indicators and adverse effects of drug therapy may cause multiorgan system involvement and complications and even death. Many studies suggest that long noncoding RNAs (lncRNAs) are involved in the mechanism of vasculitis development in children and can be used to diagnose or predict prognosis by lncRNAs. In existing drug therapies, lncRNAs are also involved in drug-mediated treatment mechanisms and are expected to improve drug toxicity. The aim of this review is to summarize the link between lncRNAs and the pathogenesis of KD and HSP. In addition, we review the potential applications of lncRNAs in multiple dimensions, such as diagnosis, treatment, and prognosis prediction. This review highlights that targeting lncRNAs may be a novel therapeutic strategy to improve and treat KD and HSP.
Collapse
Affiliation(s)
- Kangping Yang
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Jiayao Tang
- School of Pharmacy, Nanchang University, Nanchang, China
| | - Haoying Li
- Queen Mary School of Nanchang University, Nanchang, China
| | - Hejin Zhang
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Jiatong Ding
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Zelin Li
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Jinghua Luo
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
24
|
Raskov H, Orhan A, Agerbæk MØ, Gögenur I. The impact of platelets on the metastatic potential of tumour cells. Heliyon 2024; 10:e34361. [PMID: 39114075 PMCID: PMC11305202 DOI: 10.1016/j.heliyon.2024.e34361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
In cancer, activation of platelets by tumor cells is critical to disease progression. Development of precise antiplatelet targeting may improve outcomes from anticancer therapy. Alongside a distinct shift in functionality such as pro-metastatic and pro-coagulant properties, platelet production is often accelerated significantly early in carcinogenesis and the cancer-associated thrombocytosis increases the risk of metastasis formation and thromboembolic events. Tumor-activated platelets facilitate the proliferation of migrating tumor cells and shield them from immune surveillance and physical stress during circulation. Additionally, platelet-tumor cell interactions promote tumor cell intravasation, intravascular arrest, and extravasation through a repertoire of adhesion molecules, growth factors and angiogenic factors. Particularly, the presence of circulating tumor cell (CTC) clusters in association with platelets is a negative prognostic indicator. The contribution of platelets to the metastatic process is an area of intense investigation and this review provides an overview of the advances in understanding platelet-tumor cell interactions and their contribution to disease progression. Also, we review the potential of targeting platelets to interfere with the metastatic process.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
- University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Mette Ørskov Agerbæk
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Zhao J, Tian H, Zhao X, Lan L, Liu H, Sun Y, Yu F. PKCα Induced the Generation of Extracellular Vesicles in Activated Platelets to Promote Breast Cancer Metastasis. Int J Biol Sci 2024; 20:3956-3971. [PMID: 39113702 PMCID: PMC11302887 DOI: 10.7150/ijbs.89822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 07/05/2024] [Indexed: 08/10/2024] Open
Abstract
Platelet extracellular vesicles (PEVs) play an important role in tumor development. However, the mechanisms underlying their biogenesis have not been fully elucidated. Protein kinase Cα (PKCα) is an important regulator of platelet activation, but the effect of PKCα on EV generation is unclear. We used small-particle flow cytometry and found that the number of PEVs was increased in patients with breast cancer compared to those with benign breast disease. This was accompanied by increased levels of activated PKCα in breast cancer platelets. Treating platelets with the PKCα agonist phorbol 12-myristate 13-acetate (PMA) increased the phosphorylation PKCα and induced PEV production, while the PKCα inhibitor GÖ6976 showed the opposite effects. Notably, incubating platelets from patients with benign tumors with the culture supernatant of MDA-MB-231 cells induced PKCα phosphorylation in the platelets. Mass spectrometry and coimmunoprecipitation assays showed that Dynamin 2 (DNM2), a member of the guanosine-triphosphate-binding protein family, might cooperate with activated PKCα to regulate PEV production by breast cancer platelets. Similar results were observed in a mouse model of lung metastasis. In addition, PEVs were engulfed by breast cancer cells and promoted cancer cell migration and invasion via miR-1297 delivery. These findings suggested that PKCα cooperates with DNM2 to induce PEV generation, and PEV release might triggered by factors in the breast cancer environment.
Collapse
Affiliation(s)
- Jinghua Zhao
- Dept of Breast Surgery, Yat-Sen Breast Tumor Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Huan Tian
- Dept of Breast Surgery, Yat-Sen Breast Tumor Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Dept of Breast Surgery, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaona Zhao
- Dept of Breast Surgery, Yat-Sen Breast Tumor Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Lan Lan
- School of Life sciences, Guangzhou University, Guangzhou, Guangdong, China
| | - Huanhuan Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Plastic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat- sen University, Guangzhou, Guangdong, China
| | - Yi Sun
- Department of Breast Surgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Fengyan Yu
- Dept of Breast Surgery, Yat-Sen Breast Tumor Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Ricklefs FL, Wollmann K, Salviano-Silva A, Drexler R, Maire CL, Kaul MG, Reimer R, Schüller U, Heinemann S, Kolbe K, Mummert T, Glatzel M, Peine S, Gempt J, Westphal M, Dührsen L, Lamszus K. Circulating extracellular vesicles as biomarker for diagnosis, prognosis, and monitoring in glioblastoma patients. Neuro Oncol 2024; 26:1280-1291. [PMID: 38567448 PMCID: PMC11226867 DOI: 10.1093/neuonc/noae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) obtained by noninvasive liquid biopsy from patient blood can serve as biomarkers. Here, we investigated the potential of circulating plasma EVs to serve as an indicator in the diagnosis, prognosis, and treatment response of glioblastoma patients. METHODS Plasma samples were collected from glioblastoma patients at multiple timepoints before and after surgery. EV concentrations were measured by nanoparticle tracking analysis and imaging flow cytometry. Tumor burden and edema were quantified by 3D reconstruction. EVs and tumors were further monitored in glioma-bearing mice. RESULTS Glioblastoma patients displayed a 5.5-fold increase in circulating EVs compared to healthy donors (P < .0001). Patients with higher EV levels had significantly shorter overall survival and progression-free survival than patients with lower levels, and the plasma EV concentration was an independent prognostic parameter for overall survival. EV levels correlated with the extent of peritumoral fluid-attenuated inversion recovery hyperintensity but not with the size of the contrast-enhancing tumor, and similar findings were obtained in mice. Postoperatively, EV concentrations decreased rapidly back to normal levels, and the magnitude of the decline was associated with the extent of tumor resection. EV levels remained low during stable disease, but increased again upon tumor recurrence. In some patients, EV resurgence preceded the magnetic resonance imaging detectability of tumor relapse. CONCLUSIONS Our findings suggest that leakiness of the blood-brain barrier may primarily be responsible for the high circulating EV concentrations in glioblastoma patients. Elevated EVs reflect tumor presence, and their quantification may thus be valuable in assessing disease activity.
Collapse
Affiliation(s)
- Franz L Ricklefs
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kathrin Wollmann
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Amanda Salviano-Silva
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Richard Drexler
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cecile L Maire
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael G Kaul
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rudolph Reimer
- Heinrich-Pette-Institut, Leibnitz Institute for Experimental Virology, Hamburg, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
- Department of Paediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarina Heinemann
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Kolbe
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Mummert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Peine
- Institute of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Gempt
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Westphal
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lasse Dührsen
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Lamszus
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
27
|
Yang W, Wang L, Fan L, Li W, Zhao Y, Shang L, Jiang M. Photothermal Responsive Microcarriers Encapsulated With Cangrelor and 5-Fu for Colorectal Cancer Treatment. SMALL METHODS 2024; 8:e2301002. [PMID: 38127997 DOI: 10.1002/smtd.202301002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 11/29/2023] [Indexed: 12/23/2023]
Abstract
Localized chemotherapy is emerging as a potential strategy for cancer treatment due to its low systemic toxicity. However, the immune evasion of tumor cells and the lack of an intelligent design of the delivery system limit its clinical application. Herein, photothermal responsive microcarriers are designed by microfluidic electrospray for colorectal tumor treatment. The microcarriers loaded with Cangrelor, 5-FU and MXene (G-M@F/C+NIR) show sustained delivery of antiplatelet drug Cangrelor, thus inhibiting the activity of platelets, interactions of platelet-tumor cell, as well as the tumor cells invasion and epithelial-mesenchymal transition (EMT). In addition, the sustained delivery of chemotherapeutics 5-FU and the photothermal effect provided by MXene enable the microcarriers to inhibit tumor cells proliferation and migration. In vivo studies validate that the G-M@F/C+NIR microcarriers significantly inhibites tumor growth, decreased the expression of Ki-67 in tumor cells and vascular endothelial growth factor (VEGF) in the tumor microenvironment, while increased the expression of E-cadherin. It is believe that by means of the proposed photothermal responsive microcarriers, the synergistic strategy of platelet inhibition, chemotherapy, and photothermal therapy can find practical applications in cancer treatment.
Collapse
Affiliation(s)
- Wei Yang
- Clinical Laboratory Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Li Wang
- Clinical Laboratory Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Lu Fan
- Clinical Laboratory Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wenzhao Li
- Clinical Laboratory Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yuanjin Zhao
- Clinical Laboratory Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Luoran Shang
- Zhongshan-Xuhui Hospital and the Shanghai Key Laboratory of Medical Epigenetics the International Co-laboratory of Medical Epigenetics and Metabolism Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Minghua Jiang
- Clinical Laboratory Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
28
|
Liu M, Wen Z, Zhang T, Zhang L, Liu X, Wang M. The role of exosomal molecular cargo in exosome biogenesis and disease diagnosis. Front Immunol 2024; 15:1417758. [PMID: 38983854 PMCID: PMC11231912 DOI: 10.3389/fimmu.2024.1417758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024] Open
Abstract
Exosomes represent a type of extracellular vesicles derived from the endosomal pathway that transport diverse molecular cargoes such as proteins, lipids, and nucleic acids. These cargoes have emerged as crucial elements impacting disease diagnosis, treatment, and prognosis, and are integral to the process of exosome formation. This review delves into the essential molecular cargoes implicated in the phases of exosome production and release. Emphasis is placed on their significance as cancer biomarkers and potential therapeutic targets, accompanied by an exploration of the obstacles and feasible applications linked to these developments.
Collapse
Affiliation(s)
- Meijin Liu
- Laboratory Medicine, People's Hospital of Ganzhou Economic Development Zone, Ganzhou, China
| | - Zhenzhen Wen
- Laboratory Medicine, People's Hospital of Ganzhou Economic Development Zone, Ganzhou, China
| | - Tingting Zhang
- Laboratory Medicine, People's Hospital of Ganzhou Economic Development Zone, Ganzhou, China
| | - Linghan Zhang
- Laboratory Medicine, People's Hospital of Ganzhou Economic Development Zone, Ganzhou, China
| | - Xiaoyan Liu
- Laboratory Medicine, People's Hospital of Ganzhou Economic Development Zone, Ganzhou, China
| | - Maoyuan Wang
- Laboratory Medicine, People's Hospital of Ganzhou Economic Development Zone, Ganzhou, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Gannan Medical University, GanZhou, China
| |
Collapse
|
29
|
Tutuianu A, Anene CA, Shelton M, Speirs V, Whitelaw DC, Thorpe J, Roberts W, Boyne JR. Platelet-derived microvesicles isolated from type-2 diabetes mellitus patients harbour an altered miRNA signature and drive MDA-MB-231 triple-negative breast cancer cell invasion. PLoS One 2024; 19:e0304870. [PMID: 38900754 PMCID: PMC11189239 DOI: 10.1371/journal.pone.0304870] [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: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
The underlying causes of breast cancer are diverse, however, there is a striking association between type 2 diabetes and poor patient outcomes. Platelet activation is a common feature of both type 2 diabetes and breast cancer and has been implicated in tumourigenesis through a multitude of pathways. Here transcriptomic analysis of type 2 diabetes patient-derived platelet microvesicles revealed an altered miRNA signature compared with normoglycaemic control patients. Interestingly, interrogation of these data identifies a shift towards an oncogenic signature in type 2 diabetes-derived platelet microvesicles, with increased levels of miRNAs implicated in breast cancer progression and poor prognosis. Functional studies demonstrate that platelet microvesicles isolated from type 2 diabetes patient blood are internalised by triple-negative breast cancer cells in vitro, and that co-incubation with type 2 diabetes patient-derived platelet microvesicles led to significantly increased expression of epithelial to mesenchymal transition markers and triple-negative breast cancer cell invasion compared with platelet microvesicles from healthy volunteers. Together, these data suggest that circulating PMVs in type 2 diabetes patients may contribute to the progression of triple-negative breast cancer.
Collapse
Affiliation(s)
- Anca Tutuianu
- School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
| | - Chinedu A. Anene
- Biomedical Science, School of Health, Leeds Beckett University, Leeds, United Kingdom
| | - Mikayla Shelton
- Biomedical Science, School of Health, Leeds Beckett University, Leeds, United Kingdom
| | - Valerie Speirs
- Institute of Medical Science, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, Scotland
| | - Donald C. Whitelaw
- Department of Diabetes and Endocrinology, Bradford Royal Infirmary, Bradford, United Kingdom
| | - Joanne Thorpe
- Department of Diabetes and Endocrinology, Bradford Royal Infirmary, Bradford, United Kingdom
| | - Wayne Roberts
- Biomedical Science, School of Health, Leeds Beckett University, Leeds, United Kingdom
| | - James R. Boyne
- Biomedical Science, School of Health, Leeds Beckett University, Leeds, United Kingdom
| |
Collapse
|
30
|
Xi Y, Min Z, Liu M, Lin X, Yuan ZH. Role and recent progress of P2Y12 receptor in cancer development. Purinergic Signal 2024:10.1007/s11302-024-10027-w. [PMID: 38874752 DOI: 10.1007/s11302-024-10027-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024] Open
Abstract
P2Y12 receptor (P2Y12R) is an adenosine-activated G protein-coupled receptor (GPCR) that plays a central role in platelet function, hemostasis, and thrombosis. P2Y12R activation can promote platelet aggregation and adhesion to cancer cells, promote tumor angiogenesis, and affect the tumor immune microenvironment (TIME) and tumor drug resistance, which is conducive to the progression of cancers. Meanwhile, P2Y12R inhibitors can inhibit this effect, suggesting that P2Y12R may be a potential therapeutic target for cancer. P2Y12R is involved in cancer development and metastasis, while P2Y12R inhibitors are effective in inhibiting cancer. However, a new study suggests that long-term use of P2Y12R inhibitors may increase the risk of cancer and the mechanism remains to be explored. In this paper, we reviewed the structural and functional characteristics of P2Y12R and its role in cancer. We explored the role of P2Y12R inhibitors in different tumors and the latest advances by summarizing the basic and clinical studies on the effects of P2Y12R inhibitors on tumors.
Collapse
Affiliation(s)
- Yanni Xi
- Department of General Surgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332007, People's Republic of China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, Republic of China
| | - Zhenya Min
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, Republic of China
| | - Mianxue Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, Republic of China
| | - Xueqin Lin
- Department of Nursing, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
| | - Zhao-Hua Yuan
- Department of General Surgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332007, People's Republic of China.
| |
Collapse
|
31
|
Li X, Ren H, Peng L, Li J. Prognostic value of pretreatment platelet count, fibrinogen and d-dimer levels in osteosarcoma patients: A meta-analysis. Medicine (Baltimore) 2024; 103:e38463. [PMID: 39259059 PMCID: PMC11142833 DOI: 10.1097/md.0000000000038463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Previous studies explored the prognostic value of pretreatment platelet count, fibrinogen, and d-dimer level in patients with several types of cancer, however, a comprehensive conclusion has not been reached in osteosarcoma patients. METHODS PubMed, Web of Science, Embase, and CNKI databases were systematically searched for eligible studies up to May 09, 2023, and pooled hazard ratios (HRs) with corresponding 95% confidence intervals (CIs) were calculated to assess the prognostic impact of these indicators in osteosarcoma patients. RESULTS Twelve studies from China consisting of 1682 patients were finally included. Our findings revealed that an elevated level of pretreatment platelet or d-dimer was associated with a worse outcome of overall survival (platelet: HR = 1.63, 95% CI: 1.18-2.26, P = .003; d-dimer: HR = 2.29, 95% CI: 1.58-3.31, P < .001). CONCLUSION Based on current evidence, pretreatment platelet count and d-dimer level could be good prognostic biomarkers for Chinese osteosarcoma patients. However, future validation is also needed.
Collapse
Affiliation(s)
- Xianfeng Li
- Department of Orthopedic, The Second People’s Hospital of Yibin, Yibin, China
| | - Honghong Ren
- Department of Nephrology, The Second People’s Hospital of Yibin, Yibin, China
| | - Lipeng Peng
- Department of Orthopedic, The Second People’s Hospital of Yibin, Yibin, China
| | - Jie Li
- Department of Orthopedic, The Second People’s Hospital of Yibin, Yibin, China
| |
Collapse
|
32
|
Garcia-Leon MJ, Liboni C, Mittelheisser V, Bochler L, Follain G, Mouriaux C, Busnelli I, Larnicol A, Colin F, Peralta M, Osmani N, Gensbittel V, Bourdon C, Samaniego R, Pichot A, Paul N, Molitor A, Carapito R, Jandrot-Perrus M, Lefebvre O, Mangin PH, Goetz JG. Platelets favor the outgrowth of established metastases. Nat Commun 2024; 15:3297. [PMID: 38740748 DOI: 10.1038/s41467-024-47516-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Despite abundant evidence demonstrating that platelets foster metastasis, anti-platelet agents have low therapeutic potential due to the risk of hemorrhages. In addition, whether platelets can regulate metastasis at the late stages of the disease remains unknown. In this study, we subject syngeneic models of metastasis to various thrombocytopenic regimes to show that platelets provide a biphasic contribution to metastasis. While potent intravascular binding of platelets to tumor cells efficiently promotes metastasis, platelets further support the outgrowth of established metastases via immune suppression. Genetic depletion and pharmacological targeting of the glycoprotein VI (GPVI) platelet-specific receptor in humanized mouse models efficiently reduce the growth of established metastases, independently of active platelet binding to tumor cells in the bloodstream. Our study demonstrates therapeutic efficacy when targeting animals bearing growing metastases. It further identifies GPVI as a molecular target whose inhibition can impair metastasis without inducing collateral hemostatic perturbations.
Collapse
Affiliation(s)
- Maria J Garcia-Leon
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Paris, France.
- Domain therapeutics, Parc d'Innovation - 220 Boulevard Gonthier D'Andernach, 67400, Strasbourg - Illkirch, France.
| | - Cristina Liboni
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Vincent Mittelheisser
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Louis Bochler
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Gautier Follain
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Clarisse Mouriaux
- UMR_S 1255, INSERM, Etablissement Français du Sang-Alsace, Université de Strasbourg, F-67000, Strasbourg, France
| | - Ignacio Busnelli
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Annabel Larnicol
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Florent Colin
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Marina Peralta
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Naël Osmani
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Valentin Gensbittel
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Catherine Bourdon
- UMR_S 1255, INSERM, Etablissement Français du Sang-Alsace, Université de Strasbourg, F-67000, Strasbourg, France
| | - Rafael Samaniego
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Unidad de Microscopía Confocal, Madrid, Spain
| | - Angélique Pichot
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, France
| | - Nicodème Paul
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, France
| | - Anne Molitor
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, France
| | - Raphaël Carapito
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091, Strasbourg, France
| | | | - Olivier Lefebvre
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Pierre H Mangin
- UMR_S 1255, INSERM, Etablissement Français du Sang-Alsace, Université de Strasbourg, F-67000, Strasbourg, France.
| | - Jacky G Goetz
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
33
|
Merij LB, da Silva LR, Palhinha L, Gomes MT, Dib PRB, Martins-Gonçalves R, Toledo-Quiroga K, Raposo-Nunes MA, Andrade FB, de Toledo Martins S, Nascimento ALR, Rocha VN, Alves LR, Bozza PT, de Oliveira Trugilho MR, Hottz ED. Density-based lipoprotein depletion improves extracellular vesicle isolation and functional analysis. J Thromb Haemost 2024; 22:1372-1388. [PMID: 38278418 DOI: 10.1016/j.jtha.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Blood plasma is the main source of extracellular vesicles (EVs) in clinical studies aiming to identify biomarkers and to investigate pathophysiological processes, especially regarding EV roles in inflammation and thrombosis. However, EV isolation from plasma has faced the fundamental issue of lipoprotein contamination, representing an important bias since lipoproteins are highly abundant and modulate cell signaling, metabolism, and thromboinflammation. OBJECTIVES Here, we aimed to isolate plasma EVs after depleting lipoproteins, thereby improving sample purity and EV thromboinflammatory analysis. METHODS Density-based gradient ultracentrifugation (G-UC) was used for lipoprotein depletion before EV isolation from plasma through size-exclusion chromatography (SEC) or serial centrifugation (SC). Recovered EVs were analyzed by size, concentration, cellular source, ultrastructure, and bottom-up proteomics. RESULTS G-UC efficiently separated lipoproteins from the plasma, allowing subsequent EV isolation through SEC or SC. Combined analysis from EV proteomics, cholesterol quantification, and apoB-100 detection confirmed the significant reduction in lipoproteins from isolated EVs. Proteomic analysis identified similar gene ontology and cellular components in EVs, regardless of lipoprotein depletion, which was consistent with similar EV cellular sources, size, and ultrastructure by flow cytometry and transmission electron microscopy. Importantly, lipoprotein depletion increased the detection of less abundant proteins in EV proteome and enhanced thromboinflammatory responses of platelets and monocytes stimulated in vitro with EV isolates. CONCLUSION Combination of G-UC+SEC significantly reduced EV lipoprotein contamination without interfering in EV cellular source, gene ontology, and ultrastructure, allowing the recovery of highly pure EVs with potential implications for functional assays and proteomic and lipidomic analyses.
Collapse
Affiliation(s)
- Laura Botelho Merij
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Luana Rocha da Silva
- Laboratory of Toxinology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lohanna Palhinha
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Milena Tavares Gomes
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Paula Ribeiro Braga Dib
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Remy Martins-Gonçalves
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kemily Toledo-Quiroga
- Laboratory of Toxinology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Fernanda Brandi Andrade
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Sharon de Toledo Martins
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, ICC-Fiocruz, Curitiba, Paraná, Brazil
| | - Ana Lúcia Rosa Nascimento
- Laboratory of Ultrastructure and Tissue, Department of Histology and Embryology, State University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinicius Novaes Rocha
- Laboratory of Veterinary Pathology and Histology, Department of Veterinary Medicine, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Lysangela Ronalte Alves
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, ICC-Fiocruz, Curitiba, Paraná, Brazil
| | - Patrícia T Bozza
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, ICC-Fiocruz, Curitiba, Paraná, Brazil
| | - Monique Ramos de Oliveira Trugilho
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil; Center for Technological Development in Health, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Eugenio D Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| |
Collapse
|
34
|
Hua Y, He Z, Ni Y, Sun L, Wang R, Li Y, Li X, Jiang G. Silk fibroin and hydroxypropyl cellulose composite injectable hydrogel-containing extracellular vesicles for myocardial infarction repair. Biomed Phys Eng Express 2024; 10:045001. [PMID: 38640908 DOI: 10.1088/2057-1976/ad40b2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 04/19/2024] [Indexed: 04/21/2024]
Abstract
Extracellular vesicles (EVs) have been recognized as one of the promising specific drugs for myocardial infarction (MI) prognosis. Nevertheless, low intramyocardial retention of EVs remains a major impediment to their clinical application. In this study, we developed a silk fibroin/hydroxypropyl cellulose (SF/HPC) composite hydrogel combined with AC16 cell-derived EVs targeted modification by folic acid for the treatment of acute myocardial infarction repair. EVs were functionalized by distearoylphosphatidyl ethanolamine-polyethylene glycol (DSPE-PEG-FA) via noncovalent interaction for targeting and accelerating myocardial infarction repair.In vitro, cytocompatibility analyses revealed that the as-prepared hydrogels had excellent cell viability by MTT assay and the functionalized EVs had higher cell migration by scratch assay.In vivo, the composite hydrogels can promote myocardial tissue repair effects by delaying the process of myocardial fibrosis and promoting angiogenesis of infarct area in MI rat model.
Collapse
Affiliation(s)
- Yinjian Hua
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, People's Republic of China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, People's Republic of China
| | - Zhengfei He
- Department of Cardiology, The First People's Hospital, Fuyang, Hangzhou, 311400, People's Republic of China
| | - Yunjie Ni
- Department of Cardiology, The First People's Hospital, Fuyang, Hangzhou, 311400, People's Republic of China
| | - Linggang Sun
- Department of Cardiology, The First People's Hospital, Fuyang, Hangzhou, 311400, People's Republic of China
| | - Rui Wang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, People's Republic of China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, People's Republic of China
| | - Yan Li
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, People's Republic of China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, People's Republic of China
| | - Xintong Li
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, People's Republic of China
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, People's Republic of China
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, People's Republic of China
| |
Collapse
|
35
|
Zeng QY, Qin Y, Shi Y, Mu XY, Huang SJ, Yang YH, Liu SM, An ZM, Li SQ. Systemic immune-inflammation index and all-cause and cause-specific mortality in sarcopenia: a study from National Health and Nutrition Examination Survey 1999-2018. Front Immunol 2024; 15:1376544. [PMID: 38638440 PMCID: PMC11024272 DOI: 10.3389/fimmu.2024.1376544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Abstract
Background Sarcopenia, common in the elderly, often linked to chronic diseases, correlates with inflammation.The association between SII and mortality in sarcopenia patients is underexplored, this study investigates this relationship in a U.S. adult cohort. Methods We analyzed 1999-2018 NHANES data, focusing on 2,974 adults with sarcopenia. Mortality outcomes were determined by linking to National Death Index (NDI) records up to December 31, 2019. Using a weighted sampling design, participants were grouped into three groups by the Systemic Immune-Inflammation Index (SII). We used Cox regression models, adjusting for demographic and clinical variables, to explore SII's association with all-cause and cause-specific mortality in sarcopenia, performing sensitivity analyses for robustness. Results Over a median follow-up of 9.2 years, 829 deaths occurred. Kaplan-Meier analysis showed significant survival differences across SII groups. The highest SII group showed higher hazard ratios (HRs) for all-cause and cause-specific mortality in both crude and adjusted models. The highest SII group had a higher HR for all-cause(1.57, 1.25-1.98), cardiovascular(1.61, 1.00-2.58), cancer(2.13, 1.32-3.44), and respiratory disease mortality(3.21, 1.66-6.19) in fully adjusted models. Subgroup analyses revealed SII's association with all-cause mortality across various demographics, including age, gender, and presence of diabetes or cardiovascular disease. Sensitivity analyses, excluding participants with cardiovascular diseases, those who died within two years of follow-up, or those under 45 years of age, largely reflected these results, with the highest SII group consistently demonstrating higher HRs for all types of mortality in both unadjusted and adjusted models. Conclusion Our study is the first to demonstrate a significant relationship between SII and increased mortality risks in a sarcopenia population.
Collapse
Affiliation(s)
- Qing-Yue Zeng
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Qin
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Shi
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xing-Yu Mu
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shi-Jun Huang
- Department of Critical Care Medicine, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yu-Hao Yang
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Si-Min Liu
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen-Mei An
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuang-Qing Li
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
36
|
Esmaeilzadeh A, Yeganeh PM, Nazari M, Esmaeilzadeh K. Platelet-derived extracellular vesicles: a new-generation nanostructured tool for chronic wound healing. Nanomedicine (Lond) 2024; 19:915-941. [PMID: 38445377 DOI: 10.2217/nnm-2023-0344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
Chronic nonhealing wounds pose a serious challenge to regaining skin function and integrity. Platelet-derived extracellular vesicles (PEVs) are nanostructured particles with the potential to promote wound healing since they can enhance neovascularization and cell migration and reduce inflammation and scarring. This work provides an innovative overview of the technical laboratory issues in PEV production, PEVs' role in chronic wound healing and the benefits and challenges in its clinical translation. The article also explores the challenges of proper sourcing, extraction techniques and storage conditions, and discusses the necessity of further evaluations and combinational therapeutics, including dressing biomaterials, M2-derived exosomes, mesenchymal stem cells-derived extracellular vesicles and microneedle technology, to boost their therapeutic efficacy as advanced strategies for wound healing.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
| | | | - Mahdis Nazari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
| | - Kimia Esmaeilzadeh
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
| |
Collapse
|
37
|
Pang Q, Gong X, Pan H, Wang Y, Hu X, Liu H, Jin H. Platelet count as a predictor of vascular invasion and extrahepatic metastasis in hepatocellular carcinoma: A systematic review and meta-analysis. Heliyon 2024; 10:e28173. [PMID: 38545227 PMCID: PMC10966694 DOI: 10.1016/j.heliyon.2024.e28173] [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: 04/29/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Vascular invasion (VI) indicates highly invasive tumor biological behavior and is a major determining factor of poor survival and high risk of metastasis in hepatocellular carcinoma (HCC). Epidemiological evidence of the association between pretherapeutic platelet count (PLT) and the risk of VI and extrahepatic metastasis in HCC remains controversial. METHODS A systematic retrieval was executed in databases of PubMed, Embase, and Web of Science until Dec 2022. Effect size and 95% confidence interval (CI) were extracted or estimated to synthetically investigate the effects of pretherapeutic PLT on VI and extrahepatic metastasis. Meta-analyses were performed by using a random or a fixed effects model. RESULTS Finally, the current meta-analysis included 15 studies with a total of 12,378 HCC patients. It was shown that, patients with a higher pretherapeutic level of PLT had a significantly increased risk of VI (11 studies,8,759 patients; OR = 1.44, 95%CI: 1.02-2.02) and extrahepatic metastasis (6 studies,8, 951 patients; OR = 2.51, 95% CI: 2.19-2.88) in comparison with patients with a lower PLT. Funnel plots and Begg's tests indicated that there were no significant publication biases. CONCLUSION This meta-analysis shows that pretherapeutic elevated PLT is associated with an increased risk of VI and extrahepatic metastasis in HCC.
Collapse
Affiliation(s)
- Qing Pang
- Department of General Surgery, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Xuankun Gong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Hongtao Pan
- Department of General Surgery, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
| | - Yong Wang
- Department of General Surgery, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
| | - Xiaosi Hu
- Department of General Surgery, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
| | - Huichun Liu
- Department of General Surgery, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
| | - Hao Jin
- Department of General Surgery, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
| |
Collapse
|
38
|
Ebrahim T, Ebrahim AS, Kandouz M. Diversity of Intercellular Communication Modes: A Cancer Biology Perspective. Cells 2024; 13:495. [PMID: 38534339 PMCID: PMC10969453 DOI: 10.3390/cells13060495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/27/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024] Open
Abstract
From the moment a cell is on the path to malignant transformation, its interaction with other cells from the microenvironment becomes altered. The flow of molecular information is at the heart of the cellular and systemic fate in tumors, and various processes participate in conveying key molecular information from or to certain cancer cells. For instance, the loss of tight junction molecules is part of the signal sent to cancer cells so that they are no longer bound to the primary tumors and are thus free to travel and metastasize. Upon the targeting of a single cell by a therapeutic drug, gap junctions are able to communicate death information to by-standing cells. The discovery of the importance of novel modes of cell-cell communication such as different types of extracellular vesicles or tunneling nanotubes is changing the way scientists look at these processes. However, are they all actively involved in different contexts at the same time or are they recruited to fulfill specific tasks? What does the multiplicity of modes mean for the overall progression of the disease? Here, we extend an open invitation to think about the overall significance of these questions, rather than engage in an elusive attempt at a systematic repertory of the mechanisms at play.
Collapse
Affiliation(s)
- Thanzeela Ebrahim
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Abdul Shukkur Ebrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Mustapha Kandouz
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48202, USA
| |
Collapse
|
39
|
Zhang Z, Xu X, Zhang D, Zhao S, Wang C, Zhang G, Chen W, Liu J, Gong H, Rixiati Y, Li S, Shen T, Li J. Targeting Erbin-mitochondria axis in platelets/megakaryocytes promotes B cell-mediated antitumor immunity. Cell Metab 2024; 36:541-556.e9. [PMID: 38232736 DOI: 10.1016/j.cmet.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/08/2023] [Accepted: 12/15/2023] [Indexed: 01/19/2024]
Abstract
The roles of platelets/megakaryocytes (MKs), the key components in the blood system, in the tumor microenvironment and antitumor immunity are unclear. In patients with colorectal cancer, the number of platelets was significantly increased in patients with metastasis, and Erbin expression was highly expressed in platelets from patients with metastases. Moreover, Erbin knockout in platelets/MKs suppressed lung metastasis in mice and promoted aggregations of platelets. Mechanistically, Erbin-deficient platelets have increasing mitochondrial oxidative phosphorylation and secrete lipid metabolites like acyl-carnitine (Acar) by abolishing interaction with prothrombotic protein ESAM. Notably, Acar enhanced the activity of mitochondrial electron transport chain complex and mitochondrial oxidative phosphorylation in B cells by acetylation of H3K27 epigenetically. Targeting Erbin in platelets/MKs by a nanovesicle system dramatically attenuated lung metastasis in mice in vivo. Our study identifies an Erbin-mitochondria axis in platelets/MKs, which suppresses B cell-mediated antitumor immunity, suggesting a new way for the treatment of metastasis.
Collapse
Affiliation(s)
- Zilong Zhang
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xu Xu
- Department of Pathology, Soochow University Medical School, Suzhou, China
| | - Di Zhang
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Songsong Zhao
- College of Chemistry, Chemical Engineering and Materials Science, Suzhou, China
| | - Chuyi Wang
- Department of Pathology, Soochow University Medical School, Suzhou, China
| | - Guilin Zhang
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wenshu Chen
- Department of Pathology, Soochow University Medical School, Suzhou, China
| | - Jinglin Liu
- Department of Pathology, Soochow University Medical School, Suzhou, China
| | - Huimin Gong
- Department of Pathology, Soochow University Medical School, Suzhou, China
| | | | - Shi Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tong Shen
- Department of Pathology, Soochow University Medical School, Suzhou, China.
| | - Jianming Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Department of Pathology, Soochow University Medical School, Suzhou, China.
| |
Collapse
|
40
|
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: 6] [Impact Index Per Article: 6.0] [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
|
41
|
Da X, Cao B, Mo J, Xiang Y, Hu H, Qiu C, Zhang C, Lv B, Zhang H, He C, Yang Y. Inhibition of growth of hepatocellular carcinoma by co-delivery of anti-PD-1 antibody and sorafenib using biomimetic nano-platelets. BMC Cancer 2024; 24:273. [PMID: 38409035 PMCID: PMC10898182 DOI: 10.1186/s12885-024-12006-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/14/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Traditional nanodrug delivery systems have some limitations, such as eliciting immune responses and inaccuracy in targeting tumor microenvironments. MATERIALS AND METHODS Targeted drugs (Sorafenib, Sora) nanometers (hollow mesoporous silicon, HMSN) were designed, and then coated with platelet membranes to form aPD-1-PLTM-HMSNs@Sora to enhance the precision of drug delivery systems to the tumor microenvironment, so that more effective immunotherapy was achieved. RESULTS These biomimetic nanoparticles were validated to have the same abilities as platelet membranes (PLTM), including evading the immune system. The successful coating of HMSNs@Sora with PLTM was corroborated by transmission electron microscopy (TEM), western blot and confocal laser microscopy. The affinity of aPD-1-PLTM-HMSNs@Sora to tumor cells was stronger than that of HMSNs@Sora. After drug-loaded particles were intravenously injected into hepatocellular carcinoma model mice, they were demonstrated to not only directly activate toxic T cells, but also increase the triggering release of Sora. The combination of targeted therapy and immunotherapy was found to be of gratifying antineoplastic function on inhibiting primary tumor growth. CONCLUSIONS The aPD-1-PLTM-HMSNs@Sora nanocarriers that co-delivery of aPD-1 and Sorafenib integrates unique biomimetic properties and excellent targeting performance, and provides a neoteric idea for drug delivery of personalized therapy for primary hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- Xuanbo Da
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, 200092, Shanghai, China
| | - Bangping Cao
- School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, 200072, Shanghai, China
| | - Jiantao Mo
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Yukai Xiang
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, 200092, Shanghai, China
| | - Hai Hu
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, 200092, Shanghai, China
| | - Chen Qiu
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, 200092, Shanghai, China
| | - Cheng Zhang
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, 200092, Shanghai, China
| | - Beining Lv
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, 200092, Shanghai, China
| | - Honglei Zhang
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, 200092, Shanghai, China
| | - Chuanqi He
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, 200092, Shanghai, China
| | - Yulong Yang
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, 200092, Shanghai, China.
| |
Collapse
|
42
|
Saberian M, Abak N. Hydrogel-mediated delivery of platelet-derived exosomes: Innovations in tissue engineering. Heliyon 2024; 10:e24584. [PMID: 38312628 PMCID: PMC10835177 DOI: 10.1016/j.heliyon.2024.e24584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
In this scholarly review, we conduct a thorough examination of the significant role played by platelet-derived exosomes (Plt-Exos) and hydrogels in the fields of tissue engineering and regenerative medicine. Our detailed investigation highlights the central involvement of Plt-Exos in various physiological and pathological processes, underscoring their potential contributions to diverse areas such as wound healing, neural rejuvenation, and cancer progression. Despite the promising therapeutic aspects, the notable variability in the isolation and characterization of pEVs underscores the need for a more rigorous and standardized methodology. Shifting our focus to hydrogels, they have emerged as promising biomaterials relevant to tissue engineering and regenerative medicine. Their unique characteristics, especially their chemical and physical adaptability, along with the modifiability of their biochemical properties, make hydrogels a captivating subject. These exceptional features open avenues for numerous tissue engineering applications, facilitating the delivery of essential growth factors, cytokines, and microRNAs. This analysis explores the innovative integration of Plt-Exos with hydrogels, presenting a novel paradigm in tissue engineering. Through the incorporation of Plt-Exos into hydrogels, there exists an opportunity to enhance tissue regeneration endeavors by combining the bioactive features of Plt-Exos with the restorative capabilities of hydrogel frameworks. In conclusion, the cooperative interaction between platelet-derived exosomes and hydrogels indicates a promising path in tissue engineering and regenerative medicine. Nevertheless, the successful execution of this approach requires a deep understanding of molecular dynamics, coupled with a dedication to refining isolation techniques.
Collapse
Affiliation(s)
- Mostafa Saberian
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Abak
- Hematology and Transfusion Science Department, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Li S, Lu Z, Wu S, Chu T, Li B, Qi F, Zhao Y, Nie G. The dynamic role of platelets in cancer progression and their therapeutic implications. Nat Rev Cancer 2024; 24:72-87. [PMID: 38040850 DOI: 10.1038/s41568-023-00639-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 12/03/2023]
Abstract
Systemic antiplatelet treatment represents a promising option to improve the therapeutic outcomes and therapeutic efficacy of chemotherapy and immunotherapy due to the critical contribution of platelets to tumour progression. However, until recently, targeting platelets as a cancer therapeutic has been hampered by the elevated risk of haemorrhagic and thrombocytopenic (low platelet count) complications owing to the lack of specificity for tumour-associated platelets. Recent work has advanced our understanding of the molecular mechanisms responsible for the contribution of platelets to tumour progression and metastasis. This has led to the identification of the biological changes in platelets in the presence of tumours, the complex interactions between platelets and tumour cells during tumour progression, and the effects of platelets on antitumour therapeutic response. In this Review, we present a detailed picture of the dynamic roles of platelets in tumour development and progression as well as their use in diagnosis, prognosis and monitoring response to therapy. We also provide our view on how to overcome challenges faced by the development of precise antiplatelet strategies for safe and efficient clinical cancer therapy.
Collapse
Affiliation(s)
- Suping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| | - Zefang Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Suying Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Tianjiao Chu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- College of Pharmaceutical Science, Jilin University, Changchun, China
| | - Bozhao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Feilong Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
44
|
Hu Y, Wang H, Liu Y. NETosis: Sculpting tumor metastasis and immunotherapy. Immunol Rev 2024; 321:263-279. [PMID: 37712361 DOI: 10.1111/imr.13277] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/16/2023]
Abstract
The process of neutrophil extracellular traps (NETs) formation, called NETosis, is a peculiar death modality of neutrophils, which was first observed as an immune response against bacterial infection. However, recent work has revealed the unique biology of NETosis in facilitating tumor metastatic process. Neutrophil extracellular traps released by the tumor microenvironment (TME) shield tumor cells from cytotoxic immunity, leading to impaired tumor clearance. Besides, tumor cells tapped by NETs enable to travel through vessels and subsequently seed distant organs. Targeted ablation of NETosis has been proven to be beneficial in potentiating the efficacy of cancer immunotherapy in the metastatic settings. This review outlines the impact of NETosis at almost all stages of tumor metastasis. Furthermore, understanding the multifaceted interplay between NETosis and the TME components is crucial for supporting the rational development of highly effective combination immunotherapeutic strategies with anti-NETosis for patients with metastatic disease.
Collapse
Affiliation(s)
- Yanyan Hu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Houhong Wang
- Department of General Surgery, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, China
| | - Yang Liu
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
45
|
Robert M, Scherlinger M. Platelets are a major player and represent a therapeutic opportunity in systemic lupus erythematosus. Joint Bone Spine 2024; 91:105622. [PMID: 37495075 DOI: 10.1016/j.jbspin.2023.105622] [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: 04/18/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by immune dysregulation and organ injury with a premature mortality due to cardiovascular diseases. Platelets, that are primarily known for their role in hemostasis, have been shown to play an active role in the pathogenesis and in the progression of immune-mediated inflammatory diseases. Here we summarize the evidence of their roles in SLE pathogenesis which supports the development of targeted treatments. Platelets and their precursors, the megakaryocytes, are intrinsically different in SLE patients compared with healthy controls. Different triggers related to innate and adaptive immunity activate platelets which release extracellular vesicles, soluble factors and interact with immune cells, thereby perpetuating inflammation. Platelets are involved in organ damage in SLE, especially in lupus nephritis and participate in the heightened cardiovascular mortality. They also play a clear role in antiphospholipid syndrome which can be associated with both thrombocytopenia and thrombosis. To tackle platelet activation and their interactions with immune cells now constitute promising therapeutic strategies in SLE.
Collapse
Affiliation(s)
- Marie Robert
- Service de médecine interne et immunologie clinique, centre hospitalier universitaire Édouard-Herriot, hospices civils de Lyon, Lyon, France
| | - Marc Scherlinger
- Service de rhumatologie, centre hospitalier universitaire de Strasbourg, 1, avenue Molière, 67098 Strasbourg, France; Laboratoire d'immuno-rhumatologie moléculaire, Institut national de la santé et de la recherche médicale (Inserm) UMR S 1109, Strasbourg, France; Centre national de référence des maladies auto-immunes et systémiques rares, Est/Sud-Ouest (RESO), France.
| |
Collapse
|
46
|
Kryczka J, Kassassir H, Papiewska-Pająk I, Boncela J. Gelatin In Situ Zymography to Study Gelatinase Activity in Colon Cancer Cells Treated with Platelet Microparticles (PMPs). Methods Mol Biol 2024; 2747:167-176. [PMID: 38038940 DOI: 10.1007/978-1-0716-3589-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
The platelet-derived microparticles (PMPs) have been connected with tumor progression and metastatic dissemination. PMPs infiltrate solid tumors and transfer platelet-derived cargo to cancer cells. The functional roles of PMPs in cancer progression are still poorly understood. PMPs, incorporated by colorectal cancer (CRC) cells, were shown to upregulate the expression and activity of matrix metalloproteases (MMPs). To investigate the impact of PMPs on the invasive potential of CRC, we established the protocol of dequenched gelatin (DG), fluorescein conjugate assay. The procedure confirms the activity of two gelatinases, namely, MMP2 and MMP9, that digest denatured collagen (gelatin). This "step-by-step" protocol, with notes and comments implemented to human CRC lines with different phenotypes and migratory potentials, should be sufficient to obtain representative and elegant results.
Collapse
Affiliation(s)
- Jakub Kryczka
- Institute of Medical Biology, Polish Academy of Science, Lodz, Poland
| | - Hassan Kassassir
- Institute of Medical Biology, Polish Academy of Science, Lodz, Poland
| | | | - Joanna Boncela
- Institute of Medical Biology, Polish Academy of Science, Lodz, Poland.
| |
Collapse
|
47
|
Wu YW, Lee DY, Lu YL, Delila L, Nebie O, Barro L, Changou CA, Lu LS, Goubran H, Burnouf T. Platelet extracellular vesicles are efficient delivery vehicles of doxorubicin, an anti-cancer drug: preparation and in vitro characterization. Platelets 2023; 34:2237134. [PMID: 37580876 DOI: 10.1080/09537104.2023.2237134] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 08/16/2023]
Abstract
Platelet extracellular vesicles (PEVs) are an emerging delivery vehi for anticancer drugs due to their ability to target and remain in the tumor microenvironment. However, there is still a lack of understanding regarding yields, safety, drug loading efficiencies, and efficacy of PEVs. In this study, various methods were compared to generate PEVs from clinical-grade platelets, and their properties were examined as vehicles for doxorubicin (DOX). Sonication and extrusion produced the most PEVs, with means of 496 and 493 PEVs per platelet (PLT), respectively, compared to 145 and 33 by freeze/thaw and incubation, respectively. The PEVs were loaded with DOX through incubation and purified by chromatography. The size and concentration of the PEVs and PEV-DOX were analyzed using dynamic light scattering and nanoparticle tracking analysis. The results showed that the population sizes and concentrations of PEVs and PEV-DOX were in the ranges of 120-150 nm and 1.2-6.2 × 1011 particles/mL for all preparations. The loading of DOX determined using fluorospectrometry was found to be 2.1 × 106, 1.7 × 106, and 0.9 × 106 molecules/EV using freeze/thaw, extrusion, and sonication, respectively. The internalization of PEVs was determined to occur through clathrin-mediated endocytosis. PEV-DOX were more efficiently taken up by MDA-MB-231 breast cancer cells compared to MCF7/ADR breast cancer cells and NIH/3T3 cells. DOX-PEVs showed higher anticancer activity against MDA-MB-231 cells than against MCF7/ADR or NIH/3T3 cells and better than acommercial liposomal DOX formulation. In conclusion, this study demonstrates that PEVs generated by PLTs using extrusion, freeze/thaw, or sonication can efficiently load DOX and kill breast cancer cells, providing a promising strategy for further evaluation in preclinical animal models. The study findings suggest that sonication and extrusion are the most efficient methods to generate PEVs and that PEVs loaded with DOX exhibit significant anticancer activity against MDA-MB-231 breast cancer cells.
Collapse
Affiliation(s)
- Yu-Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Deng-Yao Lee
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yeh-Lin Lu
- Core Facility Center, Office of Research and Development, Taipei Medical University, Taipei, Taiwan
| | - Liling Delila
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Lassina Barro
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Chun Austin Changou
- Core Facility Center, Office of Research and Development, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Translational Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- The Ph.D. Program for Cancer Biology and Drug Discovery, Center for Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Long-Sheng Lu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
- The Ph.D. Program for Cancer Biology and Drug Discovery, Center for Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
- Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hadi Goubran
- Saskatoon Cancer Centre and College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
- The Ph.D. Program for Cancer Biology and Drug Discovery, Center for Translational Medicine, Taipei Medical University, Taipei, Taiwan
- International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
48
|
Yoo J, Kwon I, Kim S, Kim HM, Kim YD, Nam HS, Heo JH. Coagulation Factor Expression and Composition of Arterial Thrombi in Cancer-Associated Stroke. Stroke 2023; 54:2981-2989. [PMID: 37886852 DOI: 10.1161/strokeaha.123.044910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/29/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Cancer is associated with an increased risk of stroke. Tumor cells activate platelets, induce a coagulation cascade, and generate thrombin. The composition of thrombi may reflect the mechanism of thrombosis, aiding the determination of the treatment strategy. Here, we investigated the composition and expression of coagulation factors in the thrombi of patients with cancer-associated stroke. METHODS Patients with stroke who underwent endovascular thrombectomy between September 2014 and June 2020 and whose cerebral thrombi were obtained were divided into those with cancer-associated stroke (cancer group) and propensity score-matched patients without cancer (control group), using 1:1 matching based on age and sex. Immunohistochemistry was performed of the thrombi, and the composition and expression of coagulation factors were compared between groups. RESULTS Among the 320 patients who underwent endovascular thrombectomy and who had thrombi obtained, this study included 23 patients with cancer and 23 matched controls. In both groups, the median age was 65 years, and 12 patients (52.2%) were men. Platelet composition was significantly higher in the cancer group than in the control group (median [interquartile range], 51.3% [28.0%-61.4%] versus 9.5% [4.8%-14.0%]; P<0.001). Among coagulation factors, thrombin (26.2% [16.2%-52.7%] versus 4.5% [1.3%-7.2%]; P<0.001) and tissue factors (0.60% [0.34%-2.06%] versus 0.37% [0.22%-0.60%]; P=0.024) were higher and factor X was lower (1.25% [0.39%-3.60%] versus 2.33% [1.67%-4.48%]; P=0.034) in the cancer group. There was a positive correlation between thrombin and platelets in the cancer group (r=0.666; P=0.001) but not in the control group (r=-0.167; P=0.627). CONCLUSIONS Cerebral thrombi in patients with cancer-associated stroke showed higher proportions of platelets, thrombin, and tissue factors, suggesting their key roles in arterial thrombosis in cancer and providing a therapeutic perspective for preventing stroke in patients with cancer-associated stroke.
Collapse
Affiliation(s)
- Joonsang Yoo
- Department of Neurology, Yongin Severance Hospital (J.Y.), Yonsei University College of Medicine, Korea
| | - Il Kwon
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Seoul, South Korea (I.K., S.K., Y.D.K., H.S.N., J.H.H.)
| | - Sungeun Kim
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Seoul, South Korea (I.K., S.K., Y.D.K., H.S.N., J.H.H.)
| | - Hye Min Kim
- Department of Pathology, Yongin Severance Hospital (H.M.K.), Yonsei University College of Medicine, Korea
| | - Young Dae Kim
- Department of Neurology, Severance Hospital (Y.D.K., H.S.N., J.H.H.), Yonsei University College of Medicine, Korea
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Seoul, South Korea (I.K., S.K., Y.D.K., H.S.N., J.H.H.)
| | - Hyo Suk Nam
- Department of Neurology, Severance Hospital (Y.D.K., H.S.N., J.H.H.), Yonsei University College of Medicine, Korea
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Seoul, South Korea (I.K., S.K., Y.D.K., H.S.N., J.H.H.)
| | - Ji Hoe Heo
- Department of Neurology, Severance Hospital (Y.D.K., H.S.N., J.H.H.), Yonsei University College of Medicine, Korea
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Seoul, South Korea (I.K., S.K., Y.D.K., H.S.N., J.H.H.)
| |
Collapse
|
49
|
Guo S, Huang J, Li G, Chen W, Li Z, Lei J. The role of extracellular vesicles in circulating tumor cell-mediated distant metastasis. Mol Cancer 2023; 22:193. [PMID: 38037077 PMCID: PMC10688140 DOI: 10.1186/s12943-023-01909-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023] Open
Abstract
Current research has demonstrated that extracellular vesicles (EVs) and circulating tumor cells (CTCs) are very closely related in the process of distant tumor metastasis. Primary tumors are shed and released into the bloodstream to form CTCs that are referred to as seeds to colonize and grow in soil-like distant target organs, while EVs of tumor and nontumor origin act as fertilizers in the process of tumor metastasis. There is no previous text that provides a comprehensive review of the role of EVs on CTCs during tumor metastasis. In this paper, we reviewed the mechanisms of EVs on CTCs during tumor metastasis, including the ability of EVs to enhance the shedding of CTCs, protect CTCs in circulation and determine the direction of CTC metastasis, thus affecting the distant metastasis of tumors.
Collapse
Affiliation(s)
- Siyin Guo
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Huang
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Genpeng Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wenjie Chen
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jianyong Lei
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
50
|
Russo P, Marino F, Rossi F, Bizzarri FP, Ragonese M, Dibitetto F, Filomena GB, Marafon DP, Ciccarese C, Iacovelli R, Pandolfo SD, Aveta A, Cilio S, Napolitano L, Foschi N. Is Systemic Immune-Inflammation Index a Real Non-Invasive Biomarker to Predict Oncological Outcomes in Patients Eligible for Radical Cystectomy? MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2063. [PMID: 38138166 PMCID: PMC10744858 DOI: 10.3390/medicina59122063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/13/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023]
Abstract
Background and Objectives: To assess the potential prognostic role of the systemic immune-inflammation index (SII) in predicting oncological outcomes in a cohort of patients treated with radical cystectomy (RC). Materials and Methods: From 2016 to 2022, a retrospective monocentric study enrolled 193 patients who were divided into two groups based on their SII levels using the optimal cutoff determined by the Youden index. The SII was obtained from a preoperative blood test approximately one month before RC. Univariable and multivariable logistic regression analyses were conducted to investigate the capacity of SII to predict lymph node invasion (N), advanced pT stage (pT3/pT4), and locally advanced condition at the time of RC. Multivariable Cox regression models adjusted for preoperative and postoperative features were used to analyze the prognostic effect of SII on recurrence-free survival (RFS), cancer-specific survival (CSS), and overall survival (OS). Results: The optimal cutoff value of the SII was 640.27. An elevated SII was seen in 113 (58.5%) patients. Using the multivariable preoperative logistic regression models, an elevated SII was correlated with nodal invasion (N; p = 0.03), advanced pT stage (p = 0.04), and locally advanced disease (p = 0.005), with enhancement of AUCs for predicting locally advanced disease (p = 0.04). In multivariable Cox regression models that considered preoperative clinicopathologic factors, an elevated SII was linked to poorer RFS (p = 0.005) and OS (p = 0.01). Moreover, on multivariable Cox regression postoperative models, a high SII was linked to RFS (p = 0.004) and to OS (p = 0.01). Conclusions: In this monocentric retrospective study, higher preoperative SII values predicted worse oncological outcomes in patients with bladder cancer (BCa) who underwent RC.
Collapse
Affiliation(s)
- Pierluigi Russo
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168 Rome, Italy or (P.R.); (F.R.); (F.P.B.); (M.R.); (F.D.); (G.B.F.); (N.F.)
- Department of Urology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Filippo Marino
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168 Rome, Italy or (P.R.); (F.R.); (F.P.B.); (M.R.); (F.D.); (G.B.F.); (N.F.)
| | - Francesco Rossi
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168 Rome, Italy or (P.R.); (F.R.); (F.P.B.); (M.R.); (F.D.); (G.B.F.); (N.F.)
| | - Francesco Pio Bizzarri
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168 Rome, Italy or (P.R.); (F.R.); (F.P.B.); (M.R.); (F.D.); (G.B.F.); (N.F.)
| | - Mauro Ragonese
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168 Rome, Italy or (P.R.); (F.R.); (F.P.B.); (M.R.); (F.D.); (G.B.F.); (N.F.)
| | - Francesco Dibitetto
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168 Rome, Italy or (P.R.); (F.R.); (F.P.B.); (M.R.); (F.D.); (G.B.F.); (N.F.)
| | - Giovanni Battista Filomena
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168 Rome, Italy or (P.R.); (F.R.); (F.P.B.); (M.R.); (F.D.); (G.B.F.); (N.F.)
| | - Denise Pires Marafon
- Section of Hygiene, Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 20123 Milano, Italy
| | - Chiara Ciccarese
- Department of Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168 Rome, Italy; (C.C.); (R.I.)
| | - Roberto Iacovelli
- Department of Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168 Rome, Italy; (C.C.); (R.I.)
| | - Savio Domenico Pandolfo
- Division of Urology, AORN “San Giuseppe Moscati”, 83100 Avellino, Italy; (S.D.P.); (A.A.); (S.C.); (L.N.)
| | - Achille Aveta
- Division of Urology, AORN “San Giuseppe Moscati”, 83100 Avellino, Italy; (S.D.P.); (A.A.); (S.C.); (L.N.)
| | - Simone Cilio
- Division of Urology, AORN “San Giuseppe Moscati”, 83100 Avellino, Italy; (S.D.P.); (A.A.); (S.C.); (L.N.)
| | - Luigi Napolitano
- Division of Urology, AORN “San Giuseppe Moscati”, 83100 Avellino, Italy; (S.D.P.); (A.A.); (S.C.); (L.N.)
| | - Nazario Foschi
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1, 00168 Rome, Italy or (P.R.); (F.R.); (F.P.B.); (M.R.); (F.D.); (G.B.F.); (N.F.)
| |
Collapse
|