1
|
Zhang L, Pan J, Wang M, Yang J, Zhu S, Li L, Hu X, Wang Z, Pang L, Li P, Jia F, Ren G, Zhang Y, Xu D, Qiu F, Huang J. Chronic Stress-Induced and Tumor Derived SP1 + Exosomes Polarizing IL-1β + Neutrophils to Increase Lung Metastasis of Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2310266. [PMID: 39630109 PMCID: PMC11789585 DOI: 10.1002/advs.202310266] [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: 12/28/2023] [Revised: 11/07/2024] [Indexed: 01/30/2025]
Abstract
Chronic stress can significantly promote breast cancer progression. When exposed to chronic stress, exosomes released from neural and neuroendocrine cells in the central nervous system are enhanced and modified. However, whether tumor-derived exosomes (TDEs) are influenced by chronic stress and participate in chronic stress-mediated distant metastasis remains unclear. Here, it is shown that chronic stress remarkably facilitates the secretion of TDEs and modifies the contents of exosomes by activating the adrenergic β receptor in 4T1 tumor-bearing mice. Exosomes injection and blockade experiments indicate that exosomes play a crucial role in chronic stress-mediated lung metastasis of breast cancer. Chronic stress-induced TDEs are internalized by pulmonary neutrophils and strengthen neutrophil recruitment via the CXCL2 autocrine. In addition, the level of SP1 in TDEs increases, which favors the secretion of IL-1β by neutrophils through the activation of the TLR4-NFκβ pathway, ultimately aggravating lung metastasis of breast cancer. Collectively, this study provides a novel mechanism by which neutrophils within a pre-metastatic niche acquire their inflamed phenotype and establishes an important link among neuroendocrine changes, exosomes, immunity, and metastasis.
Collapse
|
2
|
Sakaue T, Dorayappan KDP, Zingarelli R, Khadraoui W, Anbalagan M, Wallbillich J, Bognar B, Wanner R, Cosgrove C, Suarez A, Koga H, Maxwell GL, O'Malley DM, Cohn DE, Selvendiran K. Obesity-induced extracellular vesicles proteins drive the endometrial cancer pathogenesis: therapeutic potential of HO-3867 and Metformin. Oncogene 2024; 43:3586-3597. [PMID: 39414985 PMCID: PMC11602708 DOI: 10.1038/s41388-024-03182-2] [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: 01/16/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/18/2024]
Abstract
Endometrial cancer (EC) is the leading gynecologic malignancy in the United States with obesity implicated in 57% of cases. This research investigates the molecular complexities of extracellular vesicles (EV) secretion as carriers of oncogenic protein and their involvement in obesity-mediated EC. An understanding of these mechanisms is pivotal for unraveling pathways relevant to obesity-associated EC, thereby guiding the development of innovative prevention and treatment strategies. Our exploration revealed a significant increase in EV secretion carrying oncogenic proteins (TMEM205, STAT5, and FAS) in adipose and uterine tissues/serum samples from obese EC patients compared to control (without cancer). We identified alterations in EV-regulating proteins (Rab7, Rab11, and Rab27a) in obesity-mediated EC patients, adipose/uterine tissues, and serum samples. Through a 24-week analysis of the effects of a 45% kcal high-fat diet (HFD) on mice, we observed increased body weight, increased adipose tissue, enlarged uterine horns, and increased inflammation in the HFD group. This correlated with elevated levels of EV secretion and increased expression of oncogenic proteins TMEM205, FAS, and STAT5 and downregulation of the tumor suppressor gene PIAS3 in adipose and uterine tissues. Furthermore, our study confirmed that adipocyte derived EV increased EC cell proliferation, migration and xenograft tumor growth. Additionally, we identified that the small molecule inhibitors (HO-3867) or Metformin inhibited EV secretion in vitro and in vivo, demonstrating significant inhibition of high glucose or adipocyte-mediated EC cell proliferation and a reduction in body weight and adipose tissue accumulation when administered to HFD mice. Moreover, HO-3867 or Metformin treatment inhibited HFD induced hyperplasia (precursor of EC) by altering the expression of EV-regulated proteins and decreasing oncogenic protein expression levels. This study provides critical insights into the mechanisms underpinning obesity-mediated EV secretion with oncogenic protein expression, shedding light on their role in EC pathogenesis. Additionally, it offers pre-clinical evidence supporting the initiation of novel studies for EV-targeted therapies aimed at preventing obesity-mediated EC.
Collapse
Affiliation(s)
- Takahiko Sakaue
- Division of GYN/ONC, The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | | | - Roman Zingarelli
- Division of GYN/ONC, The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - Wafa Khadraoui
- Division of GYN/ONC, The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | | | - John Wallbillich
- Division of GYN/ONC, The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - Balazs Bognar
- Institute of Organic and Medicinal Chemistry, Medical School, University of Pécs, Pécs, Hungary
| | - Ross Wanner
- Division of GYN/ONC, The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - Casey Cosgrove
- Division of GYN/ONC, The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - Adrian Suarez
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - G Larry Maxwell
- Inova Women's Service Line and the Inova Schar Cancer Institute, Falls Church, VA, USA
| | - David M O'Malley
- Division of GYN/ONC, The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - David E Cohn
- Division of GYN/ONC, The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - Karuppaiyah Selvendiran
- Division of GYN/ONC, The James Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
3
|
Wagner KT, Lu RXZ, Landau S, Shawky SA, Zhao Y, Bodenstein DF, Jiménez Vargas LF, Jiang R, Okhovatian S, Wang Y, Liu C, Vosoughi D, Gustafson D, Fish JE, Cummins CL, Radisic M. Endothelial extracellular vesicles enhance vascular self-assembly in engineered human cardiac tissues. Biofabrication 2024; 16:045037. [PMID: 39226913 PMCID: PMC11409464 DOI: 10.1088/1758-5090/ad76d9] [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: 01/08/2024] [Accepted: 09/03/2024] [Indexed: 09/05/2024]
Abstract
The fabrication of complex and stable vasculature in engineered cardiac tissues represents a significant hurdle towards building physiologically relevant models of the heart. Here, we implemented a 3D model of cardiac vasculogenesis, incorporating endothelial cells (EC), stromal cells, and human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) in a fibrin hydrogel. The presence of CMs disrupted vessel formation in 3D tissues, resulting in the upregulation of endothelial activation markers and altered extracellular vesicle (EV) signaling in engineered tissues as determined by the proteomic analysis of culture supernatant. miRNA sequencing of CM- and EC-secreted EVs highlighted key EV-miRNAs that were postulated to play differing roles in cardiac vasculogenesis, including the let-7 family and miR-126-3p in EC-EVs. In the absence of CMs, the supplementation of CM-EVs to EC monolayers attenuated EC migration and proliferation and resulted in shorter and more discontinuous self-assembling vessels when applied to 3D vascular tissues. In contrast, supplementation of EC-EVs to the tissue culture media of 3D vascularized cardiac tissues mitigated some of the deleterious effects of CMs on vascular self-assembly, enhancing the average length and continuity of vessel tubes that formed in the presence of CMs. Direct transfection validated the effects of the key EC-EV miRNAs let-7b-5p and miR-126-3p in improving the maintenance of continuous vascular networks. EC-EV supplementation to biofabricated cardiac tissues and microfluidic devices resulted in tissue vascularization, illustrating the use of this approach in the engineering of enhanced, perfusable, microfluidic models of the myocardium.
Collapse
Affiliation(s)
- Karl T Wagner
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Rick X Z Lu
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Shira Landau
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Sarah A Shawky
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, ON M5S 3M2, Canada
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Acceleration Consortium, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - David F Bodenstein
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5G 2C8, Canada
| | - Luis Felipe Jiménez Vargas
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Richard Jiang
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Chuan Liu
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Daniel Vosoughi
- Latner Thoracic Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Dakota Gustafson
- Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital,University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, ON M5S 3M2, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
- Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 27 King's College Circle, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
4
|
Bernáth-Nagy D, Kalinyaprak MS, Giannitsis E, Ábrahám P, Leuschner F, Frey N, Krohn JB. Circulating extracellular vesicles as biomarkers in the diagnosis, prognosis and therapy of cardiovascular diseases. Front Cardiovasc Med 2024; 11:1425159. [PMID: 39314768 PMCID: PMC11417624 DOI: 10.3389/fcvm.2024.1425159] [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: 04/29/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Cardiovascular disease (CVD) ranks among the primary contributors to worldwide mortality. Hence, the importance of constant research on new circulating biomarkers for the improvement of early diagnosis and prognostication of different CVDs and the development and refinement of therapeutic measures is critical. Extracellular vesicles (EV) have a great potential as diagnostic and prognostic markers, as they represent their parent cell by enclosing cell-specific molecules, which can differ in quality and quantity based on cell state. Assuming that all cell types of the cardiovascular system are capable of releasing EV into circulation, an emerging body of evidence has investigated the potential role of serum- or plasma-derived EV in CVD. Comprehensive research has unveiled alterations in EV quantity and EV-bound cargo in the form of RNA, proteins and lipids in the context of common CVDs such as coronary artery disease, atrial fibrillation, heart failure or inflammatory heart diseases, highlighting their diagnostic and prognostic relevance. In numerous in vitro and in vivo models, EV also showed promising therapeutic potential. However, translation of EV studies to a preclinical or clinical setting has proven to be challenging. This review is intended to provide an overview of the most relevant studies in the field of serum or plasma-derived EV.
Collapse
Affiliation(s)
- Dominika Bernáth-Nagy
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Melek Sükran Kalinyaprak
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Evangelos Giannitsis
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Pál Ábrahám
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - Florian Leuschner
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Jona Benjamin Krohn
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
5
|
Fathi E, Valipour B, Jafari S, Kazemi A, Montazersaheb S, Farahzadi R. The role of the hematopoietic stem/progenitor cells-derived extracellular vesicles in hematopoiesis. Heliyon 2024; 10:e35051. [PMID: 39157371 PMCID: PMC11327835 DOI: 10.1016/j.heliyon.2024.e35051] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are tightly regulated by specific microenvironments called niches to produce an appropriate number of mature blood cell types. Self-renewal and differentiation are two hallmarks of hematopoietic stem and progenitor cells, and their balance is critical for proper functioning of blood and immune cells throughout life. In addition to cell-intrinsic regulation, extrinsic cues within the bone marrow niche and systemic factors also affect the fate of HSCs. Despite this, many paracrine and endocrine factors that influence the function of hematopoietic cells remain unknown. In hematological malignancies, malignant cells remodel their niche into a permissive environment to enhance the survival of leukemic cells. These events are accompanied by loss of normal hematopoiesis. It is well known that extracellular vehicles (EVs) mediate intracellular interactions under physiological and pathological conditions. In other words, EVs transfer biological information to surrounding cells and contribute not only to physiological functions but also to the pathogenesis of some diseases, such as cancers. Therefore, a better understanding of cell-to-cell interactions may lead to identification of potential therapeutic targets. Recent reports have suggested that EVs are evolutionarily conserved constitutive mediators that regulate hematopoiesis. Here, we focus on the emerging roles of EVs in normal and pathological conditions, particularly in hematological malignancies. Owing to the high abundance of EVs in biological fluids, their potential use as biomarkers and therapeutic tools is discussed.
Collapse
Affiliation(s)
- Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Behnaz Valipour
- Department of Basic Sciences and Health, Sarab Faculty of Medical Sciences, Sarab, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abdolhassan Kazemi
- Medical Philosophy and History Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Hordijk S, Carter T, Bierings R. A new look at an old body: molecular determinants of Weibel-Palade body composition and von Willebrand factor exocytosis. J Thromb Haemost 2024; 22:1290-1303. [PMID: 38307391 DOI: 10.1016/j.jtha.2024.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/04/2024]
Abstract
Endothelial cells, forming a monolayer along blood vessels, intricately regulate vascular hemostasis, inflammatory responses, and angiogenesis. A key determinant of these functions is the controlled secretion of Weibel-Palade bodies (WPBs), which are specialized endothelial storage organelles housing a presynthesized pool of the hemostatic protein von Willebrand factor and various other hemostatic, inflammatory, angiogenic, and vasoactive mediators. This review delves into recent mechanistic insights into WPB biology, including the biogenesis that results in their unique morphology, the acquisition of intraluminal vesicles and other cargo, and the contribution of proton pumps to organelle acidification. Additionally, in light of a number of proteomic approaches to unravel the regulatory networks that control WPB formation and secretion, we provide a comprehensive overview of the WPB exocytotic machinery, including their molecular and cellular mechanisms.
Collapse
Affiliation(s)
- Sophie Hordijk
- Hematology, Erasmus MC University Medical Center, Rotterdam, The Netherlands. https://twitter.com/SophieHordijk
| | - Tom Carter
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Ruben Bierings
- Hematology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
Morimoto M, Maishi N, Hida K. Acquisition of drug resistance in endothelial cells by tumor-derived extracellular vesicles and cancer progression. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:1. [PMID: 38318528 PMCID: PMC10838380 DOI: 10.20517/cdr.2023.121] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/17/2023] [Indexed: 02/07/2024]
Abstract
Angiogenesis by endothelial cells (ECs) is essential for tumor growth. Angiogenesis inhibitors are used in combination with anticancer drugs in many tumor types, but tumors eventually become resistant. Previously, the underlying mechanism for developing drug resistance was considered to be a change in the characteristics of tumor cells whereas ECs were thought to be genetically stable and do not contribute to drug resistance. However, tumor endothelial cells (TECs) have been shown to differ from normal endothelial cells (NECs) in that they exhibit chromosomal abnormalities, angiogenic potential, and drug resistance. Extracellular vesicles (EVs) secreted by tumor cells have recently attracted attention as a factor involved in the acquisition of such abnormalities. Various cells communicate with each other through EVs, and it has been reported that tumor-derived EVs act on other tumor cells or stromal cells to develop drug resistance. Drug-resistant tumor cells confer drug resistance to recipient cells by transporting mRNAs encoding ATP-binding cassette subfamily B member 1 (ABCB1) and ATP-binding cassette subfamily C member 1 (ABCC1) as well as miRNAs involved in signaling such as Akt, drug efflux transporters, and P-glycoprotein modulators via EVs. However, there are limited reports on the acquisition of drug resistance in ECs by tumor-derived EVs. Since drug resistance of ECs may induce tumor metastasis and support tumor cell proliferation, the mechanism underlying the development of resistance should be elucidated to find therapeutic application. This review provides insight into the acquisition of drug resistance in ECs via tumor EVs in the tumor microenvironment.
Collapse
Affiliation(s)
- Masahiro Morimoto
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo 060-8586, Japan
- Department of Oral Diagnosis and Medicine, Hokkaido University Faculty of Dental Medicine, Sapporo 060-8586, Japan
| | - Nako Maishi
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo 060-8586, Japan
| | - Kyoko Hida
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo 060-8586, Japan
| |
Collapse
|
8
|
Beylerli O, Tamrazov R, Gareev I, Ilyasova T, Shumadalova A, Bai Y, Yang B. Role of exosomal ncRNAs in traumatic brain injury. Noncoding RNA Res 2023; 8:686-692. [PMID: 37860267 PMCID: PMC10582766 DOI: 10.1016/j.ncrna.2023.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/22/2023] [Accepted: 10/07/2023] [Indexed: 10/21/2023] Open
Abstract
Traumatic brain injury (TBI) is a complex neurological disorder that often results in long-term disabilities, cognitive impairments, and emotional disturbances. Despite significant advancements in understanding the pathophysiology of TBI, effective treatments remain limited. In recent years, exosomal non-coding RNAs (ncRNAs) have emerged as potential players in TBI pathogenesis and as novel diagnostic and therapeutic targets. Exosomal ncRNAs are small RNA molecules that are secreted by cells and transported to distant sites, where they can modulate gene expression and cell signaling pathways. They have been shown to play important roles in various aspects of TBI, such as neuroinflammation, blood-brain barrier dysfunction, and neuronal apoptosis. The ability of exosomal ncRNAs to cross the blood-brain barrier and reach the brain parenchyma makes them attractive candidates for non-invasive biomarkers and drug delivery systems. However, significant challenges still need to be addressed before exosomal ncRNAs can be translated into clinical practice, including standardization of isolation and quantification methods, validation of their diagnostic and prognostic value, and optimization of their therapeutic efficacy and safety. This review aims to summarize the current knowledge regarding the role of exosomal ncRNAs in TBI, including their biogenesis, function, and potential applications in diagnosis, prognosis, and treatment. We also discuss the challenges and future perspectives of using exosomal ncRNAs as clinical tools for TBI management.
Collapse
Affiliation(s)
- Ozal Beylerli
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Rasim Tamrazov
- Department of Oncology, Radiology and Radiotherapy, Tyumen State Medical University, 54 Odesskaya Street, 625023, Tyumen, Russia
| | - Ilgiz Gareev
- Central Research Laboratory, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 3 Lenin Street, 450008, Russia
| | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Alina Shumadalova
- Department of General Chemistry, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 3 Lenin Street, 450008, Russia
| | - Yunlong Bai
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, People's Republic of China
| |
Collapse
|
9
|
Yoo S, Choi S, Kim I, Kim IS. Hypoxic regulation of extracellular vesicles: Implications for cancer therapy. J Control Release 2023; 363:201-220. [PMID: 37739015 DOI: 10.1016/j.jconrel.2023.09.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/18/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Extracellular vesicles (EVs) play a pivotal role in intercellular communication and have been implicated in cancer progression. Hypoxia, a pervasive hallmark of cancer, is known to regulate EV biogenesis and function. Hypoxic EVs contain a specific set of proteins, nucleic acids, lipids, and metabolites, capable of reprogramming the biology and fate of recipient cells. Enhancing the intrinsic therapeutic efficacy of EVs can be achieved by strategically modifying their structure and contents. Moreover, the use of EVs as drug delivery vehicles holds great promise for cancer treatment. However, various hurdles must be overcome to enable their clinical application as cancer therapeutics. In this review, we aim to discuss the current knowledge on the hypoxic regulation of EVs. Additionally, we will describe the underlying mechanisms by which EVs contribute to cancer progression in hypoxia and outline the progress and limitations of hypoxia-related EV therapeutics for cancer.
Collapse
Affiliation(s)
- Seongkyeong Yoo
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea
| | - Sanga Choi
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea
| | - Iljin Kim
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea; Chemical and Biological Integrative Research Center, Biomedical Research Institute, Korea Institute Science and Technology, Seoul 02792, South Korea.
| |
Collapse
|
10
|
Montanari M, Guescini M, Gundogdu O, Luchetti F, Lanuti P, Ciacci C, Burattini S, Campana R, Ortolani C, Papa S, Canonico B. Extracellular Vesicles from Campylobacter jejuni CDT-Treated Caco-2 Cells Inhibit Proliferation of Tumour Intestinal Caco-2 Cells and Myeloid U937 Cells: Detailing the Global Cell Response for Potential Application in Anti-Tumour Strategies. Int J Mol Sci 2022; 24:ijms24010487. [PMID: 36613943 PMCID: PMC9820799 DOI: 10.3390/ijms24010487] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/15/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
Cytolethal distending toxin (CDT) is produced by a range of Gram-negative pathogenic bacteria such as Campylobacter jejuni. CDT represents an important virulence factor that is a heterotrimeric complex composed of CdtA, CdtB, and CdtC. CdtA and CdtC constitute regulatory subunits whilst CdtB acts as the catalytic subunit exhibiting phosphatase and DNase activities, resulting in cell cycle arrest and cell death. Extracellular vesicle (EV) secretion is an evolutionarily conserved process that is present throughout all kingdoms. Mammalian EVs play important roles in regular cell-to-cell communications but can also spread pathogen- and host-derived molecules during infections to alter immune responses. Here, we demonstrate that CDT targets the endo-lysosomal compartment, partially evading lysosomal degradation and exploiting unconventional secretion (EV release), which is largely involved in bacterial infections. CDT-like effects are transferred by Caco-2 cells to uninfected heterologous U937 and homologous Caco-2 cells. The journey of EVs derived from CDT-treated Caco-2 cells is associated with both intestinal and myeloid tumour cells. EV release represents the primary route of CDT dissemination, revealing an active toxin as part of the cargo. We demonstrated that bacterial toxins could represent suitable tools in cancer therapy, highlighting both the benefits and limitations. The global cell response involves a moderate induction of apoptosis and autophagic features may play a protective role against toxin-induced cell death. EVs from CDT-treated Caco-2 cells represent reliable CDT carriers, potentially suitable in colorectal cancer treatments. Our data present a potential bacterial-related biotherapeutic supporting a multidrug anticancer protocol.
Collapse
Affiliation(s)
- Mariele Montanari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Michele Guescini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Caterina Ciacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Sabrina Burattini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Raffaella Campana
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Claudio Ortolani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Stefano Papa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence:
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| |
Collapse
|
11
|
Iskrzak J, Zygmunciak P, Misiewicz-Krzemińska I, Puła B. Extracellular Vesicles in Multiple Myeloma-Cracking the Code to a Better Understanding of the Disease. Cancers (Basel) 2022; 14:cancers14225575. [PMID: 36428668 PMCID: PMC9688731 DOI: 10.3390/cancers14225575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell-derived malignancy that stands for around 1.5% of newly discovered cancer cases. Despite constantly improving treatment methods, the disease is incurable with over 13,000 deaths in the US and over 30,000 in Europe. Recent studies suggest that extracellular vesicles (EVs) might play a significant role in the pathogenesis and evolution of MM. Further investigation of their role could prove to be beneficial in establishing new therapies and hence, improve the prognosis of MM patients. What is more, EVs might serve as novel markers in diagnosing and monitoring the disease. Great advancements concerning the position of EVs in the pathophysiology of MM have recently been shown in research and in this review, we would like to delve into the still expanding state of knowledge.
Collapse
Affiliation(s)
- Justyna Iskrzak
- Medical University of Warsaw, 02-091 Warsaw, Poland
- Institute of Hematology and Transfusion Medicine, Indira Gandhi Str. 14, 02-776 Warsaw, Poland
| | - Przemysław Zygmunciak
- Medical University of Warsaw, 02-091 Warsaw, Poland
- Institute of Hematology and Transfusion Medicine, Indira Gandhi Str. 14, 02-776 Warsaw, Poland
| | - Irena Misiewicz-Krzemińska
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Chocimska Str. 5, 00-791 Warsaw, Poland
| | - Bartosz Puła
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Indira Gandhi Str. 14, 02-776 Warsaw, Poland
- Correspondence: ; Tel.: +48-223-496-302; Fax: +48-223-496-335
| |
Collapse
|
12
|
Kugeratski FG, Santi A, Zanivan S. Extracellular vesicles as central regulators of blood vessel function in cancer. Sci Signal 2022; 15:eaaz4742. [PMID: 36166511 DOI: 10.1126/scisignal.aaz4742] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Blood vessels deliver oxygen and nutrients that sustain tumor growth and enable the dissemination of cancer cells to distant sites and the recruitment of intratumoral immune cells. In addition, the structural and functional abnormalities of the tumor vasculature foster the development of an aggressive tumor microenvironment and impair the efficacy of existing cancer therapies. Extracellular vesicles (EVs) have emerged as major players of tumor progression, and a growing body of evidence has demonstrated that EVs derived from cancer cells trigger multiple responses in endothelial cells that alter blood vessel function in tumors. EV-mediated signaling in endothelial cells can occur through the transfer of functional cargos such as miRNAs, lncRNAs, cirRNAs, and proteins. Moreover, membrane-bound proteins in EVs can elicit receptor-mediated signaling in endothelial cells. Together, these mechanisms reprogram endothelial cells and contribute to the sustained exacerbated angiogenic signaling typical of tumors, which, in turn, influences cancer progression. Targeting these angiogenesis-promoting EV-dependent mechanisms may offer additional strategies to normalize tumor vasculature. Here, we discuss the current knowledge pertaining to the contribution of cancer cell-derived EVs in mechanisms regulating blood vessel functions in tumors. Moreover, we discuss the translational opportunities in targeting the dysfunctional tumor vasculature using EVs and highlight the open questions in the field of EV biology that can be addressed using mass spectrometry-based proteomics analysis.
Collapse
Affiliation(s)
- Fernanda G Kugeratski
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Alice Santi
- Department of Experimental and Clinical Biomedical Sciences, Università degli Studi di Firenze, 50134 Firenze, Italy
| | - Sara Zanivan
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1QH, UK
| |
Collapse
|
13
|
Carter N, Mathiesen AH, Miller N, Brown M, Colunga Biancatelli RML, Catravas JD, Dobrian AD. Endothelial cell-derived extracellular vesicles impair the angiogenic response of coronary artery endothelial cells. Front Cardiovasc Med 2022; 9:923081. [PMID: 35928931 PMCID: PMC9343725 DOI: 10.3389/fcvm.2022.923081] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/28/2022] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease (CVD) is the most prominent cause of death of adults in the United States with coronary artery disease being the most common type of CVD. Following a myocardial event, the coronary endothelium plays an important role in the recovery of the ischemic myocardium. Specifically, endothelial cells (EC) must be able to elicit a robust angiogenic response necessary for tissue revascularization and repair. However, local or distant cues may prevent effective revascularization. Extracellular vesicles (EV) are produced by all cells and endothelium is a rich source of EVs that have access to the main circulation thereby potentially impacting local and distant tissue function. Systemic inflammation associated with conditions such as obesity as well as the acute inflammatory response elicited by a cardiac event can significantly increase the EV release by endothelium and alter their miRNA, protein or lipid cargo. Our laboratory has previously shown that EVs released by adipose tissue endothelial cells exposed to chronic inflammation have angiostatic effects on naïve adipose tissue EC in vitro. Whether the observed effect is specific to EVs from adipose tissue endothelium or is a more general feature of the endothelial EVs exposed to pro-inflammatory cues is currently unclear. The objective of this study was to investigate the angiostatic effects of EVs produced by EC from the coronary artery and adipose microvasculature exposed to pro-inflammatory cytokines (PIC) on naïve coronary artery EC. We have found that EVs from both EC sources have angiostatic effects on the coronary endothelium. EVs produced by cells in a pro-inflammatory environment reduced proliferation and barrier function of EC without impacting cellular senescence. Some of these functional effects could be attributed to the miRNA cargo of EVs. Several miRNAs such as miR-451, let-7, or miR-23a impact on multiple pathways responsible for proliferation, cellular permeability and angiogenesis. Collectively, our data suggests that EVs may compete with pro-angiogenic cues in the ischemic myocardium therefore slowing down the repair response. Acute treatments with inhibitors that prevent endogenous EV release immediately after an ischemic event may contribute to better efficacy of therapeutic approaches using functionalized exogenous EVs or other pro-angiogenic approaches.
Collapse
Affiliation(s)
- Nigeste Carter
- Department of Physiological Science, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Allison H. Mathiesen
- Department of Physiological Science, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Noel Miller
- Department of Physiological Science, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Michael Brown
- Department of Physiological Science, Eastern Virginia Medical School, Norfolk, VA, United States
| | | | - John D. Catravas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States
- School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, VA, United States
| | - Anca D. Dobrian
- Department of Physiological Science, Eastern Virginia Medical School, Norfolk, VA, United States
- *Correspondence: Anca D. Dobrian,
| |
Collapse
|
14
|
Li T, Wang B, Ding H, Chen S, Cheng W, Li Y, Wu X, Wang L, Jiang Y, Lu Z, Teng Y, Su S, Han X, Zhao M. Effect of Extracellular Vesicles From Multiple Cells on Vascular Smooth Muscle Cells in Atherosclerosis. Front Pharmacol 2022; 13:857331. [PMID: 35620296 PMCID: PMC9127356 DOI: 10.3389/fphar.2022.857331] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/05/2022] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis (AS)-related diseases are still the main cause of death in clinical patients. The phenotype switching, proliferation, migration, and secretion of vascular smooth muscle cells (VSMCs) have a pivotal role in atherosclerosis. Although numerous research studies have elucidated the role of VSMCs in AS, their potential functional regulations continue to be explored. The formation of AS involves various cells, such as endothelial cells, smooth muscle cells, and macrophages. Therefore, intercellular communication of blood vessels cannot be ignored due to closely connected endothelia, media, and adventitia. Extracellular vesicles (EVs), as the vectors of cell-to-cell communication, can deliver proteins and nucleic acids of parent cells to the recipient cells. EVs have emerged as being central in intercellular communication and play a vital role in the pathophysiologic mechanisms of AS. This review summarizes the effects of extracellular vesicles (EVs) derived from multiple cells (endothelial cells, macrophages, mesenchymal stem cells, etc.) on VSMCs in AS. The key findings of this review are as follows: 1) endothelial cell–derived EVs (EEVs) have anti- or pro-atherogenic effects on VSMCs; 2) macrophage-derived EVs (MEVs) aggravate the proliferation and migration of VSMCs; 3) mesenchymal stem cells can inhibit VSMCs; and 4) the proliferation and migration of VSMCs can be inhibited by the treatment of EVs with atherosclerosis-protective factors and promoted by noxious stimulants. These results suggested that EVs have the same functional properties as treated parent cells, which might provide vital guidance for treating AS.
Collapse
Affiliation(s)
- Tong Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Baofu Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hao Ding
- Department of Oncology, Shanxi Traditional Chinese Medical Hospital, Taiyuan, China
| | - Shiqi Chen
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weiting Cheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxiao Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yangyang Jiang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ziwen Lu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Sha Su
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowan Han
- Department of Cardiac Rehabilitation, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Stratman AN, Crewe C, Stahl PD. The microenvironment‐ a general hypothesis on the homeostatic function of extracellular vesicles. FASEB Bioadv 2022; 4:284-297. [PMID: 35520390 PMCID: PMC9065581 DOI: 10.1096/fba.2021-00155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 11/23/2022] Open
Abstract
Extracellular vesicles (EVs), exosomes and microvesicles, is a burgeoning field of biological and biomedical research that may change our understanding of cell communication in plants and animals while holding great promise for the diagnosis of disease and the development of therapeutics. However, the challenge remains to develop a general hypothesis about the role of EVs in physiological homeostasis and pathobiology across kingdoms. While they can act systemically, EVs are often seen to operate locally within a microenvironment. This microenvironment is built as a collection of microunits comprised of cells that interact with each other via EV exchange, EV signaling, EV seeding, and EV disposal. We propose that microunits are part of a larger matrix at the tissue level that collectively communicates with the surrounding environment, including other end‐organ systems. Herein, we offer a working model that encompasses the various facets of EV function in the context of the cell biology and physiology of multicellular organisms.
Collapse
Affiliation(s)
- Amber N Stratman
- Department of Cell Biology and Physiology Washington University School of Medicine 660 South Euclid Avenue St. Louis Missouri USA 63110
| | - Clair Crewe
- Department of Cell Biology and Physiology Washington University School of Medicine 660 South Euclid Avenue St. Louis Missouri USA 63110
- Department of Internal Medicine Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine 660 South Euclid Avenue St. Louis Missouri USA 63110
| | - Philip D Stahl
- Department of Cell Biology and Physiology Washington University School of Medicine 660 South Euclid Avenue St. Louis Missouri USA 63110
| |
Collapse
|
16
|
Floriano JF, Emanueli C, Vega S, Barbosa AMP, Oliveira RGD, Floriano EAF, Graeff CFDO, Abbade JF, Herculano RD, Sobrevia L, Rudge MVC. Pro-angiogenic approach for skeletal muscle regeneration. Biochim Biophys Acta Gen Subj 2022; 1866:130059. [PMID: 34793875 DOI: 10.1016/j.bbagen.2021.130059] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/01/2021] [Indexed: 12/19/2022]
Abstract
The angiogenesis process is a phenomenon in which numerous molecules participate in the stimulation of the new vessels' formation from pre-existing vessels. Angiogenesis is a crucial step in tissue regeneration and recovery of organ and tissue function. Muscle diseases affect millions of people worldwide overcome the ability of skeletal muscle to self-repair. Pro-angiogenic therapies are key in skeletal muscle regeneration where both myogenesis and angiogenesis occur. These therapies have been based on mesenchymal stem cells (MSCs), exosomes, microRNAs (miRs) and delivery of biological factors. The use of different calls of biomaterials is another approach, including ceramics, composites, and polymers. Natural polymers are use due its bioactivity and biocompatibility in addition to its use as scaffolds and in drug delivery systems. One of these polymers is the natural rubber latex (NRL) which is biocompatible, bioactive, versatile, low-costing, and capable of promoting tissue regeneration and angiogenesis. In this review, the advances in the field of pro-angiogenic therapies are discussed.
Collapse
Affiliation(s)
- Juliana Ferreira Floriano
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; National Heart and Lung Institute, Imperial College London, London, UK.
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sofia Vega
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | | | | | | | | | - Joelcio Francisco Abbade
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil
| | | | - Luis Sobrevia
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland, Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD, 4029, Queensland, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands.
| | | |
Collapse
|
17
|
Exosomes in cardiovascular diseases: a blessing or a sin for the mankind. Mol Cell Biochem 2022; 477:833-847. [PMID: 35064412 DOI: 10.1007/s11010-021-04328-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022]
Abstract
Cardiovascular diseases (CVDs) comprises disorders of blood vessels and heart. Multiple cells in the heart suggests that hetero-cellular communication, which is an important aspect in heart functioning and there is a need to elucidate the way in which this inter-cellular communication occurs. Now a days, exosomal research has gained much attention. Exosomes, nano-shuttles, are EVs with diameters ranging from 40 to 160 nm (average 100 nm), secreted by body cells. These vesicles act as cell-to-cell communicators and are carriers of important biomolecules such as RNAs, miRNAs, Proteins and lipids. Exosomes can change the gene expression of the recipient cells, thereby, changes the cellular characteristics. Exosomes have known to play an essential role in protection as well as progression of various cardiovascular diseases. In the present review, role of exosomes in various CVDs have been discussed.
Collapse
|
18
|
Li D, Tang J, Gao R, Lan J, Shen W, Liu Y, Chen Y, Sun H, Yan J, Nie Y, Luo N. PFKFB4 promotes angiogenesis via IL-6/STAT5A/P-STAT5 signaling in breast cancer. J Cancer 2022; 13:212-224. [PMID: 34976184 PMCID: PMC8692682 DOI: 10.7150/jca.66773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/10/2021] [Indexed: 01/16/2023] Open
Abstract
Breast cancer has become the most newly-diagnosed cancer and the 5th leading cause of cancer death worldwide. The 5-year survival rate of breast cancer is about 90%. However, the 5-year survival rate drops to <30% when metastasis to distant sites occurs. The blood vessel formation (i.e., angiogenesis) plays a crucial role during the metastatic process. In this study, we investigated the role of PFKFB4 in angiogenesis of breast cancer. Employing in vitro HUVEC tube formation or in vivo orthotopic mouse model, and gene editing or specific small inhibitors strategy, and utilizing qPCR, western blot, ELISA, or immunofluorescent/immunohistochemistry staining methods, we found the following: 1) PFKFB4 upregulates IL-6 expression via NF-κB signaling in breast cancer cells; 2) PFKFB4-induced lactate secretion contributes to NF-κB activation in breast cancer cells; 3) IL-6 elicits angiogenesis via STAT5A/P-STAT5 in HUVEC; 4) 5-MPN (a specific PFKFB4 inhibitor) suppresses angiogenesis in vitro and in vivo. Our findings suggest a potential strategy whereby 5-MPN may lead to an improved therapeutic outcome for breast cancer patients.
Collapse
Affiliation(s)
- Dan Li
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jiaping Tang
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Ruifang Gao
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
- Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin 300131, China
| | - Jinxin Lan
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Wenzhi Shen
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| | - Yanhua Liu
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Nankai University, Tianjin 300071, China
| | - Yanan Chen
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Nankai University, Tianjin 300071, China
| | - Hongwei Sun
- Experimental Center of Operations, Chinese People's Armed Police Force Command College, Tianjin 300250, China
| | - Jie Yan
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Nankai University, Tianjin 300071, China
| | - Yongwei Nie
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Na Luo
- Department of Anatomy and Histology, School of Medicine, Nankai University, Tianjin 300071, China
- Tianjin Key Laboratory of Tumour Microenvironment and Neurovascular Regulation, Nankai University, Tianjin 300071, China
| |
Collapse
|
19
|
Xu H, Ni YQ, Liu YS. Mechanisms of Action of MiRNAs and LncRNAs in Extracellular Vesicle in Atherosclerosis. Front Cardiovasc Med 2021; 8:733985. [PMID: 34692785 PMCID: PMC8531438 DOI: 10.3389/fcvm.2021.733985] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis, a complex chronic inflammatory disease, involves multiple alterations of diverse cells, including endothelial cells (ECs), vascular smooth muscle cells (VSMCs), monocytes, macrophages, dendritic cells (DCs), platelets, and even mesenchymal stem cells (MSCs). Globally, it is a common cause of morbidity as well as mortality. It leads to myocardial infarctions, stroke and disabling peripheral artery disease. Extracellular vesicles (EVs) are a heterogeneous group of cell-derived membranous structures that secreted by multiple cell types and play a central role in cell-to-cell communication by delivering various bioactive cargos, especially microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). Emerging evidence demonstrated that miRNAs and lncRNAs in EVs are tightly associated with the initiation and development of atherosclerosis. In this review, we will outline and compile the cumulative roles of miRNAs and lncRNAs encapsulated in EVs derived from diverse cells in the progression of atherosclerosis. We also discuss intercellular communications via EVs. In addition, we focused on clinical applications and evaluation of miRNAs and lncRNAs in EVs as potential diagnostic biomarkers and therapeutic targets for atherosclerosis.
Collapse
Affiliation(s)
- Hui Xu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-related Disease Research, Central South University, Changsha, China
| | - Yu-Qing Ni
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-related Disease Research, Central South University, Changsha, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-related Disease Research, Central South University, Changsha, China
| |
Collapse
|
20
|
Femminò S, D’Ascenzo F, Ravera F, Comità S, Angelini F, Caccioppo A, Franchin L, Grosso A, Thairi C, Venturelli E, Cavallari C, Penna C, De Ferrari GM, Camussi G, Pagliaro P, Brizzi MF. Percutaneous Coronary Intervention (PCI) Reprograms Circulating Extracellular Vesicles from ACS Patients Impairing Their Cardio-Protective Properties. Int J Mol Sci 2021; 22:ijms221910270. [PMID: 34638611 PMCID: PMC8508604 DOI: 10.3390/ijms221910270] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are promising therapeutic tools in the treatment of cardiovascular disorders. We have recently shown that EVs from patients with Acute Coronary Syndrome (ACS) undergoing sham pre-conditioning, before percutaneous coronary intervention (PCI) were cardio-protective, while EVs from patients experiencing remote ischemic pre-conditioning (RIPC) failed to induce protection against ischemia/reperfusion Injury (IRI). No data on EVs from ACS patients recovered after PCI are currently available. Therefore, we herein investigated the cardio-protective properties of EVs, collected after PCI from the same patients. EVs recovered from 30 patients randomly assigned (1:1) to RIPC (EV-RIPC) or sham procedures (EV-naive) (NCT02195726) were characterized by TEM, FACS and Western blot analysis and evaluated for their mRNA content. The impact of EVs on hypoxia/reoxygenation damage and IRI, as well as the cardio-protective signaling pathways, were investigated in vitro (HMEC-1 + H9c2 co-culture) and ex vivo (isolated rat heart). Both EV-naive and EV-RIPC failed to drive cardio-protection both in vitro and ex vivo. Consistently, EV treatment failed to activate the canonical cardio-protective pathways. Specifically, PCI reduced the EV-naive Dusp6 mRNA content, found to be crucial for their cardio-protective action, and upregulated some stress- and cell-cycle-related genes in EV-RIPC. We provide the first evidence that in ACS patients, PCI reprograms the EV cargo, impairing EV-naive cardio-protective properties without improving EV-RIPC functional capability.
Collapse
Affiliation(s)
- Saveria Femminò
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.F.); (F.R.); (A.C.); (A.G.); (E.V.); (G.C.)
| | - Fabrizio D’Ascenzo
- Department of Medical Sciences, Division of Cardiology, University of Turin, 10126 Turin, Italy; (F.D.); (F.A.); (L.F.); (G.M.D.F.)
| | - Francesco Ravera
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.F.); (F.R.); (A.C.); (A.G.); (E.V.); (G.C.)
| | - Stefano Comità
- Department of Clinical and Biological Sciences, University of Turin, 10143 Orbassano, Italy; (S.C.); (C.T.); (C.P.); (P.P.)
| | - Filippo Angelini
- Department of Medical Sciences, Division of Cardiology, University of Turin, 10126 Turin, Italy; (F.D.); (F.A.); (L.F.); (G.M.D.F.)
| | - Andrea Caccioppo
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.F.); (F.R.); (A.C.); (A.G.); (E.V.); (G.C.)
| | - Luca Franchin
- Department of Medical Sciences, Division of Cardiology, University of Turin, 10126 Turin, Italy; (F.D.); (F.A.); (L.F.); (G.M.D.F.)
| | - Alberto Grosso
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.F.); (F.R.); (A.C.); (A.G.); (E.V.); (G.C.)
| | - Cecilia Thairi
- Department of Clinical and Biological Sciences, University of Turin, 10143 Orbassano, Italy; (S.C.); (C.T.); (C.P.); (P.P.)
| | - Emilio Venturelli
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.F.); (F.R.); (A.C.); (A.G.); (E.V.); (G.C.)
| | | | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10143 Orbassano, Italy; (S.C.); (C.T.); (C.P.); (P.P.)
| | - Gaetano Maria De Ferrari
- Department of Medical Sciences, Division of Cardiology, University of Turin, 10126 Turin, Italy; (F.D.); (F.A.); (L.F.); (G.M.D.F.)
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.F.); (F.R.); (A.C.); (A.G.); (E.V.); (G.C.)
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, 10143 Orbassano, Italy; (S.C.); (C.T.); (C.P.); (P.P.)
| | - Maria Felice Brizzi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.F.); (F.R.); (A.C.); (A.G.); (E.V.); (G.C.)
- Correspondence: ; Tel.: +39-011-670-6653
| |
Collapse
|
21
|
Lin B, Yang J, Song Y, Dang G, Feng J. Exosomes and Atherogenesis. Front Cardiovasc Med 2021; 8:738031. [PMID: 34513963 PMCID: PMC8427277 DOI: 10.3389/fcvm.2021.738031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/04/2021] [Indexed: 01/08/2023] Open
Abstract
Myocardial infarction and ischemic stroke are the leading causes of mortality worldwide. Atherosclerosis is their common pathological foundation. It is known that atherosclerosis is characterized by endothelial activation/injury, accumulation of inflammatory immune cells and lipid-rich foam cells, followed by the development of atherosclerotic plaque. Either from arterial vessel wall or blood circulation, endothelial cells, smooth muscle cells, macrophages, T-lymphocytes, B-lymphocytes, foam cells, and platelets have been considered to contribute to the pathogenesis of atherosclerosis. Exosomes, as natural nano-carriers and intercellular messengers, play a significant role in modulation of cell-to-cell communication. Under physiological or pathological conditions, exosomes can deliver their cargos including donor cell-specific proteins, lipids, and nucleic acids to target cells, which in turn affect the function of the target cells. In this review, we will describe the pathophysiological significance of various exosomes derived from different cell types associated with atherosclerosis, and the potential applications of exosome in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Bingbing Lin
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Juan Yang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yuwei Song
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Guohui Dang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Juan Feng
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
22
|
Escudé Martinez de Castilla P, Tong L, Huang C, Sofias AM, Pastorin G, Chen X, Storm G, Schiffelers RM, Wang JW. Extracellular vesicles as a drug delivery system: A systematic review of preclinical studies. Adv Drug Deliv Rev 2021; 175:113801. [PMID: 34015418 DOI: 10.1016/j.addr.2021.05.011] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/10/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023]
Abstract
During the past decades, extracellular vesicles (EVs) have emerged as an attractive drug delivery system. Here, we assess their pre-clinical applications, in the form of a systematic review. For each study published in the past decade, disease models, animal species, EV donor cell types, active pharmaceutical ingredients (APIs), EV surface modifications, API loading methods, EV size and charge, estimation of EV purity, presence of biodistribution studies and administration routes were quantitatively analyzed in a defined and reproducible way. We have interpreted the trends we observe over the past decade, to define the niches where to apply EVs for drug delivery in the future and to provide a basis for regulatory guidelines.
Collapse
|
23
|
Abstract
[Figure: see text].
Collapse
|
24
|
Gebara N, Correia Y, Wang K, Bussolati B. Angiogenic Properties of Placenta-Derived Extracellular Vesicles in Normal Pregnancy and in Preeclampsia. Int J Mol Sci 2021; 22:5402. [PMID: 34065595 PMCID: PMC8160914 DOI: 10.3390/ijms22105402] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is one of the main processes that coordinate the biological events leading to a successful pregnancy, and its imbalance characterizes several pregnancy-related diseases, including preeclampsia. Intracellular interactions via extracellular vesicles (EVs) contribute to pregnancy's physiology and pathophysiology, and to the fetal-maternal interaction. The present review outlines the implications of EV-mediated crosstalk in the angiogenic process in healthy pregnancy and its dysregulation in preeclampsia. In particular, the effect of EVs derived from gestational tissues in pro and anti-angiogenic processes in the physiological and pathological setting is described. Moreover, the application of EVs from placental stem cells in the clinical setting is reported.
Collapse
Affiliation(s)
- Natalia Gebara
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| | - Yolanda Correia
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Keqing Wang
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| |
Collapse
|
25
|
Mazzariol M, Camussi G, Brizzi MF. Extracellular Vesicles Tune the Immune System in Renal Disease: A Focus on Systemic Lupus Erythematosus, Antiphospholipid Syndrome, Thrombotic Microangiopathy and ANCA-Vasculitis. Int J Mol Sci 2021; 22:ijms22084194. [PMID: 33919576 PMCID: PMC8073859 DOI: 10.3390/ijms22084194] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 01/02/2023] Open
Abstract
Extracellular vesicles (EV) are microparticles released in biological fluids by different cell types, both in physiological and pathological conditions. Owing to their ability to carry and transfer biomolecules, EV are mediators of cell-to-cell communication and are involved in the pathogenesis of several diseases. The ability of EV to modulate the immune system, the coagulation cascade, the angiogenetic process, and to drive endothelial dysfunction plays a crucial role in the pathophysiology of both autoimmune and renal diseases. Recent studies have demonstrated the involvement of EV in the control of renal homeostasis by acting as intercellular signaling molecules, mediators of inflammation and tissue regeneration. Moreover, circulating EV and urinary EV secreted by renal cells have been investigated as potential early biomarkers of renal injury. In the present review, we discuss the recent findings on the involvement of EV in autoimmunity and in renal intercellular communication. We focused on EV-mediated interaction between the immune system and the kidney in autoimmune diseases displaying common renal damage, such as antiphospholipid syndrome, systemic lupus erythematosus, thrombotic microangiopathy, and vasculitis. Although further studies are needed to extend our knowledge on EV in renal pathology, a deeper investigation of the impact of EV in kidney autoimmune diseases may also provide insight into renal biological processes. Furthermore, EV may represent promising biomarkers of renal diseases with potential future applications as diagnostic and therapeutic tools.
Collapse
|
26
|
Lee SA, Choi C, Yoo TH. Extracellular vesicles in kidneys and their clinical potential in renal diseases. Kidney Res Clin Pract 2021; 40:194-207. [PMID: 33866768 PMCID: PMC8237124 DOI: 10.23876/j.krcp.20.209] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/26/2021] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs), such as exosomes and microvesicles, are cell-derived lipid bilayer membrane particles, which deliver information from host cells to recipient cells. EVs are involved in various biological processes including the modulation of the immune response, cell-to-cell communications, thrombosis, and tissue regeneration. Different types of kidney cells are known to release EVs under physiologic as well as pathologic conditions, and recent studies have found that EVs have a pathophysiologic role in different renal diseases. Given the recent advancement in EV isolation and analysis techniques, many studies have shown the diagnostic and therapeutic potential of EVs in various renal diseases, such as acute kidney injury, polycystic kidney disease, chronic kidney disease, kidney transplantation, and renal cell carcinoma. This review updates recent clinical and experimental findings on the role of EVs in renal diseases and highlights the potential clinical applicability of EVs as novel diagnostics and therapeutics.
Collapse
Affiliation(s)
- Sul A Lee
- Department of Internal Medicine and Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, MetroWest Medical Center, Framingham, MA, USA
| | - Chulhee Choi
- ILIAS Biologics Inc., Daejeon, Republic of Korea.,Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Tae-Hyun Yoo
- Department of Internal Medicine and Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
27
|
Qin Z, Liao R, Xiong Y, Jiang L, Li J, Wang L, Han M, Sun S, Geng J, Yang Q, Zhang Z, Li Y, Du H, Su B. A narrative review of exosomes in vascular calcification. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:579. [PMID: 33987277 DOI: 10.21037/atm-20-7355] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Vascular calcification (VC) is the abnormal deposition of calcium, phosphorus, and other minerals in the vessel wall and can be commonly observed in diabetes, chronic kidney disease, and chronic inflammatory disease. It is closely associated with mortality from cardiovascular events. Traditionally, calcification is considered as a degenerative disease associated with the aging process, while increasing evidence has shown that the occurrence and development of calcification is an active biological process, which is highly regulated by multiple factors. The molecular mechanisms of VC have not yet been fully elucidated. Exosomes, as important transporters of substance transport and intercellular communication, have been shown to participate in VC. The regulation of VC by exosomes involves a number of complex biological processes, which occur through a variety of interaction mechanisms. However, the specific role and mechanism of exosomes in the process of VC are still not fully understood and require further study. This review will briefly describe the roles of exosomes in the process of VC including in the promotion of extracellular mineral deposits, induction of phenotypic conversion of vascular smooth muscle cells (VSMCs), transport of microRNA between cells, and regulation on autophagy and oxidative stress, with the aim of providing novel ideas for the clinical diagnosis and treatment of VC.
Collapse
Affiliation(s)
- Zheng Qin
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Ruoxi Liao
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Yuqin Xiong
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Luojia Jiang
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Jiameng Li
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Liya Wang
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Mei Han
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Si Sun
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Jiwen Geng
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Qinbo Yang
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Zhuyun Zhang
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Yupei Li
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China.,Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, China
| | - Heyue Du
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Baihai Su
- Department of nephrology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Wu Y, Liu J. Effect of exosome -derived non -coding RNA on traumatic brain injury. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:183-188. [PMID: 33678656 PMCID: PMC10929786 DOI: 10.11817/j.issn.1672-7347.2021.190702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Indexed: 11/03/2022]
Abstract
Traumatic brain injury (TBI) is a main cause of death and disability worldwide, posing a serious threat to public health. But currently, the diagnosis and treatments for TBI are still very limited. Exosomes are a group of extracellular vesicles and participate in multiple physiological processes including intercellular communication and substance transport. Non-coding RNAs (ncRNA) are of great abundancy as cargo of exosomes. Previous studies have shown that ncRNAs are involved in several pathophysiological processes of TBI. However, the concrete mechanisms involved in the effects induced by exosome-derived ncRNA remain largely unknown. As an important component of exosomes, ncRNA is of great significance for diagnosis, precise treatment, response evaluation, prognosis prediction, and complication management after TBI.
Collapse
Affiliation(s)
- Yun Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Jinfang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
29
|
Dalirfardouei R, Gholoobi A, Vahabian M, Mahdipour E, Afzaljavan F. Therapeutic role of extracellular vesicles derived from stem cells in cutaneous wound models: A systematic review. Life Sci 2021; 273:119271. [PMID: 33652035 DOI: 10.1016/j.lfs.2021.119271] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/07/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
A growing body of evidence has shown that extracellular vesicles can be efficient as experimental therapeutics in pre-clinical models of skin wounds, but there is a significant unmet need to translate this to clinical utilization. The objectives of the current systematic review were to identify the strength of the therapeutic effects of EVs derived from stem cells in cutaneous wounds and to assess which EV-mediated mechanisms could be involved in the therapeutic response. PubMed, ISI Web of Science, and Scopus databases were systematically searched. We retrieved English-language articles published through June 2020. In vivo studies which applied stem cell-derived EVs were included for further analysis. The Risk of bias was assessed by the SYRCLE tool. We identified thirty-nine pre-clinical studies that evaluated the effects of EVs on the wound healing process. The included studies varied greatly in EVs isolation techniques, route of administration, EVs producing cells, and follow-up time. In vivo application revealed beneficial effects of EVs on accelerating wound closure and re-epithelialization in a dose-dependent manner. Elevated angiogenesis was reported in twelve eligible studies through multiple signaling pathways such as PI3K/Akt, MAPK/ERK, and JAK/STAT. The well-known signaling pathway to inhibit scar formation was TGF-β2/SMAD2. However, all included studies were not blinded enough which may have introduced bias. Therefore, the transition of EV's efficacy into the clinics is deeply rooted in the following important factors: 1) pre-clinical studies with a lower risk of bias and longer follow-up time, and 2) consistent, reproducible, and feasible manufacturing of EVs production in a large-scale commercial program.
Collapse
Affiliation(s)
- Razieh Dalirfardouei
- Research center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Aida Gholoobi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrangiz Vahabian
- Department of English Language and Persian Literature, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Elahe Mahdipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Afzaljavan
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
30
|
Phenotypic and Proteomic Analysis Identifies Hallmarks of Blood Circulating Extracellular Vesicles in NSCLC Responders to Immune Checkpoint Inhibitors. Cancers (Basel) 2021; 13:cancers13040585. [PMID: 33546102 PMCID: PMC7913165 DOI: 10.3390/cancers13040585] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Purpose of this study was to investigate the prognostic and predictive role of blood circulating extracellular vesicles (EVs) in patients with advanced non-small cell lung cancer treated with immunotherapy. A newly optimized flow cytometry protocol was applied for identification and subtyping of blood circulating EVs in a total cohort of 59 NSCLC patients, which included 31 patients treated with anti-PD-1/PD-L1 agents and 28 patients treated with traditional chemotherapy. Our results show that pre-treatment concentration of blood circulating endothelial-derived EVs was correlated with overall survival and clinical response in patients treated with immunotherapy. Additionally, proteomic analysis of purified blood circulating EVs indicated differences in EV protein cargo between responders and non-responders to immunotherapy. These findings may pave the way to the identification of novel immunotherapy biomarkers in patients with advanced NSCLC. Abstract Immune checkpoint inhibitors (ICIs) induce durable clinical responses only in a subset of advanced non-small cell lung cancer (NSCLC) patients. There is a need to identify mechanisms of ICI resistance and immunotherapy biomarkers to improve clinical benefit. In this study, we evaluated the prognostic and predictive value of circulating endothelial and leukocyte-derived extracellular vesicles (EV) in patients with advanced NSCLC treated with anti-PD-1/PD-L1 agents. In addition, the relationship between total blood circulating EV proteome and response to ICIs was investigated. An optimized flow cytometry method was employed for the identification and subtyping of blood circulating EVs in 59 patients with advanced NSCLC. Blood samples were collected from patients receiving anti-PD-1/PD-L1 inhibitors (n = 31) or chemotherapy (n = 28). An exploratory proteomic analysis of sorted blood EVs was conducted in a subset of patients. Our results show that a low blood concentration of circulating endothelial-derived EVs before treatment was strongly associated to longer overall survival (p = 0.0004) and higher disease control rate (p = 0.045) in patients treated with ICIs. Interestingly, shotgun proteomics revealed that EVs of responders to anti-PD-1 therapy had a specific protein cargo before treatment. In addition, EV protein cargo was specifically modulated during immunotherapy. We identified a previously unknown association between circulating endothelial-derived extracellular vesicle concentration and immunotherapy-related clinical outcomes. We also observed differences in circulating extracellular vesicle proteome according to anti-PD-1-based treatment response in NSCLC patients. Overall, these results may contribute to the identification of novel circulating biomarkers for rational immunotherapy approaches in patients affected by NSCLC.
Collapse
|
31
|
Herrmann M, Diederichs S, Melnik S, Riegger J, Trivanović D, Li S, Jenei-Lanzl Z, Brenner RE, Huber-Lang M, Zaucke F, Schildberg FA, Grässel S. Extracellular Vesicles in Musculoskeletal Pathologies and Regeneration. Front Bioeng Biotechnol 2021; 8:624096. [PMID: 33553127 PMCID: PMC7855463 DOI: 10.3389/fbioe.2020.624096] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
The incidence of musculoskeletal diseases is steadily increasing with aging of the population. In the past years, extracellular vesicles (EVs) have gained attention in musculoskeletal research. EVs have been associated with various musculoskeletal pathologies as well as suggested as treatment option. EVs play a pivotal role in communication between cells and their environment. Thereby, the EV cargo is highly dependent on their cellular origin. In this review, we summarize putative mechanisms by which EVs can contribute to musculoskeletal tissue homeostasis, regeneration and disease, in particular matrix remodeling and mineralization, pro-angiogenic effects and immunomodulatory activities. Mesenchymal stromal cells (MSCs) present the most frequently used cell source for EV generation for musculoskeletal applications, and herein we discuss how the MSC phenotype can influence the cargo and thus the regenerative potential of EVs. Induced pluripotent stem cell-derived mesenchymal progenitor cells (iMPs) may overcome current limitations of MSCs, and iMP-derived EVs are discussed as an alternative strategy. In the last part of the article, we focus on therapeutic applications of EVs and discuss both practical considerations for EV production and the current state of EV-based therapies.
Collapse
Affiliation(s)
- Marietta Herrmann
- Interdisciplinary Center for Clinical Research (IZKF) Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Solvig Diederichs
- Research Centre for Experimental Orthopaedics, Centre for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Svitlana Melnik
- Research Centre for Experimental Orthopaedics, Centre for Orthopaedics, Trauma Surgery and Paraplegiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Drenka Trivanović
- Interdisciplinary Center for Clinical Research (IZKF) Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
- Bernhard-Heine-Centrum for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Shushan Li
- Department of Orthopedic Surgery, Experimental Orthopedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, Regensburg, Germany
| | - Zsuzsa Jenei-Lanzl
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, Frankfurt, Germany
| | - Rolf E. Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, Frankfurt, Germany
| | - Frank A. Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Susanne Grässel
- Department of Orthopedic Surgery, Experimental Orthopedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, Regensburg, Germany
| |
Collapse
|
32
|
The Inflammatory Cytokine IL-3 Hampers Cardioprotection Mediated by Endothelial Cell-Derived Extracellular Vesicles Possibly via Their Protein Cargo. Cells 2020; 10:cells10010013. [PMID: 33374685 PMCID: PMC7822476 DOI: 10.3390/cells10010013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/09/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
The biological relevance of extracellular vesicles (EV) released in an ischemia/reperfusion setting is still unclear. We hypothesized that the inflammatory microenvironment prevents cardioprotection mediated by endothelial cell (EC)-derived extracellular vesicles. The effects of naïve EC-derived EV (eEV) or eEV released in response to interleukin-3 (IL-3) (eEV-IL-3) were evaluated in cardiomyoblasts (H9c2) and rat hearts. In transwell assay, eEV protected the H9c2 exposed to hypoxia/reoxygenation (H/R) more efficiently than eEV-IL-3. Conversely, only eEV directly protected H9c2 cells to H/R-induced damage. Consistent with this latter observation, eEV, but not eEV-IL-3, exerted beneficial effects in the whole heart. Protein profiles of eEV and eEV-IL-3, established using label-free mass spectrometry, demonstrated that IL-3 drives changes in eEV-IL-3 protein cargo. Gene ontology analysis revealed that both eEV and eEV-IL-3 were equipped with full cardioprotective machinery, including the Nitric Oxide Signaling in the Cardiovascular System. eEV-IL-3 were also enriched in the endothelial-nitric oxide-synthase (eNOS)-antagonist caveolin-1 and proteins related to the inflammatory response. In vitro and ex vivo experiments demonstrated that a functional Mitogen-Activated Protein Kinase Kinase (MEK1/2)/eNOS/guanylyl-cyclase (GC) pathway is required for eEV-mediated cardioprotection. Consistently, eEV were found enriched in MEK1/2 and able to induce the expression of B-cell-lymphoma-2 (Bcl-2) and the phosphorylation of eNOS in vitro. We conclude that an inflammatory microenvironment containing IL-3 changes the eEV cargo and impairs eEV cardioprotective action.
Collapse
|
33
|
Jurj A, Pop-Bica C, Slaby O, Ştefan CD, Cho WC, Korban SS, Berindan-Neagoe I. Tiny Actors in the Big Cellular World: Extracellular Vesicles Playing Critical Roles in Cancer. Int J Mol Sci 2020; 21:7688. [PMID: 33080788 PMCID: PMC7589964 DOI: 10.3390/ijms21207688] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/04/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023] Open
Abstract
Communications among cells can be achieved either via direct interactions or via secretion of soluble factors. The emergence of extracellular vesicles (EVs) as entities that play key roles in cell-to-cell communication offer opportunities in exploring their features for use in therapeutics; i.e., management and treatment of various pathologies, such as those used for cancer. The potential use of EVs as therapeutic agents is attributed not only for their cell membrane-bound components, but also for their cargos, mostly bioactive molecules, wherein the former regulate interactions with a recipient cell while the latter trigger cellular functions/molecular mechanisms of a recipient cell. In this article, we highlight the involvement of EVs in hallmarks of a cancer cell, particularly focusing on those molecular processes that are influenced by EV cargos. Moreover, we explored the roles of RNA species and proteins carried by EVs in eliciting drug resistance phenotypes. Interestingly, engineered EVs have been investigated and proposed as therapeutic agents in various in vivo and in vitro studies, as well as in several clinical trials.
Collapse
Affiliation(s)
- Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.J.); (C.P.-B.)
| | - Cecilia Pop-Bica
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.J.); (C.P.-B.)
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic;
- Department of Pathology, Faculty Hospital Brno and Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Cristina D. Ştefan
- SingHealth Duke-NUS Global Health Institute, Singapore 169857, Singapore;
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China;
| | - Schuyler S. Korban
- Department of Natural Resources and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA;
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.J.); (C.P.-B.)
- Department of Functional Genomics and Experimental Pathology, “Prof. Dr. Ion Chiricuta” Oncology Institute, 400015 Cluj-Napoca, Romania
| |
Collapse
|
34
|
Roefs MT, Sluijter JPG, Vader P. Extracellular Vesicle-Associated Proteins in Tissue Repair. Trends Cell Biol 2020; 30:990-1013. [PMID: 33069512 DOI: 10.1016/j.tcb.2020.09.009] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
The administration of (stem) cell-derived extracellular vesicles (EVs) promotes tissue repair through management of different inflammatory, proliferative and remodeling processes in the body. Despite the widely observed biological and therapeutic roles of EVs in wound healing and tissue repair, knowledge on how EVs activate recipient cells and which EV cargo is responsible for the subsequent functional effects is limited. Recent studies hint toward an important role for proteins as functional EV cargo. Here, we provide an overview of how EV-associated proteins promote tissue repair processes and discuss current challenges in evaluating their contribution to EV function and future directions for translating fundamental insights into clinically relevant EV therapies.
Collapse
Affiliation(s)
- Marieke T Roefs
- Department of Cardiology, Experimental Cardiology Laboratory, University Utrecht, University Medical Center Utrecht, The Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, University Utrecht, University Medical Center Utrecht, The Netherlands.
| | - Pieter Vader
- Department of Cardiology, Experimental Cardiology Laboratory, University Utrecht, University Medical Center Utrecht, The Netherlands; CDL Research, University Medical Center Utrecht, The Netherlands.
| |
Collapse
|
35
|
Niazi V, Parseh B, Ahani M, Karami F, Gilanchi S, Atarodi K, Soufi M, Soleimani M, Ghafouri-Fard S, Taheri M, Zali H. Communication between stromal and hematopoietic stem cell by exosomes in normal and malignant bone marrow niche. Biomed Pharmacother 2020; 132:110854. [PMID: 33059261 DOI: 10.1016/j.biopha.2020.110854] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/26/2020] [Accepted: 10/04/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) have been regarded as important tools for cell-cell communication. They act as carriers for the transfer of various molecules such as genes, proteins and miRNA. EVs shift and transfer their ingredients to target cells in an active form. These particles have prominent roles in modulation of bone marrow (BM) niche; therefore they can regulate proliferation, differentiation, and other properties of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). This review discusses the different roles of EVs on BM niche; HPCs fate regulation and downstream effects of them on HSCs. Moreover, cellular and molecular mechanisms of BM microenvironment cross-talking are explained in healthy and malignant settings.
Collapse
Affiliation(s)
- Vahid Niazi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19857-17443, Iran
| | - Benyamin Parseh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 14177-55469, Iran
| | - Milad Ahani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19857-17443, Iran
| | - Farshid Karami
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19857-17443, Iran
| | - Samira Gilanchi
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, 19716-53313, Iran
| | - Kamran Atarodi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, 14665-1157, Iran
| | - Mina Soufi
- Department of Hematology and Cell Therapy, Faculty of Medical Science, Tarbiat Modares University, Tehran, 14117-13116, Iran
| | - Masoud Soleimani
- Department of Hematology and Cell Therapy, Faculty of Medical Science, Tarbiat Modares University, Tehran, 14117-13116, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19857-17443, Iran; Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, 19716-53313, Iran.
| |
Collapse
|
36
|
Targeting IL-3Rα on tumor-derived endothelial cells blunts metastatic spread of triple-negative breast cancer via extracellular vesicle reprogramming. Oncogenesis 2020; 9:90. [PMID: 33040091 PMCID: PMC7548009 DOI: 10.1038/s41389-020-00274-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/10/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
The lack of approved targeted therapies highlights the need for new treatments for triple-negative breast cancer (TNBC) patients. Interleukin-3 (IL-3) acts as an autocrine factor for tumor-endothelial cells (TEC), and exerts pro-angiogenic paracrine action via extracellular vesicles (EVs). IL-3Rα blockade on TEC changes TEC-EV (anti-IL-3R-EV) microRNA (miR) content and promotes the regression of established vessels. As TEC is the doorway for "drug" entry into tumors, we aimed to assess whether IL-3R blockade on TEC impacts tumor progression via its unique EV cargo. First, the expression of IL-3Rα was evaluated in 27 human TNBC samples. It was noticed that, besides TEC and inflammatory cells, tumor cells from 55.5% of the human TNBC samples expressed IL-3Rα. Using human TNBC cell lines for in vitro studies, we found that, unlike native TEC-EVs (nEVs), anti-IL-3R-EVs increase apoptosis and reduced cell viability and migration. In vivo, anti-IL-3R-EV treatment induced vessel regression in established tumors formed of MDA-MB-231 cells, decreased Vimentin, β-catenin, and TWIST1 expression, almost abolished liver and lung metastases from primary tumors, and reduced lung metastasis generated via the intravenous injection of MDA-MB-231 cells. nEVs depleted of miR-24-3p (antago-miR-24-3p-EVs) were effective as anti-IL-3R-EVs in downregulating TWIST1 and reducing metastatic lesions in vivo. Consistent with network analyses of miR-24-3p gene targeting, anti-IL-3R-EVs and antago-miR-24-3p-EVs upregulate SPRY2 in MDA-MB-231 cells. Finally, SPRY2 silencing prevented anti-IL-3R-EV and antago-miR-24-3p-EV-mediated apoptotic cues.Overall, these data provide the first evidence that IL-3Rα is highly expressed in TNBC cells, TEC, and inflammatory cells, and that IL-3Rα blockade on TEC impacts tumor progression.
Collapse
|
37
|
Kesidou D, da Costa Martins PA, de Windt LJ, Brittan M, Beqqali A, Baker AH. Extracellular Vesicle miRNAs in the Promotion of Cardiac Neovascularisation. Front Physiol 2020; 11:579892. [PMID: 33101061 PMCID: PMC7546892 DOI: 10.3389/fphys.2020.579892] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide claiming almost 17. 9 million deaths annually. A primary cause is atherosclerosis within the coronary arteries, which restricts blood flow to the heart muscle resulting in myocardial infarction (MI) and cardiac cell death. Despite substantial progress in the management of coronary heart disease (CHD), there is still a significant number of patients developing chronic heart failure post-MI. Recent research has been focused on promoting neovascularisation post-MI with the ultimate goal being to reduce the extent of injury and improve function in the failing myocardium. Cardiac cell transplantation studies in pre-clinical models have shown improvement in cardiac function; nonetheless, poor retention of the cells has indicated a paracrine mechanism for the observed improvement. Cell communication in a paracrine manner is controlled by various mechanisms, including extracellular vesicles (EVs). EVs have emerged as novel regulators of intercellular communication, by transferring molecules able to influence molecular pathways in the recipient cell. Several studies have demonstrated the ability of EVs to stimulate angiogenesis by transferring microRNA (miRNA, miR) molecules to endothelial cells (ECs). In this review, we describe the process of neovascularisation and current developments in modulating neovascularisation in the heart using miRNAs and EV-bound miRNAs. Furthermore, we critically evaluate methods used in cell culture, EV isolation and administration.
Collapse
Affiliation(s)
- Despoina Kesidou
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Paula A. da Costa Martins
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, Maastricht, Netherlands
- Faculty of Health, Medicine and Life Sciences, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Leon J. de Windt
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, Maastricht, Netherlands
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Abdelaziz Beqqali
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew Howard Baker
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Li S, Yuan L, Su L, Lian Z, Liu C, Zhang F, Cui Y, Wu M, Chen H. Decreased miR‑92a‑3p expression potentially mediates the pro‑angiogenic effects of oxidative stress‑activated endothelial cell‑derived exosomes by targeting tissue factor. Int J Mol Med 2020; 46:1886-1898. [PMID: 32901851 PMCID: PMC7521555 DOI: 10.3892/ijmm.2020.4713] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is an essential pathological feature of vulnerable atherosclerotic plaque. Exosome‑derived microRNAs (miRNAs or miRs) have been proven to be important regulators of angiogenesis. However, the role of exosomes, which are secreted by endothelial cells (ECs) under conditions of oxidative stress, in angiogenesis remain unclear. The present study aimed to investigate the effects and mechanism of oxidative stress‑activated endothelial‑derived exosomes in angiogenesis. Exosomes were isolated from H2O2‑stimulated human umbilical vein ECs (HUVECs; termed Exo-H2O2) by differential centrifugation and characterized by transmission electron microscopy, nanoparticle tracking analysis and western blot analysis. Exo-H2O2 enhanced HUVEC proliferation, migration and tube formation, as determined by EdU incorporation assay, scratch wound migration assay and tube formation assay, respectively. miR‑92a‑3p was identified as the predominantly downregulated miRNA in the Exo-H2O2‑treated HUVECs by small RNA sequencing, and the expression of primary miR‑92a (pri‑miR‑92a‑1) was also decreased, as shown by RT‑qPCR. Similarly, the inhibition of miR‑92a‑3p promoted angiogenesis in vitro and in vivo. miR‑92a‑3p overexpression blocked the pro‑angiogenic effects of Exo-H2O2 on target ECs. Tissue factor (TF), a molecule involved in angiogenesis, was increased in HUVECs in which miR‑92a‑3p expression was downregulated, as shown by mRNA sequencing. TF was also predicted as a target of miR‑92a‑3p by using the RNAhybrid program. The overexpression or suppression of miR‑92a‑3p modified TF expression at both the mRNA and protein level, as measured by RT‑qPCR and western blot analysis, respectively. Luciferase reporter assays suggested that miR‑92a‑3p inhibited TF expression by binding to the 3' untranslated region of TF. On the whole, the findings of the present study demonstrate that exosomes released from oxidative stress‑activated ECs stimulate angiogenesis by inhibiting miR‑92a‑3p expression in recipient ECs, and TF may be involved in the regulatory effects of miR‑92a‑3p on angiogenesis.
Collapse
Affiliation(s)
- Sufang Li
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Lan Yuan
- Medical and Healthy Analytical Center, Peking University, Beijing 100191, P.R. China
| | - Lina Su
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Zheng Lian
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Chuanfen Liu
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Feng Zhang
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Yuxia Cui
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Manyan Wu
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Hong Chen
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| |
Collapse
|
39
|
Targeting Angiopoietin in Retinal Vascular Diseases: A Literature Review and Summary of Clinical Trials Involving Faricimab. Cells 2020; 9:cells9081869. [PMID: 32785136 PMCID: PMC7464130 DOI: 10.3390/cells9081869] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022] Open
Abstract
This review summarizes the latest findings in the literature of Angiopoietin-2 (Ang-2), Tyrosine-protein kinase receptor (Tie-2) complex, and faricimab along with their involvement for the treatment of retinal vascular diseases in various clinical trials. In ischemic diseases, such as diabetic retinopathy, Ang-2 is upregulated, deactivating Tie-2, resulting in vascular leakage, pericyte loss, and inflammation. Recombinant Angiopeotin-1 (Ang-1), Ang-2-blocking molecules, and inhibitors of vascular endothelial protein tyrosine phosphatase (VE-PTP) decrease inflammation-associated vascular leakage, showing therapeutic effects in diabetes, atherosclerosis, and ocular neovascular diseases. In addition, novel studies show that angiopoietin-like proteins may play an important role in cellular metabolism leading to retinal vascular diseases. Current therapeutic focus combines Ang-Tie targeted drugs with other anti-angiogenic or immune therapies. Clinical studies have identified faricimab, a novel bispecific antibody designed for intravitreal use, to simultaneously bind and neutralize Ang-2 and VEGF-A for treatment of diabetic eye disease. By targeting both Ang-2 and vascular endothelial growth factor-A (VEGF-A), faricimab displays an improved and sustained efficacy over longer treatment intervals, delivering superior vision outcomes for patients with diabetic macular edema and reducing the treatment burden for patients with neovascular age-related macular degeneration and diabetic macular edema. Phase 2 results have produced promising outcomes with regard to efficacy and durability. Faricimab is currently being evaluated in global Phase 3 studies.
Collapse
|
40
|
Effect of Physical Exercise on the Release of Microparticles with Angiogenic Potential. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10144871] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cellular communication has a fundamental role in both human physiological and pathological states and various mechanisms are involved in the crosstalk between organs. Among these, microparticles (MPs) have an important involvement. MPs are a subtype of extracellular vesicles produced by a variety of cells following activation or apoptosis. They are normally present in physiological conditions, but their concentration varies in pathological states such as cardiovascular disease, diabetes mellitus, or cancer. Acute and chronic physical exercise are able to modify MPs amounts as well. Among various actions, exercise-responsive MPs affect angiogenesis, the process through which new blood vessels grow from pre-existing vessels. Usually, the neo vascular growth has functional role; but an aberrant neovascularization accompanies several oncogenic, ischemic, or inflammatory diseases. In addition, angiogenesis is one of the key adaptations to physical exercise and training. In the present review, we report evidence regarding the effect of various typologies of exercise on circulating MPs that are able to affect angiogenesis.
Collapse
|
41
|
Boomgarden AC, Sheehan C, D'Souza-Schorey C. Extracellular Vesicles in the Tumor Microenvironment: Various Implications in Tumor Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1259:155-170. [PMID: 32578176 DOI: 10.1007/978-3-030-43093-1_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Extracellular vesicle (EV) shedding is a biologically conserved cellular process across virtually every cell type. In cancer, EVs shed from tumor and stromal cells to the tumor microenvironment play a major role in determining tumor fate, which to a large extent is dictated by the biologically active cargo contained in EVs. Current understanding of various cancer-associated EVs has enabled the outlining of mechanistic connections between cargo and tumor-promoting functions. In this chapter, we describe examples of EV-mediated communication between tumor cells and stromal cells, highlighting the molecular constituents responsible for pro-tumorigenic effects. Furthermore, we discuss the roles of matrix-degrading EVs in cell invasion. Finally, we summarize research on the potential use of EVs as a novel approach to cancer therapeutics.
Collapse
Affiliation(s)
- Alex C Boomgarden
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Colin Sheehan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | | |
Collapse
|
42
|
Su KY, Lee WL. Fourier Transform Infrared Spectroscopy as a Cancer Screening and Diagnostic Tool: A Review and Prospects. Cancers (Basel) 2020; 12:E115. [PMID: 31906324 PMCID: PMC7017192 DOI: 10.3390/cancers12010115] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 02/07/2023] Open
Abstract
Infrared spectroscopy has long been used to characterize chemical compounds, but the applicability of this technique to the analysis of biological materials containing highly complex chemical components is arguable. However, recent advances in the development of infrared spectroscopy have significantly enhanced the capacity of this technique in analyzing various types of biological specimens. Consequently, there is an increased number of studies investigating the application of infrared spectroscopy in screening and diagnosis of various diseases. The lack of highly sensitive and specific methods for early detection of cancer has warranted the search for novel approaches. Being more simple, rapid, accurate, inexpensive, non-destructive and suitable for automation compared to existing screening, diagnosis, management and monitoring methods, Fourier transform infrared spectroscopy can potentially improve clinical decision-making and patient outcomes by detecting biochemical changes in cancer patients at the molecular level. Besides the commonly analyzed blood and tissue samples, extracellular vesicle-based method has been gaining popularity as a non-invasive approach. Therefore, infrared spectroscopic analysis of extracellular vesicles could be a useful technique in the future for biomedical applications. In this review, we discuss the potential clinical applications of Fourier transform infrared spectroscopic analysis using various types of biological materials for cancer. Additionally, the rationale and advantages of using extracellular vesicles in the spectroscopic analysis for cancer diagnostics are discussed. Furthermore, we highlight the challenges and future directions of clinical translation of the technique for cancer.
Collapse
Affiliation(s)
| | - Wai-Leng Lee
- School of Science, Monash University Malaysia, Subang Jaya 47500, Malaysia
| |
Collapse
|
43
|
Embryonic stem cell-derived extracellular vesicle-mimetic nanovesicles rescue erectile function by enhancing penile neurovascular regeneration in the streptozotocin-induced diabetic mouse. Sci Rep 2019; 9:20072. [PMID: 31882614 PMCID: PMC6934510 DOI: 10.1038/s41598-019-54431-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 09/02/2019] [Indexed: 12/30/2022] Open
Abstract
Extracellular vesicles (EVs) have attracted particular interest in various fields of biology and medicine. However, one of the major hurdles in the clinical application of EV-based therapy is their low production yield. We recently developed cell-derived EV-mimetic nanovesicles (NVs) by extruding cells serially through filters with diminishing pore sizes (10, 5, and 1 μm). Here, we demonstrate in diabetic mice that embryonic stem cell (ESC)-derived EV-mimetic NVs (ESC-NVs) completely restore erectile function (~96% of control values) through enhanced penile angiogenesis and neural regeneration in vivo, whereas ESC partially restores erectile function (~77% of control values). ESC-NVs promoted tube formation in primary cultured mouse cavernous endothelial cells and pericytes under high-glucose condition in vitro; and accelerated microvascular and neurite sprouting from aortic ring and major pelvic ganglion under high-glucose condition ex vivo, respectively. ESC-NVs enhanced the expression of angiogenic and neurotrophic factors (hepatocyte growth factor, angiopoietin-1, nerve growth factor, and neurotrophin-3), and activated cell survival and proliferative factors (Akt and ERK). Therefore, it will be a better strategy to use ESC-NVs than ESCs in patients with erectile dysfunction refractory to pharmacotherapy, although it remains to be solved for future clinical application of ESC.
Collapse
|
44
|
Mbagwu SI, Lannes N, Walch M, Filgueira L, Mantel PY. Human Microglia Respond to Malaria-Induced Extracellular Vesicles. Pathogens 2019; 9:pathogens9010021. [PMID: 31878288 PMCID: PMC7168629 DOI: 10.3390/pathogens9010021] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
Microglia are the chief immune cells of the brain and have been reported to be activated in severe malaria. Their activation may drive towards neuroinflammation in cerebral malaria. Malaria-infected red blood cell derived-extracellular vesicles (MiREVs) are produced during the blood stage of malaria infection. They mediate intercellular communication and immune regulation, among other functions. During cerebral malaria, the breakdown of the blood–brain barrier can promote the migration of substances such as MiREVs from the periphery into the brain, targeting cells such as microglia. Microglia and extracellular vesicle interactions in different pathological conditions have been reported to induce neuroinflammation. Unlike in astrocytes, microglia–extracellular vesicle interaction has not yet been described in malaria infection. Therefore, in this study, we aimed to investigate the uptake of MiREVs by human microglia cells and their cytokine response. Human blood monocyte-derived microglia (MoMi) were generated from buffy coats of anonymous healthy donors using Ficoll-Paque density gradient centrifugation. The MiREVs were isolated from the Plasmodium falciparum cultures. They were purified by ultracentrifugation and labeled with PKH67 green fluorescent dye. The internalization of MiREVs by MoMi was observed after 4 h of co-incubation on coverslips placed in a 24-well plate at 37 °C using confocal microscopy. Cytokine-gene expression was investigated using rt-qPCR, following the stimulation of the MoMi cells with supernatants from the parasite cultures at 2, 4, and 24 h, respectively. MiREVs were internalized by the microglia and accumulated in the perinuclear region. MiREVs-treated cells increased gene expression of the inflammatory cytokine TNFα and reduced gene expression of the immune suppressive IL-10. Overall, the results indicate that MiREVs may act on microglia, which would contribute to enhanced inflammation in cerebral malaria.
Collapse
Affiliation(s)
- Smart Ikechukwu Mbagwu
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
- Department of Anatomy, Faculty of Basic Medical Sciences, Nnamdi Azikiwe University, Nnewi Campus, Nnewi 435101, Nigeria
- Correspondence: (S.I.M.); (L.F.)
| | - Nils Lannes
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| | - Michael Walch
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| | - Luis Filgueira
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
- Correspondence: (S.I.M.); (L.F.)
| | - Pierre-Yves Mantel
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
45
|
Cardiac Extracellular Vesicles (EVs) Released in the Presence or Absence of Inflammatory Cues Support Angiogenesis in Different Manners. Int J Mol Sci 2019; 20:ijms20246363. [PMID: 31861211 PMCID: PMC6940836 DOI: 10.3390/ijms20246363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
Cells release extracellular vesicles (EVs) to communicate in a paracrine manner with other cells, and thereby influence processes, such as angiogenesis. The conditioned medium of human cardiac-derived adherent proliferating (CardAP) cells was recently shown to enhance angiogenesis. To elucidate whether their released EVs are involved, we isolated them by differential centrifugation from the conditioned medium derived either in the presence or absence of a pro-inflammatory cytokine cocktail. Murine recipient cells internalized CardAP-EVs as determined by an intracellular detection of human proteins, such as CD63, by a novel flow cytometry method for studying EV–cell interaction. Moreover, endothelial cells treated for 24 h with either unstimulated or cytokine stimulated CardAP-EVs exhibited a higher tube formation capability on Matrigel. Interestingly, unstimulated CardAP-EVs caused endothelial cells to release significantly more vascular endothelial growth factor and interleukin (IL)-6, while cytokine stimulated CardAP-EVs significantly enhanced the release of IL-6 and IL-8. By nCounter® miRNA expression assay (NanoString Technologies) we identified microRNA 302d-3p to be enhanced in unstimulated CardAP-EVs compared to their cytokine stimulated counterparts, which was verified by quantitative polymerase chain reaction. This study demonstrates that both CardAP-EVs are pro-angiogenic by inducing different factors from endothelial cells. This would allow to select potent targets for a safe and efficient therapeutic application.
Collapse
|
46
|
Lv L, Sheng C, Zhou Y. Extracellular vesicles as a novel therapeutic tool for cell-free regenerative medicine in oral rehabilitation. J Oral Rehabil 2019; 47 Suppl 1:29-54. [PMID: 31520537 DOI: 10.1111/joor.12885] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 07/26/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022]
Abstract
Oral maxillofacial defects may always lead to complicated hard and soft tissue loss, including bone, nerve, blood vessels, teeth and skin, which are difficult to restore and severely influence the life quality of patients. Extracellular vesicles (EVs), including exosomes, microvesicles and apoptotic bodies, are emerging as potential solutions for complex tissue regeneration through cell-free therapies. In this review, we highlight the functional roles of EVs in the regenerative medicine for oral maxillofacial rehabilitation, specifically bone, skin, blood vessels, peripheral nerve and tooth-related tissue regeneration. Publications were reviewed by two researchers independently basing on three databases (PubMed, MEDLINE and Web of Science), until 31 December 2018. Basing on current researches, we classified the origin of EVs for regenerative medicine into four categories: related cells in the regenerative niche, mesenchymal stem cells, immune cells and body fluids. The secretome of different cells are distinct, while the same cells secrete different EVs under varied conditions; therefore, the content profiles of EVs and regulatory mechanisms on target cells are compared and emphasised. By unravelling the regulatory mechanisms of EVs in tissue regeneration, modified cells and tailored EVs with specific target may be produced for precision medicine with high efficacy.
Collapse
Affiliation(s)
- Longwei Lv
- Department of Prosthodontics, National Clinical Research Center for Oral Disease, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Chunhui Sheng
- Department of Prosthodontics, National Clinical Research Center for Oral Disease, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yongsheng Zhou
- Department of Prosthodontics, National Clinical Research Center for Oral Disease, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
47
|
Li D, Tian Y, Hu Y, Qi Y, Tian N, Li S, Hu P, Wu F, Wei Q, Wei Z, Wang S, Yin B, Jiang T, Yuan J, Qiang B, Han W, Peng X. Glioma-associated human endothelial cell-derived extracellular vesicles specifically promote the tumourigenicity of glioma stem cells via CD9. Oncogene 2019; 38:6898-6912. [DOI: 10.1038/s41388-019-0903-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 10/07/2018] [Accepted: 05/01/2019] [Indexed: 11/09/2022]
|
48
|
Wang Y, Xie Y, Zhang A, Wang M, Fang Z, Zhang J. Exosomes: An emerging factor in atherosclerosis. Biomed Pharmacother 2019; 115:108951. [PMID: 31078042 DOI: 10.1016/j.biopha.2019.108951] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is the main reason for morbidity and death caused by cardiovascular disease which leads to approximately 20% of total death around the world. Exosomes secreted by the cells is a kind of extracellular vesicles with lipid bilayer structure, containing a variety of cell specific lipid, nucleic acid and protein, involved in intercellular communication, plays an important role in different physiological and pathological process. In recent years, with the deepening of research, the role of exosomes in cardiovascular diseases has received extensive attention. This review summarizes the roles of exosomes and exosome-derived from microRNAs, proteins and DNA as biomarkers in the development of atherosclerosis, and explores the mechanism of exosome-mediated intercellular crosstalk in atherosclerosis, providing potential roles for diagnosis and treatment.
Collapse
Affiliation(s)
- Yanan Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, 312 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China
| | - Yingyu Xie
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, 312 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China
| | - Ao Zhang
- 726 broadway, Epidemiology, College of global public health, New York University, New York, 10003, United States
| | - Mingyang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, 312 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China
| | - Zihan Fang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, 312 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China.
| |
Collapse
|
49
|
Deng W, Tang T, Hou Y, Zeng Q, Wang Y, Fan W, Qu S. Extracellular vesicles in atherosclerosis. Clin Chim Acta 2019; 495:109-117. [PMID: 30959044 DOI: 10.1016/j.cca.2019.04.051] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs), which exist in human blood, are increased in some inflammation-related cardiovascular diseases. EVs are involved in inflammation, immunity, signal transduction, cell survival and apoptosis, angiogenesis, thrombosis, and autophagy, all of which are highly significant for maintaining homeostasis and disease progression. Therefore, EVs are also associated with key steps in atherosclerosis, including cellular lipid metabolism, endothelial dysfunction and vascular wall inflammation, ultimately resulting in vascular remodelling. In this review, we summarize recent studies on EV contents and biological function, focusing on their potential effect in atherosclerosis, including cholesterol metabolism, vascular inflammation, angiogenesis, coagulation and the development of atherosclerotic lesions. EVs may represent potential biomarkers and pharmacological targets for atherosclerotic diseases.
Collapse
Affiliation(s)
- WenYi Deng
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - TingTing Tang
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - YangFeng Hou
- Clinic Medicine Department, Hengyang Medical School, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Qian Zeng
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - YuFei Wang
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - WenJing Fan
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China; Emergency Department, The Second Affiliated Hospital, University of south China, Hengyang City, Hunan Province 421001, PR China.
| | - ShunLin Qu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang City, Hunan Province 421001, PR China.
| |
Collapse
|
50
|
Li K, Chen Y, Li A, Tan C, Liu X. Exosomes play roles in sequential processes of tumor metastasis. Int J Cancer 2019; 144:1486-1495. [PMID: 30155891 DOI: 10.1002/ijc.31774] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/22/2018] [Accepted: 07/17/2018] [Indexed: 02/05/2023]
Abstract
Overwhelming evidence demonstrates that exosomes, a series of biologically functional small vesicles of endocytic origin carrying a variety of active constituents, especially tumor-derived exosomes, contribute to tumor progression and metastasis. This review focuses on the specific multifaceted roles of exosomes in affecting sequenced four crucial processes of metastasis, through which cancer cells spread from primary to secondary organs and finally form macroscopic metastatic lesions. First, exosomes modulate the primary tumor sites to assist cancer growth and dissemination. In this part, five main biological events are reviewed, including the transfer of oncogenic constituents, the recruitment and activation of fibroblasts, the induction of angiogenesis, immunosuppression and epithelial-mesenchymal transition (EMT) promotion. In Step 2, we list two recently disclosed mechanisms during the organ-specific homing process: the exosomal integrin model and exosomal epidermal growth factor receptor (EGFR)/miR-26/hepatocyte growth factor (HGF) model. Subsequently, Step 3 focuses on the interactions between exosomes and pre-metastatic niche, in which we highlight the specific functions of exosomes in angiogenesis, lymphangiogenesis, immune modulation and metabolic, epigenetic and stromal reprogramming of pre-metastatic niche. Finally, we summarize the mechanisms of exosomes in helping the metastatic circulating tumor cells escape from immunologic surveillance, survive in the blood circulation and proliferate in host organs.
Collapse
Affiliation(s)
- Keyu Li
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yonghua Chen
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ang Li
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chunlu Tan
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xubao Liu
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|