1
|
D’Amico G, Carista A, Manna OM, Paladino L, Picone D, Sarullo S, Sausa M, Cappello F, Vitale AM, Caruso Bavisotto C. Brain-Periphery Axes: The Potential Role of Extracellular Vesicles-Delivered miRNAs. BIOLOGY 2024; 13:1056. [PMID: 39765723 PMCID: PMC11673379 DOI: 10.3390/biology13121056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025]
Abstract
Bidirectional communication between the central nervous system (CNS) and peripheral organs and tissue has been widely documented in physiological and pathological conditions. This communication relies on the bilateral transmission of signaling molecules and substances that circulate throughout the body and reach their target site(s) via the blood and other biological fluids (e.g., the cerebrospinal fluid, the lymph). One of the mechanisms by which these molecular messengers are exchanged is through the secretion of extracellular vesicles (EVs). EVs are known to mediate cell-to-cell communication by delivering biological molecules, including nucleic acids, proteins, lipids, and various other bioactive regulators. Moreover, EVs can cross the blood-brain barrier (BBB), enabling direct communication between the periphery and the brain. In particular, the delivery of microRNAs (miRNAs) can modulate the expression profiles of recipient cells, thereby influencing their functions. This review synthesizes current findings about the brain-periphery cross-talk mediated by EVs-delivered miRNAs. Although this mechanism has been definitively shown in a few cases, much evidence indirectly indicates that it could mediate brain-peripherical organs/tissue communication, especially in pathological conditions. Therefore, understanding this process could provide valuable insights for the treatment and management of neurological and systemic diseases.
Collapse
Affiliation(s)
- Giuseppa D’Amico
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.D.); (A.C.); (O.M.M.); (L.P.); (D.P.); (S.S.); (F.C.); (C.C.B.)
| | - Adelaide Carista
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.D.); (A.C.); (O.M.M.); (L.P.); (D.P.); (S.S.); (F.C.); (C.C.B.)
| | - Olga Maria Manna
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.D.); (A.C.); (O.M.M.); (L.P.); (D.P.); (S.S.); (F.C.); (C.C.B.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Via Michele Miraglia 20, 90139 Palermo, Italy
| | - Letizia Paladino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.D.); (A.C.); (O.M.M.); (L.P.); (D.P.); (S.S.); (F.C.); (C.C.B.)
| | - Domiziana Picone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.D.); (A.C.); (O.M.M.); (L.P.); (D.P.); (S.S.); (F.C.); (C.C.B.)
| | - Silvia Sarullo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.D.); (A.C.); (O.M.M.); (L.P.); (D.P.); (S.S.); (F.C.); (C.C.B.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Via Michele Miraglia 20, 90139 Palermo, Italy
| | - Martina Sausa
- Department of Theoretical and Applied Sciences, eCampus University, 22060 Novedrate, Italy;
| | - Francesco Cappello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.D.); (A.C.); (O.M.M.); (L.P.); (D.P.); (S.S.); (F.C.); (C.C.B.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Via Michele Miraglia 20, 90139 Palermo, Italy
| | - Alessandra Maria Vitale
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.D.); (A.C.); (O.M.M.); (L.P.); (D.P.); (S.S.); (F.C.); (C.C.B.)
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.D.); (A.C.); (O.M.M.); (L.P.); (D.P.); (S.S.); (F.C.); (C.C.B.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Via Michele Miraglia 20, 90139 Palermo, Italy
| |
Collapse
|
2
|
Moreno A, Alarcón-Zapata P, Guzmán-Gútierrez E, Radojkovic C, Contreras H, Nova-Lampeti E, A Zúñiga F, Rodriguez-Alvárez L, Escudero C, Lagos P, Aguayo C. Changes in the Release of Endothelial Extracellular Vesicles CD144+, CCR6+, and CXCR3+ in Individuals with Acute Myocardial Infarction. Biomedicines 2024; 12:2119. [PMID: 39335632 PMCID: PMC11430588 DOI: 10.3390/biomedicines12092119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/09/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Acute myocardial infarction (AMI) results from vulnerable plaque rupture, causing ischemic cardiomyocyte necrosis and intense inflammation. Paradoxically, this inflammation releases factors that aid heart repair. Recent findings suggest a role for extracellular vesicles (EVs) in intercellular communication during post-AMI cardiac repair. However, EVs' tissue origin and chemokine profile in the blood of patients with AMI remains unclear. This study characterized the tissue origin and chemokine receptor profile of EVs in the coronary and peripheral blood of patients with AMI. The results reveal that vesicles isolated from coronary and peripheral blood plasma are enriched in tetraspanin (CD9) and express CD81+, CD90+, and CD144+. The vesicle size ranged between 145 and 162 nm, with the control group exhibiting smaller vesicles (D10) than the AMI group. Furthermore, all vesicles expressed CCR6 and CXCR3, whereas a small percentage expressed CCR4. In addition, a decrease in CXCR3 and CCR6 expression was observed in coronary and peripheral AMI blood vesicles compared with the control; however, no difference was found between AMI coronary and AMI peripheral blood vesicles. In conclusion, our study demonstrates, for the first time, changes in the number of extracellular vesicles expressing CD144+, CXCR3, and CCR6 in the peripheral circulation of patients with AMI. Extracellular vesicles present in the circulation of patients with AMI hold excellent promise as a potential diagnostic tool.
Collapse
Affiliation(s)
- Alexa Moreno
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
- Clinical Laboratory Program, Faculty of Health Sciences, State University of Southern Manabí, Jipijapa 130402, Ecuador
| | - Pedro Alarcón-Zapata
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Enrique Guzmán-Gútierrez
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Claudia Radojkovic
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Héctor Contreras
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Estefanía Nova-Lampeti
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Felipe A Zúñiga
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Llerenty Rodriguez-Alvárez
- Department of Animal Science, Faculty of Veterinary Sciences, University of Concepcion, Chillán 3780000, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bio-Bio, Chillán 3780000, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán 3780000, Chile
| | - Paola Lagos
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Claudio Aguayo
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán 3780000, Chile
| |
Collapse
|
3
|
Hervás C, Peirotén I, González L, Alonso de Leciñana M, Alonso-López E, Casado L, De Celis-Ruíz E, Fernández Prieto AF, Frutos R, Gallego-Ruiz R, González Pérez de Villar N, Gutiérrez-Fernández M, Navia P, Otero-Ortega L, Pozo-Novoa J, Rigual R, Rodríguez-Pardo J, Ruiz G, Fuentes B. Glycaemia and ischaemia-reperfusion brain injury in patients with ischaemic stroke treated with mechanical thrombectomy (GLIAS-MT): an observational, unicentric, prospective study protocol. BMJ Open 2024; 14:e086745. [PMID: 39117402 PMCID: PMC11404171 DOI: 10.1136/bmjopen-2024-086745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
INTRODUCTION Poststroke hyperglycaemia is an independent risk factor for poorer outcomes in patients treated with mechanical thrombectomy (MT) and is associated with a lower probability of functional recovery and higher mortality at 3 months. This study aims to evaluate the association between glucose levels during cerebral reperfusion with MT and functional recovery at 3 months, measured by subcutaneous continuous glucose monitoring (CGM) devices. METHODS This prospective observational study aims to recruit 100 patients with ischaemic stroke and large anterior circulation vessel occlusion, in whom MT is indicated. CGM will be performed using a Freestyle Libre ProIQ device (FSL-CGM, Abbott Diabetes Care, Alameda, California, USA), which will be implanted on admission to the emergency department, to monitor glucose levels before, during and after reperfusion. The study's primary endpoint will be the functional status at 3 months, as measured by the dichotomised modified Rankin Scale (0-2 indicating good recovery and 3-6 indicating dependency or death). We will analyse expression profiles of microRNA (miRNA) at the time of reperfusion and 24 hours later, as potential biomarkers of ischaemic-reperfusion injury. The most promising miRNAs include miR-100, miR-29b, miR-339, miR-15a and miR-424. All patients will undergo treatment according to current international recommendations and local protocols for the treatment of stroke, including intravenous thrombolysis if indicated. ETHICS AND DISSEMINATION This study (protocol V.1.1, dated 29 October 2021, code 6017) has been approved by the Clinical Research Ethics Committee of La Paz University Hospital (Madrid, Spain) and has been registered in ClinicalTrials.gov (NCT05871502). Study results will be disseminated through peer-reviewed publications in Open Access format and at conference presentations. TRIAL REGISTRATION NUMBER NCT05871502.
Collapse
Affiliation(s)
- Carlos Hervás
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Irene Peirotén
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Laura González
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - María Alonso de Leciñana
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Elisa Alonso-López
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Laura Casado
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Elena De Celis-Ruíz
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Andrés Francisco Fernández Prieto
- Department of Radiology (Neurointerventional Radiology), La Paz University Hospital, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Remedios Frutos
- Department of Radiology (Neurointerventional Radiology), La Paz University Hospital, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Rebeca Gallego-Ruiz
- Neurological Sciences and Cerebrovascular Research Laboratory, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Noemí González Pérez de Villar
- Diabetes Unit, Department of Endocrinology, La Paz University Hospital, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Pedro Navia
- Department of Radiology (Neurointerventional Radiology), La Paz University Hospital, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Laura Otero-Ortega
- Neurological Sciences and Cerebrovascular Research Laboratory, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Javier Pozo-Novoa
- Neurological Sciences and Cerebrovascular Research Laboratory, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Ricardo Rigual
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Jorge Rodríguez-Pardo
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Gerardo Ruiz
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Blanca Fuentes
- Department of Neurology and Stroke Centre, La Paz University Hospital and Department of Medicine, Universidad Autónoma de Madrid, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| |
Collapse
|
4
|
Fullerton JL, Cosgrove CC, Rooney RA, Work LM. Extracellular vesicles and their microRNA cargo in ischaemic stroke. J Physiol 2023; 601:4907-4921. [PMID: 35421904 PMCID: PMC10952288 DOI: 10.1113/jp282050] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/15/2022] [Indexed: 11/08/2022] Open
Abstract
Acute ischaemic stroke (AIS) is a leading cause of death and disability. MicroRNAs (miRNAs) are short non-coding RNAs which hold the potential to act as a novel biomarker in AIS. The majority of circulating miRNAs are actively encapsulated by extracellular vesicles (EVs) produced by many cells and organs endogenously. EVs released by mesenchymal stem cells (MSCs) have been extensively studied for their therapeutic potential. In health and disease, EVs are vital for intercellular communication, as the cargo within EVs can be exchanged between neighbouring cells or transported to distant sites. It is clear here from both current preclinical and clinical studies that AIS is associated with specific EV-derived miRNAs, including those transported via MSC-derived EVs. In addition, current studies provide evidence to show that modulating levels of specific EV-derived miRNAs in AIS provides a novel therapeutic potential of miRNAs in the treatment of stroke. Commonalities exist in altered miRNAs across preclinical and clinical studies. Of those EV-packaged miRNAs, miRNA-124 was described both as an EV-packaged biomarker and as a potential EV-loaded therapeutic in experimental models. Alterations of miRNA-17 family and miRNA-17-92 cluster were identified in preclinical, clinical and MSC-EV-mediated neuroprotection in experimental stroke. Finally, miRNA-30d and -30a were found to mediate therapeutic effect when overexpressed from MSC and implicated as a biomarker clinically. Combined, EV-derived miRNAs will further our understanding of the neuropathological processes triggered by AIS. In addition, this work will help determine the true clinical value of circulating EV-packaged miRNAs as biomarkers of AIS or as novel therapeutics in this setting.
Collapse
Affiliation(s)
- Josie L. Fullerton
- Institute of Cardiovascular and Medical Sciences College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Caitlin C. Cosgrove
- Institute of Cardiovascular and Medical Sciences College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Rebecca A. Rooney
- Institute of Cardiovascular and Medical Sciences College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Lorraine M. Work
- Institute of Cardiovascular and Medical Sciences College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
5
|
Zhou W, Zhao L, Mao Z, Wang Z, Zhang Z, Li M. Bidirectional Communication Between the Brain and Other Organs: The Role of Extracellular Vesicles. Cell Mol Neurobiol 2023; 43:2675-2696. [PMID: 37067749 PMCID: PMC10106324 DOI: 10.1007/s10571-023-01345-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/03/2023] [Indexed: 04/18/2023]
Abstract
A number of substances released by the brain under physiological and pathological conditions exert effects on other organs. In turn, substances produced primarily by organs such as bone marrow, adipose tissue, or the heart may have an impact on the metabolism and function and metabolism of the healthy and diseased brain. Despite a mounting amount of evidence supports such bidirectional communication between the brain and other organs, research on the function of molecular mediators carried by extracellular vesicles (EVs) is in the early stages. In addition to being able to target or reach practically any organ, EVs have the ability to cross the blood-brain barrier to transport a range of substances (lipids, peptides, proteins, and nucleic acids) to recipient cells, exerting biological effects. Here, we review the function of EVs in bidirectional communication between the brain and other organs. In a small number of cases, the role has been explicitly proven; yet, in most cases, it relies on indirect evidence from EVs in cell culture or animal models. There is a dearth of research currently available on the function of EVs-carrying mediators in the bidirectional communication between the brain and bone marrow, adipose tissue, liver, heart, lungs, and gut. Therefore, more studies are needed to determine how EVs facilitate communication between the brain and other organs.
Collapse
Affiliation(s)
- Wu Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China
| | - Lihong Zhao
- Department of Radiotherapy, Jilin Cancer Hospital, 1018 Huguang Street, Changchun, 130012, Jilin, China
| | - Zelu Mao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China
| | - Zhihua Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China
| | - Zhixiong Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
6
|
Extracellular vesicles: Critical bilateral communicators in periphery-brain crosstalk in central nervous system disorders. Biomed Pharmacother 2023; 160:114354. [PMID: 36753954 DOI: 10.1016/j.biopha.2023.114354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Growing evidence shows that there is a comorbid mechanism between the central nervous system (CNS) and the peripheral organs. The bilateral transmission of signal molecules in periphery-brain crosstalk plays an important role in the underlying mechanism, which result from complex networks of neurohumoral circuits. Secreted by almost all cells and considered innovative information transport systems, extracellular vesicles (EVs) encapsulate and deliver nucleic acids, proteins, lipids, and various other bioactive regulators. Moreover, EVs can cross the blood-brain barrier (BBB), they are also identified primarily as essential communicators between the periphery and the CNS. In addition to transporting molecules under physiological or pathological conditions, EVs also show novel potential in targeted drug delivery. In this review, we discuss the mechanisms implicated in the transport of EVs in crosstalk between the peripheral and the central immune systems as well as in crosstalk between the peripheral organs and the brain in CNS disorders, especially in neurodegenerative diseases, stroke, and trauma. This work will help in elucidating the contributions of EVs to brain health and disorders, and promote the development of new strategies for minimally invasive treatment.
Collapse
|
7
|
Kim SY, Jang S, Lee S, Park JT, Lee SJ, Kim HS. Characterization of Exosomes and Exosomal RNAs Isolated from Post-Mortem Body Fluids for Molecular Forensic Diagnosis. Diagnostics (Basel) 2022; 12:2153. [PMID: 36140554 PMCID: PMC9498102 DOI: 10.3390/diagnostics12092153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/01/2022] [Accepted: 09/03/2022] [Indexed: 11/25/2022] Open
Abstract
Exosomes have been mainly studied for their potential applications in biomarker detection and drug delivery for diagnosis and treatment. However, in the field of forensic research, the potential value of exosomes derived from post-mortem body fluids has not been investigated to date. Here, we isolated the exosomes and exosomal RNAs from post-mortem body fluids, including cardiac blood, pericardial fluid, and urine. We also compared commercial exosome isolation kits to determine the optimal method for post-mortem exosome isolation. Transmission electron microscopy (TEM), the Agilent bioanalyzer system, and western blotting were used to evaluate the efficiencies of alternative isolation methods and the characteristics of isolated exosomes. There were no significant differences between exosomes obtained from post-mortem and ante-mortem body fluids in the expression of exosome surface markers or morphology. The exosomes were well-preserved even under simulated post-mortem conditions. Among the isolation procedures tested, the membrane affinity column-based method was the most suitable for post-mortem exosomal RNA isolation. These results suggest that exosomes are well-preserved in post-mortem body fluids and could be utilized for forensic diagnosis.
Collapse
Affiliation(s)
- So-Yeon Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Sinae Jang
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Korea
- Brain Korea 21 Plus Program, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Sookyoung Lee
- Division of Forensic Medical Examination, National Forensic Service, Wonju 26460, Korea
| | - Jong-Tae Park
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Su-Jin Lee
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Korea
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Korea
| |
Collapse
|
8
|
Abstract
Stroke remains a leading cause of death and disability, with limited therapeutic options and suboptimal tools for diagnosis and prognosis. High throughput technologies such as proteomics generate large volumes of experimental data at once, thus providing an advanced opportunity to improve the status quo by facilitating identification of novel therapeutic targets and molecular biomarkers. Proteomics studies in animals are largely designed to decipher molecular pathways and targets altered in brain tissue after stroke, whereas studies in human patients primarily focus on biomarker discovery in biofluids and, more recently, in thrombi and extracellular vesicles. Here, we offer a comprehensive review of stroke proteomics studies conducted in both animal and human specimen and present our view on limitations, challenges, and future perspectives in the field. In addition, as a unique resource for the scientific community, we provide extensive lists of all proteins identified in proteomic studies as altered by stroke and perform postanalysis of animal data to reveal stroke-related cellular processes and pathways.
Collapse
Affiliation(s)
- Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY (K.H.)
| | - Wei Yang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University School of Medicine, Durham, NC (W.Y.)
| |
Collapse
|
9
|
Gelosa P, Castiglioni L, Rzemieniec J, Muluhie M, Camera M, Sironi L. Cerebral derailment after myocardial infarct: mechanisms and effects of the signaling from the ischemic heart to brain. J Mol Med (Berl) 2022; 100:23-41. [PMID: 34674004 PMCID: PMC8724191 DOI: 10.1007/s00109-021-02154-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/07/2021] [Accepted: 10/14/2021] [Indexed: 12/04/2022]
Abstract
Myocardial infarction (MI) is the leading cause of death among ischemic heart diseases and is associated with several long-term cardiovascular complications, such as angina, re-infarction, arrhythmias, and heart failure. However, MI is frequently accompanied by non-cardiovascular multiple comorbidities, including brain disorders such as stroke, anxiety, depression, and cognitive impairment. Accumulating experimental and clinical evidence suggests a causal relationship between MI and stroke, but the precise underlying mechanisms have not yet been elucidated. Indeed, the risk of stroke remains a current challenge in patients with MI, in spite of the improvement of medical treatment among this patient population has reduced the risk of stroke. In this review, the effects of the signaling from the ischemic heart to the brain, such as neuroinflammation, neuronal apoptosis, and neurogenesis, and the possible actors mediating these effects, such as systemic inflammation, immunoresponse, extracellular vesicles, and microRNAs, are discussed.
Collapse
Affiliation(s)
- Paolo Gelosa
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Laura Castiglioni
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Joanna Rzemieniec
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Majeda Muluhie
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Marina Camera
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
- Centro Cardiologico Monzino, 20138, Milan, Italy
| | - Luigi Sironi
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy.
| |
Collapse
|
10
|
Otero-Ortega L, Gutiérrez-Fernández M, Díez-Tejedor E. Recovery After Stroke: New Insight to Promote Brain Plasticity. Front Neurol 2021; 12:768958. [PMID: 34867756 PMCID: PMC8639681 DOI: 10.3389/fneur.2021.768958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/18/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Laura Otero-Ortega
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of Hospital La Paz Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of Hospital La Paz Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Exuperio Díez-Tejedor
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of Hospital La Paz Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
11
|
Potential Applications and Functional Roles of Exosomes in Cardiometabolic Disease. Pharmaceutics 2021; 13:pharmaceutics13122056. [PMID: 34959338 PMCID: PMC8703910 DOI: 10.3390/pharmaceutics13122056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/13/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Abstract
Despite diagnostic and therapeutic advances, cardiometabolic disease remains the leading cause of death worldwide. Extracellular vesicles (EVs), which include exosomes and microvesicles, have gained particular interest because of their role in metabolic homeostasis and cardiovascular physiology. Indeed, EVs are recognized as critical mediators of intercellular communication in the cardiovascular system. Exosomes are naturally occurring nanocarriers that transfer biological information in the setting of metabolic abnormalities and cardiac dysfunction. The study of these EVs can increase our knowledge on the pathophysiological mechanisms of metabolic disorders and their cardiovascular complications. Because of their inherent properties and composition, exosomes have been proposed as diagnostic and prognostic biomarkers and therapeutics for specific targeting and drug delivery. Emerging fields of study explore the use exosomes as tools for gene therapy and as a cell-free alternative for regenerative medicine. Furthermore, innovative biomaterials can incorporate exosomes to enhance tissue regeneration and engineering. In this work, we summarize the most recent knowledge on the role of exosomes in cardiometabolic pathophysiology while highlighting their potential therapeutic applications.
Collapse
|
12
|
Zheng S, Wu R, Deng Y, Zhang Q. Dihydroartemisinin represses oral squamous cell carcinoma progression through downregulating mitochondrial calcium uniporter. Bioengineered 2021; 13:227-241. [PMID: 34847839 PMCID: PMC8805845 DOI: 10.1080/21655979.2021.2012951] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Dysregulation of mitochondrial calcium uniporter (MCU) exerts a carcinogenic effect in several cancers. Nevertheless, the roles of MCU in oral squamous cell carcinoma (OSCC) remain elusive. It has been reported that dihydroartemisinin (DHA) may suppress the progression of OSCC but its associated mechanisms have not been investigated. The purpose of our research was to observe the biological function of MCU on OSCC and its regulatory relationship with DHA. MCU, MICU1, MICU2, N-cadherin, TGF-β and vimentin expression was detected in OSCC and peritumoral tissues by immunohistochemistry and Western blot. Following DHA treatment, the expression of the aforementioned proteins was detected in CAL-27 cells transfected with shMCU or pcDNA3.1-MCU by Western blot or immunofluorescence. Furthermore, clone formation, mitochondrial membrane potential (MMP), wound healing and transwell assays were presented in CAL-27 cells treated with DHA, shMCU or pcDNA3.1-MCU. Our results showed that the members of MCU complex (MCU, MICU1 and MICU2) were overexpressed in OSCC than peritumoral tissues. Furthermore, TGF-β and epithelial to mesenchymal transition (EMT) proteins (N-cadherin and vimentin) exhibited higher expression in OSCC. DHA treatment significantly lowered the expression of MCU in CAL-27 cells. MCU overexpression reversed the inhibitory effects of DHA on MICU1, MICU2, N-cadherin, TGF-β and vimentin. MCU knockdown or DHA suppressed proliferation, MMP and migration of CAL-27 cells. DHA treatment could reverse the effects of MCU overexpression. Collectively, our study demonstrated that MCU was an oncogene of OSCC and DHA exerted a suppressive role on proliferation and migration of OSCC cells by suppressing MCU expression.
Collapse
Affiliation(s)
- Shen Zheng
- Department of Orthodontics and Prosthodontics, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Ran Wu
- Department of Stomatology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Yunlong Deng
- Department of Stomatology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Qiang Zhang
- Department of Orthodontics and Prosthodontics, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| |
Collapse
|
13
|
Li C, Ke C, Su Y, Wan C. Exercise Intervention Promotes the Growth of Synapses and Regulates Neuroplasticity in Rats With Ischemic Stroke Through Exosomes. Front Neurol 2021; 12:752595. [PMID: 34777222 PMCID: PMC8581302 DOI: 10.3389/fneur.2021.752595] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Stroke is the leading cause of death and disability. Exercise produces neuroprotection by improving neuroplasticity. Exercise can induce exosome production. According to several studies, exosomes are involved in repairing brain function, but the relationship and mechanism of exercise, exosomes, and neuroprotection have not been elucidated. This study intends to explore the relationship and potential mechanism by observing the changes in the exosome level, infarct volume, neurological function and behavioral scores, synapses, and corticospinal tract (CST). Methods: Rats were randomly divided into four groups: a sham operation (SHAM) group, middle cerebral artery occlusion (MCAO) with sedentary intervention (SED-MCAO) group, MCAO with exercise intervention (EX-MCAO) group, and MCAO with exercise intervention and exosome injection (EX-MCAO-EXO) group. The exercise intervention was started 1 day after MCAO and lasted for 4 weeks. All rats were assessed using the modified neurological severity score (mNSS). The levels of exosomes in serum and brain, gait analysis, and magnetic resonance scan were performed 1 and 4 weeks after the intervention. After 4 weeks of intervention, the number of synapses, synaptophysin (Syn), and postsynaptic density protein 95(PSD-95) expression was detected. Results: After 4 weeks of intervention, (1) the EX-MCAO and EX-MCAO-EXO groups showed higher serum exosome (pEX−MCAO = 0.000, pEX−MCAO−EXO = 0.000) and brain exosome (pEX−MCAO = 0.001, pEX−MCAO−EXO = 0.000) levels than the SED-MCAO group, of which the EX-MCAO group had the highest serum exosome (p = 0.000) and the EX-MCAO-EXO group had the highest brain exosome (p = 0.03) levels. (2) The number of synapses in the EX-MCAO (p = 0.032) and EX-MCAO-EXO groups (p = 0.000) was significantly higher than that in the SED-MCAO group. The EX-MCAO-EXO group exhibited a greater number of synapses than the EX-MCAO (p = 0.000) group. (3) The synaptic plasticity-associated proteins were expressed significantly higher in the EX-MCAO (pSyn = 0.010, pPSD−95 = 0.044) and EX-MCAO-EXO (pSyn = 0.000, pPSD−95 = 0.000) groups than in the SED-MCAO group, and the EX-MCAO-EXO group (pSyn = 0.000, pPSD−95 = 0.046) had the highest expression. (4) Compared with the SED-MCAO group, the EX-MCAO group had significantly improved infarct volume ratio (p = 0.000), rFA value (p = 0.000), and rADC (p = 0.000). Compared with the EX-MCAO group, the EX-MCAO-EXO group had a significantly improved infarct volume ratio (p = 0.000), rFA value (p = 0.000), and rADC value (p = 0.001). (5) Compared with the SED-MCAO group, the EX-MCAO group (p = 0.001) and EX-MCAO-EXO group (p = 0.000) had significantly lower mNSS scores and improved gait. (6) The brain exosome levels were negatively correlated with the mNSS score, infarct volume ratio, and rADC value and positively correlated with the rFA value, Syn, and PSD-95 expression. The serum and brain exosome levels showed a positive correlation. Conclusions: Exercise intervention increases the serum exosome level in MCAO rats, which are recruited into the brain, leading to improved synaptic growth and CST integrity, a reduced infarct volume, and improved neurological function and gait.
Collapse
Affiliation(s)
- Chen Li
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Changkai Ke
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Yue Su
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunxiao Wan
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
14
|
Circulating Extracellular Vesicle Proteins and MicroRNA Profiles in Subcortical and Cortical-Subcortical Ischaemic Stroke. Biomedicines 2021; 9:biomedicines9070786. [PMID: 34356850 PMCID: PMC8301391 DOI: 10.3390/biomedicines9070786] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/28/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
In order to investigate the role of circulating extracellular vesicles (EVs), proteins, and microRNAs as damage and repair markers in ischaemic stroke depending on its topography, subcortical (SC), and cortical-subcortical (CSC) involvement, we quantified the total amount of EVs using an enzyme-linked immunosorbent assay technique and analysed their global protein content using proteomics. We also employed a polymerase chain reaction to evaluate the circulating microRNA profile. The study included 81 patients with ischaemic stroke (26 SC and 55 CSC) and 22 healthy controls (HCs). No differences were found in circulating EV levels between the SC, CSC, and HC groups. We detected the specific expression of C1QA and Casp14 in the EVs of patients with CSC ischaemic stroke and the specific expression of ANXA2 in the EVs of patients with SC involvement. Patients with CSC ischaemic stroke showed a lower expression of miR-15a, miR-424, miR-100, and miR-339 compared with those with SC ischaemic stroke, and the levels of miR-339, miR-100, miR-199a, miR-369a, miR-424, and miR-15a were lower than those of the HCs. Circulating EV proteins and microRNAs from patients with CSC ischaemic stroke could be considered markers of neurite outgrowth, neurogenesis, inflammation process, and atherosclerosis. On the other hand, EV proteins and microRNAs from patients with SC ischaemic stroke might be markers of an anti-inflammatory process and blood–brain barrier disruption reduction.
Collapse
|