1
|
Ramos A, Youssef L, Molina P, Torramadé-Moix S, Martinez-Sanchez J, Moreno-Castaño AB, Blasco M, Guillén-Olmos E, De Moner B, Pino M, Tortajada M, Camacho M, Borrell M, Crovetto F, Ramirez-Bajo MJ, Ventura-Aguiar P, Banon-Maneus E, Rovira J, Escolar G, Carreras E, Gratacos E, Diaz-Ricart M, Crispi F, Palomo M. Circulating extracellular vesicles and neutrophil extracellular traps contribute to endothelial dysfunction in preeclampsia. Front Immunol 2024; 15:1488127. [PMID: 39735539 PMCID: PMC11671372 DOI: 10.3389/fimmu.2024.1488127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Background Preeclampsia (PE) is a pregnancy complication characterized by hypertension, proteinuria, endothelial dysfunction, and complement dysregulation. Placenta-derived extracellular vesicles (EVs), necessary in maternal-fetal communication, might contribute to PE pathogenesis. Moreover, neutrophil extracellular traps (NETs) play a pathogenic role in other complement-mediated pathologies, and their contribution in PE remains unexplored. Materials and methods EVs were isolated from PE (peEVs) and normotensive pregnant women sera. NETs were obtained incubating donor-pre-activated neutrophils with PE or control sera. Microvascular (HMEC) endothelial cells (ECs) were incubated with PE or control sera with or without (depleted sera) EVs or NETs, to assess changes in VCAM-1, ICAM-1, VE-cadherin, eNOS, VWF, ROS, and C5b-9 deposits. Results were expressed as fold increase vs. control. Results VWF, VCAM-1, and ROS expression was significantly higher in cells exposed to PE sera vs. control (12.3 ± 8.1, 3.6 ± 2.3, and 1.8 ± 0.2, respectively, p < 0.05), though significantly lower in cells exposed to depleted PE (dPE) sera (6.1 ± 2.7, 0.7 ± 0.6, and 1.2 ± 0.1, respectively, vs. control, p < 0.05). EC exposure to depleted control sera supplemented with peEVs (dC+peEVs) significantly increased VWF, VCAM-1, and ROS compared to non-supplemented sera (4.5 ± 0.3, 2.8 ± 2.0, and 1.4 ± 0.2, respectively, p < 0.05). ICAM-1, VE-cadherin, and C5b-9 did not differ among groups. ECs incubated with PE-NETs increased VWF and VCAM-1 and decreased VE-cadherin expression vs. control (4 ± 1.6, 5.9 ± 1.2, and 0.5 ± 0.1, respectively, p < 0.05), and notably increased C5b-9 deposit (7.5 ± 2.9, p < 0.05). ICAM-1 and ROS did not differ. Conclusions Both circulating EVs and NETs from PE pregnant women exhibit a deleterious effect on ECs. Whereas EVs trigger a pro-oxidant and proinflammatory state, NETs potentiate the activation of the complement system, as already described in PE.
Collapse
Affiliation(s)
- Alex Ramos
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lina Youssef
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Patricia Molina
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Sergi Torramadé-Moix
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Julia Martinez-Sanchez
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ana Belen Moreno-Castaño
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Miquel Blasco
- Nephrology and Kidney Transplant Department, Center of Reference in Complex Glomerular Disease (CSUR), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud RICORS2040, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elena Guillén-Olmos
- Nephrology and Kidney Transplant Department, Center of Reference in Complex Glomerular Disease (CSUR), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Blanca De Moner
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Marc Pino
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Marta Tortajada
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Marta Camacho
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Maria Borrell
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Francesca Crovetto
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Maria Jose Ramirez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pedro Ventura-Aguiar
- Nephrology and Kidney Transplant Department, Center of Reference in Complex Glomerular Disease (CSUR), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elisenda Banon-Maneus
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gines Escolar
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Enric Carreras
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Eduard Gratacos
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
| | - Maribel Diaz-Ricart
- Hemostasis and Erythropathology Laboratory, Hematopathology, Department of Pathology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Fatima Crispi
- BCNatal | Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Palomo
- Barcelona Endothelium Team, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematology External Quality Assessment Laboratory, Biomedical Diagnostic Center, Hospital Clinic of Barcelona, Barcelona, Spain
| |
Collapse
|
2
|
Aquino A, Abutalimova N, Ma Y, Ismail-zade I, Grebennik V, Rubinstein A, Kudryavtsev I, Zaikova E, Sambur D, Marichev A, Kalinina O, Bautin A, Kostareva A, Vaage J, Golovkin A. Differences in Plasma Extracellular Vesicles of Different Origin in On-Pump Versus Off-Pump Cardiac Surgery. Curr Issues Mol Biol 2024; 46:13058-13077. [PMID: 39590373 PMCID: PMC11593215 DOI: 10.3390/cimb46110779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Coronary artery bypass grafting (CABG) using cardiopulmonary bypass (CPB) causes a systemic inflammatory response that can worsen patient outcomes. Off-pump surgery has been associated with a reduced inflammatory response. The precise mechanisms and the role of extracellular vesicles (EVs) in this context are not fully understood. This study aimed to investigate the early immune response, including main T- and B-lymphocyte subsets, cytokine profiles, and plasma EVs, in patients undergoing off-pump (n = 18) and on-pump (n = 18) CABG. Thirty-six patients undergoing isolated CABG were enrolled in this randomized control study. Pre- and 24 h postoperative blood samples were analyzed for immune cell populations, cytokine levels, and plasma EV phenotyping. Off-pump CABG triggered a milder immune response than on-pump surgery. On-pump surgery led to greater changes in circulating EVs, particularly platelet- (CD62P+), endothelial- (CD31+), and B-cell-derived (CD19+), as well as platelet- and erythrocyte-derived aggregates (CD41+CD235a+). Levels of platelet-derived EVs, expressing both constitutional and activation markers (CD41+CD62P+) decreased in both groups of patients 24 h after surgery. On-pump cardiac procedures led to an increase in T-regulatory cell-derived EVs (CD73+CD39+), suggesting a potential mechanism for immune suppression compared to off-pump surgery. There were numerous correlations between EV levels and cytokine profiles following on-pump surgery, hinting at a close relationship. Leucocyte-derived EVs exhibited positive correlations with each other and with GRO but showed negative correlations with endothelial-derived EVs (CD90+ and CD31+). Additionally, CD73+ EVs demonstrated positive correlations with platelet counts and with erythrocyte-derived CD235a+ EVs. EV changes were significantly greater after on-pump surgery, highlighting a more pronounced response to this type of surgery and emphasizing the role of EVs as regulators of post-surgical inflammation.
Collapse
Affiliation(s)
- Arthur Aquino
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Napisat Abutalimova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Yi Ma
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Imran Ismail-zade
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Vadim Grebennik
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Artem Rubinstein
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
- Institute of Experimental Medicine, 197022 St. Petersburg, Russia
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
- Institute of Experimental Medicine, 197022 St. Petersburg, Russia
| | - Ekatherina Zaikova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Darina Sambur
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Alexander Marichev
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Olga Kalinina
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Andrey Bautin
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Anna Kostareva
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Jarle Vaage
- Oslo University Hospital, University of Oslo, 0372 Oslo, Norway;
| | - Alexey Golovkin
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| |
Collapse
|
3
|
Kaur M, Fusco S, Van den Broek B, Aseervatham J, Rostami A, Iacovitti L, Grassi C, Lukomska B, Srivastava AK. Most recent advances and applications of extracellular vesicles in tackling neurological challenges. Med Res Rev 2024; 44:1923-1966. [PMID: 38500405 DOI: 10.1002/med.22035] [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: 01/02/2024] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
Over the past few decades, there has been a notable increase in the global burden of central nervous system (CNS) diseases. Despite advances in technology and therapeutic options, neurological and neurodegenerative disorders persist as significant challenges in treatment and cure. Recently, there has been a remarkable surge of interest in extracellular vesicles (EVs) as pivotal mediators of intercellular communication. As carriers of molecular cargo, EVs demonstrate the ability to traverse the blood-brain barrier, enabling bidirectional communication. As a result, they have garnered attention as potential biomarkers and therapeutic agents, whether in their natural form or after being engineered for use in the CNS. This review article aims to provide a comprehensive introduction to EVs, encompassing various aspects such as their diverse isolation methods, characterization, handling, storage, and different routes for EV administration. Additionally, it underscores the recent advances in their potential applications in neurodegenerative disorder therapeutics. By exploring their unique capabilities, this study sheds light on the promising future of EVs in clinical research. It considers the inherent challenges and limitations of these emerging applications while incorporating the most recent updates in the field.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Bram Van den Broek
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jaya Aseervatham
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Lorraine Iacovitti
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Jefferson Stem Cell and Regenerative Neuroscience Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Amit K Srivastava
- Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Hussain MT, Austin-Williams S, Wright TD, Dhawan UK, Pinto AL, Cooper D, Norling LV. β1-Integrin-Mediated Uptake of Chondrocyte Extracellular Vesicles Regulates Chondrocyte Homeostasis. Int J Mol Sci 2024; 25:4756. [PMID: 38731975 PMCID: PMC11083596 DOI: 10.3390/ijms25094756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Osteoarthritis (OA) is the most prevalent age-related degenerative disorder, which severely reduces the quality of life of those affected. Whilst management strategies exist, no cures are currently available. Virtually all joint resident cells generate extracellular vesicles (EVs), and alterations in chondrocyte EVs during OA have previously been reported. Herein, we investigated factors influencing chondrocyte EV release and the functional role that these EVs exhibit. Both 2D and 3D models of culturing C28I/2 chondrocytes were used for generating chondrocyte EVs. We assessed the effect of these EVs on chondrogenic gene expression as well as their uptake by chondrocytes. Collectively, the data demonstrated that chondrocyte EVs are sequestered within the cartilage ECM and that a bi-directional relationship exists between chondrocyte EV release and changes in chondrogenic differentiation. Finally, we demonstrated that the uptake of chondrocyte EVs is at least partially dependent on β1-integrin. These results indicate that chondrocyte EVs have an autocrine homeostatic role that maintains chondrocyte phenotype. How this role is perturbed under OA conditions remains the subject of future work.
Collapse
Affiliation(s)
- Mohammed Tayab Hussain
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Shani Austin-Williams
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Thomas Dudley Wright
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Umesh Kumar Dhawan
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Andreia L. Pinto
- Royal Brompton & Harefield NHS Foundation Trust, London SW3 6PY, UK;
| | - Dianne Cooper
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
| | - Lucy V. Norling
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
5
|
Kaurani L. Clinical Insights into MicroRNAs in Depression: Bridging Molecular Discoveries and Therapeutic Potential. Int J Mol Sci 2024; 25:2866. [PMID: 38474112 PMCID: PMC10931847 DOI: 10.3390/ijms25052866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Depression is a major contributor to the overall global burden of disease. The discovery of biomarkers for diagnosis or prediction of treatment responses and as therapeutic agents is a current priority. Previous studies have demonstrated the importance of short RNA molecules in the etiology of depression. The most extensively researched of these are microRNAs, a major component of cellular gene regulation and function. MicroRNAs function in a temporal and tissue-specific manner to regulate and modify the post-transcriptional expression of target mRNAs. They can also be shuttled as cargo of extracellular vesicles between the brain and the blood, thus informing about relevant mechanisms in the CNS through the periphery. In fact, studies have already shown that microRNAs identified peripherally are dysregulated in the pathological phenotypes seen in depression. Our article aims to review the existing evidence on microRNA dysregulation in depression and to summarize and evaluate the growing body of evidence for the use of microRNAs as a target for diagnostics and RNA-based therapies.
Collapse
Affiliation(s)
- Lalit Kaurani
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany
| |
Collapse
|
6
|
Chang H, Chen E, Hu Y, Wu L, Deng L, Ye‐Lehmann S, Mao X, Zhu T, Liu J, Chen C. Extracellular Vesicles: The Invisible Heroes and Villains of COVID-19 Central Neuropathology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305554. [PMID: 38143270 PMCID: PMC10933635 DOI: 10.1002/advs.202305554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/18/2023] [Indexed: 12/26/2023]
Abstract
Acknowledging the neurological symptoms of COVID-19 and the long-lasting neurological damage even after the epidemic ends are common, necessitating ongoing vigilance. Initial investigations suggest that extracellular vesicles (EVs), which assist in the evasion of the host's immune response and achieve immune evasion in SARS-CoV-2 systemic spreading, contribute to the virus's attack on the central nervous system (CNS). The pro-inflammatory, pro-coagulant, and immunomodulatory properties of EVs contents may directly drive neuroinflammation and cerebral thrombosis in COVID-19. Additionally, EVs have attracted attention as potential candidates for targeted therapy in COVID-19 due to their innate homing properties, low immunogenicity, and ability to cross the blood-brain barrier (BBB) freely. Mesenchymal stromal/stem cell (MSCs) secreted EVs are widely applied and evaluated in patients with COVID-19 for their therapeutic effect, considering the limited antiviral treatment. This review summarizes the involvement of EVs in COVID-19 neuropathology as carriers of SARS-CoV-2 or other pathogenic contents, as predictors of COVID-19 neuropathology by transporting brain-derived substances, and as therapeutic agents by delivering biotherapeutic substances or drugs. Understanding the diverse roles of EVs in the neuropathological aspects of COVID-19 provides a comprehensive framework for developing, treating, and preventing central neuropathology and the severe consequences associated with the disease.
Collapse
Affiliation(s)
- Haiqing Chang
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Erya Chen
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Yi Hu
- Department of Cardiology, Honghui hospitalXi'an Jiaotong UniversityXi'an710049China
| | - Lining Wu
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Liyun Deng
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Shixin Ye‐Lehmann
- Diseases and Hormones of the Nervous System University of Paris‐Scalay Bicêtre Hosptial BâtGrégory Pincus 80 Rue du Gal Leclerc, CedexLe Kremlin Bicêtre94276France
| | - Xiaobo Mao
- Department of NeurologyInstitute of Cell EngineeringSchool of MedicineJohns Hopkins UniversityBaltimoreMD21218USA
| | - Tao Zhu
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Jin Liu
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Chan Chen
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| |
Collapse
|
7
|
Feng Q, Zhang Y, Fang Y, Kong X, He Z, Ji J, Yang X, Zhai G. Research progress of exosomes as drug carriers in cancer and inflammation. J Drug Target 2023; 31:335-353. [PMID: 36543743 DOI: 10.1080/1061186x.2022.2162059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Extracellular vesicles (EVs) could be produced by most cells and play an important role in disease development. As a subtype of EVs, exosomes exhibit suitable size, rich surface markers and diverse contents, making them more appealing as potential drug carriers. Compared with traditional synthetic nanoparticles, exosomes possess superior biocompatibility and much lower immunogenicity. This work reviewed the most up-to-date research progress of exosomes as carriers for nucleic acids, proteins and small molecule drugs for cancer and inflammation management. The drug loading strategies and potential cellular uptake behaviour of exosomes are highlighted, trying to provide reference for future exosome design and application.
Collapse
Affiliation(s)
- Qixiang Feng
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Yu Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Yuelin Fang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Xinru Kong
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Zhijing He
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
8
|
Groves AM, Paris N, Hernady E, Johnston CJ, Aljitawi O, Lee YF, Kerns SL, Marples B. Prevention of Radiation-Induced Bladder Injury: A Murine Study Using Captopril. Int J Radiat Oncol Biol Phys 2023; 115:972-982. [PMID: 36400304 DOI: 10.1016/j.ijrobp.2022.10.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 10/04/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022]
Abstract
PURPOSE Pelvic radiation therapy (RT) can cause debilitating bladder toxicities but few clinical interventions exist to prevent injury or alleviate symptoms. From a large genome-wide association study in patients with prostate cancer it was previously reported that SNPs tagging AGT, part of the renin-angiotensin system (RAS), correlated with patient-reported late hematuria, identifying a potential targetable pathway to prevent RT-induced bladder injury. To investigate this association, we performed a preclinical study to determine whether RAS modulation protected the bladder against RT injury. METHODS AND MATERIALS C57BL/6 male mice were treated with an oral angiotensin converting enzyme inhibitor (ACEi: 0.3g/L captopril) 5 days before focal bladder X-irradiation with either single dose (SD) 30 Gy or 3 fractions of 8 Gy (8 Gy × 3 in 5 days). RT was delivered using XStrahl SARRP Muriplan CT-image guidance with parallel-opposed lateral beams. ACEi was maintained for 20 weeks post RT. Bladder toxicity was assessed using assays to identify local injury that included urinalysis, functional micturition, bladder-released exosomes, and histopathology, as well as an assessment of systemic changes in inflammatory-mediated circulating immune cells. RESULTS SD and fractionated RT increased urinary frequency and reduced the volume of individual voids at >14 weeks, but not at 4 weeks, compared with nonirradiated animals. Urothelial layer width was positively correlated with mean volume of individual voids (P = .0428) and negatively correlated with number of voids (P = .028), relating urothelial thinning to changes in RT-mediated bladder dysfunction. These chronic RT-induced changes in micturition patterns were prevented by captopril treatment. Focal bladder irradiation significantly increased the mean particle count of urine extracellular vesicles and the monocyte and neutrophil chemokines CCL2 and MIP-2, and the proportions of circulating inflammatory-mediated neutrophils and monocytes, which was also prevented by captopril. Exploratory transcriptomic analysis of bladder tissue implicated inflammatory and erythropoietic pathways. CONCLUSIONS This study demonstrated that systemic modulation of the RAS protected against and alleviated RT-induced late bladder injury but larger confirmatory studies are needed.
Collapse
Affiliation(s)
- Angela M Groves
- Departments of Radiation Oncology, University of Rochester, Rochester, New York
| | - Nicole Paris
- Departments of Radiation Oncology, University of Rochester, Rochester, New York
| | - Eric Hernady
- Departments of Radiation Oncology, University of Rochester, Rochester, New York
| | - Carl J Johnston
- Departments of Pediatrics, University of Rochester, Rochester, New York
| | - Omar Aljitawi
- Departments of Medicine, Hematology/Oncology, University of Rochester, Rochester, New York
| | - Yi-Fen Lee
- Departments of Urology, University of Rochester, Rochester, New York
| | - Sarah L Kerns
- Departments of Radiation Oncology, University of Rochester, Rochester, New York
| | - Brian Marples
- Departments of Radiation Oncology, University of Rochester, Rochester, New York.
| |
Collapse
|
9
|
Chan L, Chung CC, Yu RC, Hong CT. Cytokine profiles of plasma extracellular vesicles as progression biomarkers in Parkinson's disease. Aging (Albany NY) 2023; 15:1603-1614. [PMID: 36897204 PMCID: PMC10042681 DOI: 10.18632/aging.204575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/01/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND Inflammation contributes substantially to the pathogenesis of Parkinson's disease (PD). Plasma extracellular vesicle (EV)-derived cytokines are emerging biomarkers of inflammation. We conducted a longitudinal study of the plasma EV-derived cytokine profiles of people with PD (PwP). METHODS A total of 101 people with mild to moderate PD and 45 healthy controls (HCs) were recruited, and they completed motor assessments (Unified Parkinson Disease Rating Scale [UPDRS]) and cognitive tests at baseline and 1-year follow-up. We isolated the participants' plasma EVs and analyzed their levels of cytokines, including interleukin (IL)-1β, IL-6, IL-10, tumor necrosis factor (TNF)-α, and transforming growth factor (TGF)-β. RESULTS We noted no significant changes in the plasma EV-derived cytokine profiles of the PwPs and HCs between baseline and the 1-year follow-up. Among the PwP, changes in plasma EV-derived IL-1β, TNF-α and IL-6 levels were significantly associated with changes in the severity of postural instability and gait disturbance (PIGD) and cognition. Baseline plasma EV-derived IL-1β, TNF-α, IL-6, and IL-10 levels were significantly associated with the severity of PIGD and cognitive symptoms at follow-up, and PwP with elevated IL-1β and IL-6 levels exhibited significant progression of PIGD over the study period. CONCLUSION These results suggested the role of inflammation in PD progression. In addition, baseline levels of plasma EV-derived proinflammatory cytokines can be used to predict the progression of PIGD, the most severe motor symptom of PD. Additional studies with longer follow-up periods are necessary, and plasma EV-derived cytokines may serve as effective biomarkers of PD progression.
Collapse
Affiliation(s)
- Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Chen-Chih Chung
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Ruan-Ching Yu
- Division of Psychiatry, University College London, London, UK
| | - Chien-Tai Hong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
10
|
Exosomes: A missing link between chronic systemic inflammation and Alzheimer's disease? Biomed Pharmacother 2023; 159:114161. [PMID: 36641928 DOI: 10.1016/j.biopha.2022.114161] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023] Open
Abstract
Exosomes are potent mediators of physiological and pathological processes. In Alzheimer's disease and inflammatory disorders, due to exosomes' distinctive ability to cross the blood-brain barrier, a bidirectional communication between the periphery and the central nervous system exists. Since exosomes can carry various biochemical molecules, this review investigates the role of exosomes as possible mediators between chronic systemic inflammatory diseases and Alzheimer's disease. Exosomes carry pro-inflammatory molecules generated in the periphery, travel to the central nervous system, and target glial and neuronal cells. Microglia and astrocytes then become activated, initiating chronic neuroinflammation. As the aging brain is more susceptible to such changes, this state of neuroinflammation can stimulate neuropathologies, impair amyloid-beta clearance capabilities, and generate dysregulated microRNAs that alter the expression of genes critical in Alzheimer's disease pathology. These processes, individually and collectively, become significant risk factors for the development of Alzheimer's disease.
Collapse
|
11
|
Zamboni S, D'Ambrosio A, Margutti P. Extracellular vesicles as contributors in the pathogenesis of multiple sclerosis. Mult Scler Relat Disord 2023; 71:104554. [PMID: 36842311 DOI: 10.1016/j.msard.2023.104554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/22/2023]
Abstract
Extracellular vesicles (EVs) are a heterogeneous family of extracellular structures bounded by a phospholipid bilayer, released by all cell types in various biological fluids, such as blood and cerebrospinal fluid (CSF), playing important roles in intercellular communication, both locally and systemically. EVs carry and deliver a variety of bioactive molecules (proteins, nucleic acids, lipids and metabolites), conferring epigenetic and phenotypic changes to the recipient cells and thus resulting as important mediators of both homeostasis and pathogenesis. In neurological diseases, such as multiple sclerosis (MS), the EV ability to cross Blood-Brain Barrier (BBB), moving from central nervous system (CNS) to the peripheral circulation and vice versa, has increased the interest in EV study in the neurological field. In the present review, we will provide an overview of the recent advances made in understanding the pathogenic role of EVs regarding the immune response, the BBB dysfunction and the CNS inflammatory processes.
Collapse
Affiliation(s)
- Silvia Zamboni
- Department of Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | | | - Paola Margutti
- Department of Neurosciences, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
12
|
Yuan Y, Ma Y, Aili Z, Nijiati M. Reductions in extracellular vesicle-associated microRNA-126 levels in coronary blood after acute myocardial infarction: A retrospective study. Front Cardiovasc Med 2022; 9:1046839. [DOI: 10.3389/fcvm.2022.1046839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/11/2022] [Indexed: 11/30/2022] Open
Abstract
BackgroundAcute Myocardial Infarction (AMI) is a kind of cardiovascular disease with high mortality and incidence. Extracellular vesicles (EVs) and microRNA-126 (miR-126) are known to play important role in the development and prognosis of several cardiovascular diseases. Therefore, this study aimed to investigate the changes in Extracellular vesicle (EV)-associated miR-126 levels in the coronary blood of patients with AMI to explore the relationship between miR-126 levels and AMI.Materials and methodsWe analyzed EV-associated miR-126 in the coronary blood of patients with AMI and stable coronary artery disease (SCAD) using quantitative reverse transcription polymerase chain reaction (qRT-PCR).ResultsWe tested the coronary blood of 20 patients with AMI and 20 with SCAD. The mean age of the patients was 58.8 ± 10.3 years and 32 (80%) were men. We observed that the EV-associated miR-126 levels were lower in patients with AMI [median = 0.13; interquartile range (IQR): 0.08–0.22] than in patients with SCAD (median = 0.37; IQR: 0.26–0.48) (P < 0.001). In addition, the levels of miR-126 were negatively associated with the Thrombolysis in Myocardial Infarction (TIMI) score (r = −0.66, P = 0.001).ConclusionReduction of EV-associated miR-126 levels in the coronary blood of patients with AMI may be involved in acute coronary thrombosis events.
Collapse
|
13
|
Catitti G, De Bellis D, Vespa S, Simeone P, Canonico B, Lanuti P. Extracellular Vesicles as Players in the Anti-Inflammatory Inter-Cellular Crosstalk Induced by Exercise Training. Int J Mol Sci 2022; 23:14098. [PMID: 36430575 PMCID: PMC9697937 DOI: 10.3390/ijms232214098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 07/28/2023] Open
Abstract
Extracellular Vesicles (EVs) are circulating particles surrounded by a plasma membrane carrying a cargo consisting of proteins, lipids, RNAs, and DNA fragments, stemming from the cells from which they originated. EV factors (i.e., miRNAs) play relevant roles in intercellular crosstalk, both locally and systemically. As EVs increasingly gained attention as potential carriers for targeted genes, the study of EV effects on the host immune response became more relevant. It has been demonstrated that EVs regulate the host immune response, executing both pro- and anti-inflammatory functions. It is also known that physical exercise triggers anti-inflammatory effects. This review underlines the role of circulating EVs as players in the anti-inflammatory events associated with the regulation of the host's immune response to physical exercise.
Collapse
Affiliation(s)
- Giulia Catitti
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.C.); (D.D.B.); (S.V.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Domenico De Bellis
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.C.); (D.D.B.); (S.V.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Simone Vespa
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.C.); (D.D.B.); (S.V.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.C.); (D.D.B.); (S.V.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy;
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.C.); (D.D.B.); (S.V.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
14
|
Elevated G-CSF, IL8, and HGF in patients with definite Meniere's disease may indicate the role of NET formation in triggering autoimmunity and autoinflammation. Sci Rep 2022; 12:16309. [PMID: 36175465 PMCID: PMC9522806 DOI: 10.1038/s41598-022-20774-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/19/2022] [Indexed: 11/08/2022] Open
Abstract
The etiology and mechanism causing Meniere’s disease (MD) are not understood. The present study investigated the possible molecular mechanism of autoimmunity and autoinflammation associated with MD. Thirty-eight patients with definite MD and 39 normal volunteers were recruited, and 48 human cytokines/chemokines were quantified. In patients with MD pure tone audiograms, tympanograms and standard blood tests were performed. The mean hearing loss in the worse ear was 44.1 dB nHL. Compared to the referents, the concentrations of TNFα, IL1α, IL8, CTACK, MIP1α, MIP1β, G-CSF, and HGF in the sera of patients with MD were significantly elevated, while those of TRAIL and PDGFBB were significantly decreased. The area under the receiver operating characteristic curve (AUC) showed that G-CSF, MIP1α, and IL8 were above 0.8 and could be used to diagnose MD (p < 0.01), and the AUCs of CTACK and HGF were above 0.7 and acceptable to discriminate the MD group from the control group (p < 0.01). The revised AUCs (1 − AUC) of TRAIL and PDGFBB were above 0.7 and could also be used in the diagnosis of MD (p < 0.01). The linear regression showed significant correlations between MIP1α and GCSF, between IL2Rα and GCSF, between IL8 and HGF, between MIP1α and IL8, and between SCF and CTACK; there was a marginal linear association between IP10 and MIP1α. Linear regression also showed that there were significant age-related correlations of CTACK and MIG expression in the MD group (p < 0.01, ANOVA) but not in the control group. We hypothesize that G-CSF, IL8, and HGF, which are involved in the development of neutrophil extracellular traps (NETs) and through various mechanisms influence the functions of macrophages, lymphocytes, and dendritic cells, among others, are key players in the development of EH and MD and could be useful in elucidating the pathophysiological mechanisms leading to MD. Biomarkers identified in the present study may suggest that both autoimmune and autoinflammatory mechanisms are involved in MD. In the future, it will be valuable to develop a cost-effective method to detect G-CSF, IL8, HGF, CTACK, MIP1α, TRAIL, and PDGFBB in the serum of patient that have diagnostic relevance.
Collapse
|
15
|
Beckner ME, Conkright WR, Mi Q, Martin BJ, Sahu A, Flanagan SD, Ledford AK, Wright M, Susmarski A, Ambrosio F, Nindl BC. Neuroendocrine, Inflammatory, and Extracellular Vesicle Responses During the Navy Special Warfare Screener Selection Course. Physiol Genomics 2022; 54:283-295. [PMID: 35695270 PMCID: PMC9291410 DOI: 10.1152/physiolgenomics.00184.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Military operational stress is known to increase adrenal hormones and inflammatory cytokines, while decreasing hormones associated with the anabolic milieu and neuroendocrine system. Less is known about the role of extracellular vesicles (EVs), a form of cell-to-cell communication, in military operational stress and their relationship to circulating hormones. PURPOSE To characterize the neuroendocrine, cytokine, and EV response to an intense, 24-h selection course known as the Naval Special Warfare (NSW) Screener and identify associations between EVs and cytokines. METHODS Blood samples were collected the morning of and following the NSW Screener in 29 men (18 - 26 years). Samples were analyzed for concentrations of cortisol, insulin-like growth factor I (IGF-I), neuropeptide-Y (NPY), brain-derived neurotrophic factor (BDNF), α-klotho, tumor necrosis factor- α (TNFα), and interleukins (IL) -1β, -6, and -10. EVs stained with markers associated with exosomes (CD63), microvesicles (VAMP3), and apoptotic bodies (THSD1) were characterized using imaging flow cytometry and vesicle flow cytometry. RESULTS The selection event induced significant changes in circulating BDNF (-43.2%), IGF-I (-24.56%), TNFα (+17.7%), IL-6 (+13.6%), accompanied by increases in intensities of THSD1+ and VAMP3+ EVs (all p<0.05). Higher concentrations of IL-1β and IL-10 were positively associated with THSD1+ EVs (p<0.05). CONCLUSION Military operational stress altered the EV profile. Surface markers associated with apoptotic bodies were positively correlated with an inflammatory response. Future studies should consider a multi-omics assessment of EV cargo to discern canonical pathways that may be mediated by EVs during military stress.
Collapse
Affiliation(s)
- Meaghan E Beckner
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, United States
| | - William R Conkright
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, United States
| | - Qi Mi
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brian J Martin
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, United States
| | - Amrita Sahu
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shawn D Flanagan
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, United States
| | - Andrew K Ledford
- Department of Leadership, Ethics, and Law, U.S. Naval Academy, Annapolis, MD, United States
| | - Martin Wright
- Human Performance Lab, Physical Education Department, U.S. Naval Academy, Annapolis, MD, United States
| | - Adam Susmarski
- Brigade Orthopedics and Sports Medicine, U.S. Navy Academy, Annapolis, MD, United States
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bradley C Nindl
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
16
|
Reinicke M, Shamkeeva S, Hell M, Isermann B, Ceglarek U, Heinemann ML. Targeted Lipidomics for Characterization of PUFAs and Eicosanoids in Extracellular Vesicles. Nutrients 2022; 14:nu14071319. [PMID: 35405932 PMCID: PMC9000901 DOI: 10.3390/nu14071319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/16/2022] [Accepted: 03/19/2022] [Indexed: 02/06/2023] Open
Abstract
Lipids are increasingly recognized as bioactive mediators of extracellular vesicle (EV) functions. However, while EV proteins and nucleic acids are well described, EV lipids are insufficiently understood due to lack of adequate quantitative methods. We adapted an established targeted and quantitative mass spectrometry (LC-MS/MS) method originally developed for analysis of 94 eicosanoids and seven polyunsaturated fatty acids (PUFA) in human plasma. Additionally, the influence of freeze–thaw (FT) cycles, injection volume, and extraction solvent were investigated. The modified protocol was applied to lipidomic analysis of differently polarized macrophage-derived EVs. We successfully quantified three PUFAs and eight eicosanoids within EVs. Lipid extraction showed reproducible PUFA and eicosanoid patterns. We found a particularly high impact of FT cycles on EV lipid profiles, with significant reductions of up to 70%. Thus, repeated FT will markedly influence analytical results and may alter EV functions, emphasizing the importance of a standardized sample pretreatment protocol for the analysis of bioactive lipids in EVs. EV lipid profiles differed largely depending on the polarization of the originating macrophages. Particularly, we observed major changes in the arachidonic acid pathway. We emphasize the importance of a standardized sample pretreatment protocol for the analysis of bioactive lipids in EVs.
Collapse
|
17
|
The miRNome of Depression. Int J Mol Sci 2021; 22:ijms222111312. [PMID: 34768740 PMCID: PMC8582693 DOI: 10.3390/ijms222111312] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/10/2021] [Accepted: 10/18/2021] [Indexed: 02/07/2023] Open
Abstract
Depression is an effect of complex interactions between genetic, epigenetic and environmental factors. It is well established that stress responses are associated with multiple modest and often dynamic molecular changes in the homeostatic balance, rather than with a single genetic factor that has a strong phenotypic penetration. As depression is a multifaceted phenotype, it is important to study biochemical pathways that can regulate the overall allostasis of the brain. One such biological system that has the potential to fine-tune a multitude of diverse molecular processes is RNA interference (RNAi). RNAi is an epigenetic process showing a very low level of evolutionary diversity, and relies on the posttranscriptional regulation of gene expression using, in the case of mammals, primarily short (17–23 nucleotides) noncoding RNA transcripts called microRNAs (miRNA). In this review, our objective was to examine, summarize and discuss recent advances in the field of biomedical and clinical research on the role of miRNA-mediated regulation of gene expression in the development of depression. We focused on studies investigating post-mortem brain tissue of individuals with depression, as well as research aiming to elucidate the biomarker potential of miRNAs in depression and antidepressant response.
Collapse
|
18
|
Ohayon L, Zhang X, Dutta P. The role of extracellular vesicles in regulating local and systemic inflammation in cardiovascular disease. Pharmacol Res 2021; 170:105692. [PMID: 34182130 DOI: 10.1016/j.phrs.2021.105692] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/25/2021] [Accepted: 05/21/2021] [Indexed: 10/21/2022]
Abstract
Extracellular vesicles are heterogeneous structures surrounded by cell membranes and carry complex contents including nucleotides, proteins, and lipids. These proteins include cytokines and chemokines that are important for exaggerating local and systemic inflammation in disease. Extracellular vesicles are mainly categorized as exosomes and micro-vesicles, which are directly shed from the endosomal system or originated from the cell membrane, respectively. By transporting several bioactive molecules to recipient cells and tissues, extracellular vesicles have favorable, neutral, or detrimental impacts on their targets, such as switching cell phenotype, modulating gene expression, and controlling biological pathways such as inflammatory cell recruitment, activation of myeloid cells and cell proliferation. Extracellular vesicles mediate these functions via both autocrine and paracrine signaling. In the cardiovascular system, extracellular vesicles can be secreted by multiple cell types like cardiomyocytes, smooth muscle cells, macrophages, monocytes, fibroblasts, and endothelial cells, and affect functions of cells or tissues in distant organs. These effects involve maintaining homeostasis, regulating inflammation, and triggering pathological process in cardiovascular disease. In this review, we mainly focus on the role of micro-vesicles and exosomes, two important subtypes of extracellular vesicles, in local and systemic inflammation in cardiovascular diseases such as myocardial infarction, atherosclerosis and heart failure. We summarize recent findings and knowledge on the effect of extracellular vesicles in controlling both humoral and cellular immunity, and the therapeutic approaches to harness this knowledge to control exacerbated inflammation in cardiovascular diseases.
Collapse
Affiliation(s)
- Lee Ohayon
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xinyi Zhang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
19
|
Chen Y, Yuan X, Li Y, Chen J, Wu S, Jiang A, Miao X, Shu Q. Circulating exosomal microRNA-18a-5p accentuates intestinal inflammation in Hirschsprung-associated enterocolitis by targeting RORA. Am J Transl Res 2021; 13:4182-4196. [PMID: 34150007 PMCID: PMC8205693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 03/15/2021] [Indexed: 06/12/2023]
Abstract
The relevance of stem cell-derived exosomes has been implicated in necrotizing enterocolitis, while the involvement of serum-derived exosomes from children with Hirschsprung-associated enterocolitis (HAEC) in pathogenesis of HAEC remains unclear. This study set to identify the roles of exosomal microRNA (miR)-18a-5p from sera of HAEC patients in human-derived colonic epithelial NCM460 cells and in mice with HAEC. Exosomes were isolated from the sera of healthy children (Healthy-exo), patients with Hirschsprung's disease (HSCR) (HSCR-exo) or HAEC (HAEC-exo). A microarray analysis of miRNAs was implemented to assess the enrichment of miRNAs in these exosomes. HAEC-exo was significantly enriched in miR-18a-5p. HAEC-exo led to the generation of a pro-inflammatory microenvironment, inhibition of cellular DNA synthesis, and promotion of apoptosis in NCM460 cells. Mechanistically, miR-18a-5p targeted and repressed retinoid-related orphan receptor α (RORA) expression, thereby regulating the Sirtuin 1 (SIRT1)/nuclear factor-kappa B (NFκB) pathway. Overexpression of RORA ameliorated inflammatory damage in NCM460 cells caused by exosomal miR-18a-5p. HAEC-exo exacerbated inflammatory damage in HAEC mice, and this facilitation was reversed after RORA overexpression. Collectively, exosomal miR-18a-5p was a promoter of HAEC, which induces the intestine cell apoptosis and inflammatory responses through the inhibition of SIRT1/NFκB pathway by targeting RORA.
Collapse
Affiliation(s)
- Yi Chen
- Department of Nosocomial Infection, Yiwu Maternity and Children HospitalJinhua 322000, Zhejiang, P. R. China
| | - Xiaojian Yuan
- Department of Pediatrics, Zhejiang University School of MedicineHangzhou 310011, Zhejiang, P. R. China
| | - Yonglin Li
- Department of Pediatric Surgery, Yiwu Maternity and Child Health HospitalJinhua 322000, Zhejiang, P. R. China
| | - Jie Chen
- Department of Pediatric Surgery, Jiaxing Maternal and Child Health HospitalJiaxing 314051, Zhejiang, P. R. China
| | - Shannan Wu
- Department of Pediatric Surgery, Yiwu Maternity and Child Health HospitalJinhua 322000, Zhejiang, P. R. China
| | - Amin Jiang
- Department of Pediatric Surgery, Yiwu Maternity and Child Health HospitalJinhua 322000, Zhejiang, P. R. China
| | - Xuefeng Miao
- Department of Pediatric Surgery, Yiwu Maternity and Child Health HospitalJinhua 322000, Zhejiang, P. R. China
| | - Qiang Shu
- Department of Cardio-Thoracic Surgery, Children’s Hospital Affiliated to Zhejiang UniversityHangzhou 310000, Zhejiang, P. R. China
| |
Collapse
|
20
|
Chan L, Chung CC, Chen JH, Yu RC, Hong CT. Cytokine Profile in Plasma Extracellular Vesicles of Parkinson's Disease and the Association with Cognitive Function. Cells 2021; 10:cells10030604. [PMID: 33803292 PMCID: PMC7999703 DOI: 10.3390/cells10030604] [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: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/06/2021] [Indexed: 12/22/2022] Open
Abstract
Plasma extracellular vesicles (EVs) containing various molecules, including cytokines, can reflect the intracellular condition and participate in cell-to-cell signaling, thus emerging as biomarkers for Parkinson’s disease (PD). Inflammation may be a crucial risk factor for PD development and progression. The present study investigated the role of plasma EV cytokines as the biomarkers of PD. This cross-sectional study recruited 113 patients with PD, with mild to moderate stage disease, and 48 controls. Plasma EVs were isolated, and the levels of cytokines, including pro-interleukin (IL)-1β, IL-6, IL-10, tumor necrosis factor (TNF)-α, and transforming growth factor (TGF)-β1, were evaluated. Patients with PD had significantly increased plasma EV pro-IL-1β and TNF-α levels compared with controls after adjustment for age and sex. Despite the lack of a significant association between plasma EV cytokines and motor symptom severity in patients with PD, cognitive dysfunction severity, assessed using the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment, was significantly associated with plasma EV pro-IL-1β, IL-6, IL-10, and TNF-α levels. This association was PD specific and not found in controls. Furthermore, patients with PD cognitive deficit (MMSE < 26) exhibited a distinguished EV cytokine profile compared to those without cognitive deficit. The findings support the concept of inflammatory pathogenesis in the development and progression of PD and indicate that plasma EV cytokines may serve as PD biomarkers in future.
Collapse
Affiliation(s)
- Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (L.C.); (C.-C.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chen-Chih Chung
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (L.C.); (C.-C.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei 11031, Taiwan
| | - Jia-Hung Chen
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (L.C.); (C.-C.C.); (J.-H.C.)
| | - Ruan-Ching Yu
- Division of Psychiatry, University College London, London W1T 7NF, UK;
| | - Chien-Tai Hong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (L.C.); (C.-C.C.); (J.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: ; Tel.: +886-2-2249-0088 (ext. 8112)
| |
Collapse
|