1
|
Balic N, Nikolac Perkovic M, Milos T, Vuic B, Kurtovic Kodzoman M, Svob Strac D, Nedic Erjavec G. Extracellular vesicles as a promising tool in neuropsychiatric pharmacotherapy application and monitoring. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111393. [PMID: 40340017 DOI: 10.1016/j.pnpbp.2025.111393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
This review deals with the application of extracellular vesicles (EVs) in the treatment of various neuropsychiatric disorders, including mood disorders, neurodegeneration, psychosis, neurological insults and injuries, epilepsy and substance use disorders. The main challenges of most neuropsychiatric pharmaceuticals nowadays are how to reach the central nervous system at therapeutic concentration and maintain it long enough and how to avoid undesirable side effects caused by unsatisfying toxicity. Extracellular vesicles, as very important mediators of intercellular communication, can have a variety of therapeutic qualities. They can act neuroprotective, regenerative and anti-inflammatory, but they also have characteristics of a good drug delivery system, including their nano- scale size, biological safety and abilities to cross BBB, to pack drugs within the lipid bilayer, and not to trigger an immunological response. Besides, due to their presence in readily accessible biofluids, they are good candidates for biomarkers of the disease, its progression and therapy response monitoring. Alternations in EVs' cargo profiles can reflect the pathogenesis of neuropsychiatric disorders, but they could also affect the disease outcomes. In the future, EVs could help physicians to tailor treatment strategies for individual patients, however, more extensive studies are needed to standardize isolation, purification and production procedures, increase efficacy of drug loading and limit unwanted effects of innate EVs' content.
Collapse
Affiliation(s)
- Nikola Balic
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | | | - Tina Milos
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | - Barbara Vuic
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | | | | | | |
Collapse
|
2
|
Sánchez SV, Otavalo GN, Gazeau F, Silva AKA, Morales JO. Intranasal delivery of extracellular vesicles: A promising new approach for treating neurological and respiratory disorders. J Control Release 2025; 379:489-523. [PMID: 39800240 DOI: 10.1016/j.jconrel.2025.01.018] [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: 09/13/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Extracellular vesicles (EVs) are membrane vesicles secreted by all types of cells, including bacteria, animals, and plants. These vesicles contain proteins, nucleic acids, and lipids from their parent cells and can transfer these components between cells. EVs have attracted attention for their potential use in diagnosis and therapy due to their natural properties, such as low immunogenicity, high biocompatibility, and ability to cross the blood-brain barrier. They can also be engineered to carry therapeutic molecules. EVs can be delivered via various routes. The intranasal route is particularly advantageous for delivering them to the central nervous system, making it a promising approach for treating neurological disorders. SCOPE OF REVIEW This review delves into the promising potential of intranasally administered EVs-based therapies for various medical conditions, with a particular focus on those affecting the brain and central nervous system. Additionally, the potential use of these therapies for pulmonary conditions, cancer, and allergies is examined, offering a hopeful outlook for the future of medical treatments. MAJOR CONCLUSIONS The intranasal administration of EVs offers significant advantages over other delivery methods. By directly delivering EVs to the brain, specifically targeting areas that have been injured, this administration proves to be highly efficient and effective, providing reassurance about the progress in medical treatments. Intranasal delivery is not limited to brain-related conditions. It can also benefit other organs like the lungs and stimulate a mucosal immune response against various pathogens due to the highly vascularized nature of the nasal cavity and airways. Moreover, it has the added benefit of minimizing toxicity to non-targeted organs and allows the EVs to remain longer in the body. As a result, there is a growing emphasis on conducting clinical trials for intranasal administration of EVs, particularly in treating respiratory tract pathologies such as coronavirus disease.
Collapse
Affiliation(s)
- Sofía V Sánchez
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile
| | - Gabriela N Otavalo
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile
| | - Florence Gazeau
- Université Paris Cité, CNRS UMR8175, INSERM U1334, Laboratory NABI (Nanomédecine, Biologie Extracellulaire, Intégratome et Innovations en santé), Paris, France
| | - Amanda K A Silva
- Université Paris Cité, CNRS UMR8175, INSERM U1334, Laboratory NABI (Nanomédecine, Biologie Extracellulaire, Intégratome et Innovations en santé), Paris, France
| | - Javier O Morales
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile.
| |
Collapse
|
3
|
Quintanilla ME, Santapau D, Diaz E, Valenzuela Martinez I, Medina N, Landskron G, Dominguez A, Morales P, Ramírez D, Hermoso M, Olivares B, Berríos-Cárcamo P, Ezquer M, Herrera-Marschitz M, Israel Y, Ezquer F. Intragastric administration of short chain fatty acids greatly reduces voluntary ethanol intake in rats. Sci Rep 2024; 14:29260. [PMID: 39587197 PMCID: PMC11589138 DOI: 10.1038/s41598-024-80228-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
Alcohol use disorder (AUD) represents a public health crisis with few FDA-approved medications for its treatment. Growing evidence supports the key role of the bidirectional communication between the gut microbiota and the central nervous system (CNS) during the initiation and progression of alcohol use disorder. Among the different protective molecules that could mediate this communication, short chain fatty acids (SCFAs) have emerged as attractive candidates, since these gut microbiota-derived molecules have multi-target effects that could normalize several of the functional and structural parameters altered by chronic alcohol abuse. The present study, conducted in male alcohol-preferring UChB rats, shows that the initiation of voluntary ethanol intake was inhibited in 85% by the intragastric administration of a combination of SCFAs (acetate, propionate and butyrate) given before ethanol exposure, while SCFAs administration after two months of ethanol intake induced a 90% reduction in its consumption. These SCFAs therapeutic effects were associated with (1) a significant reduction of ethanol-induced intestinal inflammation and damage; (2) reduction of plasma lipopolysaccharide levels and hepatic inflammation; (3) reduction of ethanol-induced astrocyte and microglia activation; and (4) attenuation of the ethanol-induced gene expression changes within the nucleus accumbens. Finally, we determined that among the different SCFAs evaluated, butyrate was the most potent, reducing chronic ethanol intake in a dose-response manner. These findings support a key role of SCFAs, and especially butyrate, in regulating AUD, providing a simple, inexpensive, and safe approach as a preventive and intervention-based strategy to address this devastating disease.
Collapse
Affiliation(s)
- María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Specialized Center for the Prevention of Substance Use and the Treatment of Addictions (CESA), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Daniela Santapau
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Eugenio Diaz
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | | | - Nicolas Medina
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Glauben Landskron
- Center for Biomedical Research, CIBMED, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Antonia Dominguez
- Center for Biomedical Research, CIBMED, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Specialized Center for the Prevention of Substance Use and the Treatment of Addictions (CESA), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Research Center for the Development of Novel Therapeutics Alternatives for Alcohol Use Disorders, Santiago, Chile
| | - David Ramírez
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Research Center for the Development of Novel Therapeutics Alternatives for Alcohol Use Disorders, Santiago, Chile
| | - Marcela Hermoso
- Department of Gastroenterology and Hepatology, University Medical Center, Groningen, The Netherlands
- Laboratorio de Inmunidad Innata, Programa Disciplinario de Inmunología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Belén Olivares
- Center for Medical Chemistry, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Pablo Berríos-Cárcamo
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Marcelo Ezquer
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | | | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Specialized Center for the Prevention of Substance Use and the Treatment of Addictions (CESA), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile.
- Research Center for the Development of Novel Therapeutics Alternatives for Alcohol Use Disorders, Santiago, Chile.
| |
Collapse
|
4
|
Severtsev VV, Pavkina MA, Ivanets NN, Vinnikova MA, Yakovlev AA. Extracellular Vesicles as Potential Biomarkers in Addictive Disorders. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1970-1984. [PMID: 39647826 DOI: 10.1134/s0006297924110117] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 12/10/2024]
Abstract
Small extracellular vesicles (sEVs) and their role in mental and addictive disorders are extremely promising research areas. Because of their small size, sEVs can pass through the blood-brain barrier. The membrane of sEVs contain proteins that protect them against destruction by the organism's immune system. Due to these properties, sEVs circulating in the blood can be used as potential biomarkers of processes occurring in the brain. Exposure to psychoactive substances in vitro and in vivo affects sEV biogenesis and significantly alters the amount of sEVs and chemical composition of their cargo. Based on the published reports, sEVs carry numerous potential biomarkers of addictive pathologies, although the diagnostic significance of these markers still has to be evaluated. A large body of evidence indicates that psychoactive substances influence Rab family GTPases, Toll-like receptors, complement system components, and cytokines. In some studies, the effect of psychoactive substances on sEVs was found to be sex-dependent. It has become commonly accepted that sEVs are involved in the regulation of neuroinflammation and interaction between glial cells and neurons, as well as between peripheral cells and cells of the central nervous system. Here, we formulated a hypothesis on the existence of two mechanisms/stages involved in the effect of psychoactive substances on sEVs: the "fast" mechanism that provides neuroplasticity, and the "slow" one, resulting from the impaired biogenesis of sEVs and formation of aberrant vesicles.
Collapse
Affiliation(s)
- Vsevolod V Severtsev
- Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, 119048, Russia.
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical-Biological Agency of the Russian Federation, Moscow, 143007, Russia
| | - Margarita A Pavkina
- Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, 119048, Russia
| | - Nikolay N Ivanets
- Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, 119048, Russia
| | - Maria A Vinnikova
- Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, 119048, Russia
- Moscow Scientific and Practical Center of Narcology, Moscow Healthcare Department, Moscow, 109390, Russia
| | - Alexander A Yakovlev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
- Research and Clinical Center for Neuropsychiatry, Moscow Healthcare Department, Moscow, 115419, Russia
| |
Collapse
|
5
|
Meinhardt MW, Gerlach B, Spanagel R. Good Practice Guideline for Preclinical Alcohol Research: The STRINGENCY Framework. Curr Top Behav Neurosci 2024. [PMID: 39117860 DOI: 10.1007/7854_2024_484] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Research in the field of preclinical alcohol research, but also science in general, has a problem: Many published scientific results cannot be repeated. As a result, findings from preclinical research often do not translate well to humans, causing increasing disappointment and calls for restructuring of preclinical research, that is, better reproducibility of preclinical research. However, the replication crisis is an inherent problem in biomedical research. Replication failures are not only due to small experimental variations but are often the result of poor methodology. In response to the replication crisis, numerous guidelines and recommendations have been proposed to promote transparency, rigor, and reproducibility in scientific research. What is missing today is a framework that integrates all the confusing information that results from all these guidelines and recommendations. Here we present STRINGENCY, an integrative approach to good practice guidelines for preclinical alcohol research, which can also apply to behavioral research in general and which aims to improve preclinical research to better prepare it for translation and minimize the "valley of death" in translational research. STRINGENCY includes systematic review and, when possible, meta-analysis prior to study design, sample size calculation, preregistration, multisite experiments, scientific data management (FAIR), reporting of data using ARRIVE, generalization of research data, and transparent publications that allow reporting of null results. We invite the scientific community to adopt STRINGENCY to improve the reliability and impact of preclinical alcohol research.
Collapse
Affiliation(s)
- Marcus W Meinhardt
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
- Department of Molecular Neuroimaging, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany.
| | - Björn Gerlach
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Guarantors of EQIPD e.V., Heidelberg, Germany
- PAASP GmbH, Heidelberg, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
- German Center for Mental Health (DZPG), Mannheim, Heidelberg, Ulm, Germany.
| |
Collapse
|
6
|
Gallardo J, Berríos-Cárcamo P, Ezquer F. Mesenchymal stem cells as a promising therapy for alcohol use disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:179-211. [PMID: 39523054 DOI: 10.1016/bs.irn.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Alcohol Use Disorder (AUD) is a highly prevalent medical condition characterized by impaired control over alcohol consumption, despite negative consequences on the individual's daily life and health. There is increasing evidence suggesting that chronic alcohol intake, like other addictive drugs, induces neuroinflammation and oxidative stress, disrupting glutamate homeostasis in the main brain areas related to drug addiction. This review explores the potential application of mesenchymal stem cells (MSCs)-based therapy for the treatment of AUD. MSCs secrete a broad array of anti-inflammatory and antioxidant molecules, thus, the administration of MSCs, or their secretome, could reduce neuroinflammation and oxidative stress in the brain. These effects correlate with an increase in the expression of the main glutamate transporter, GLT1, which, through the normalization of the extracellular glutamate levels, could mediate the inhibitory effect of MSCs' secretome on chronic alcohol consumption, thus highlighting GLT1 as a central target to reduce chronic alcohol consumption.
Collapse
Affiliation(s)
- Javiera Gallardo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Pablo Berríos-Cárcamo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile; Research Center for the Development of Novel Therapeutics Alternatives for Alcohol Use Disorders, Santiago, Chile.
| |
Collapse
|
7
|
Abdal Dayem A, Yan E, Do M, Kim Y, Lee Y, Cho SG, Kim DH. Engineering extracellular vesicles for ROS scavenging and tissue regeneration. NANO CONVERGENCE 2024; 11:24. [PMID: 38922501 PMCID: PMC11208369 DOI: 10.1186/s40580-024-00430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024]
Abstract
Stem cell therapy holds promise for tissue regeneration, yet significant challenges persist. Emerging as a safer and potentially more effective alternative, extracellular vesicles (EVs) derived from stem cells exhibit remarkable abilities to activate critical signaling cascades, thereby facilitating tissue repair. EVs, nano-scale membrane vesicles, mediate intercellular communication by encapsulating a diverse cargo of proteins, lipids, and nucleic acids. Their therapeutic potential lies in delivering cargos, activating signaling pathways, and efficiently mitigating oxidative stress-an essential aspect of overcoming limitations in stem cell-based tissue repair. This review focuses on engineering and applying EVs in tissue regeneration, emphasizing their role in regulating reactive oxygen species (ROS) pathways. Additionally, we explore strategies to enhance EV therapeutic activity, including functionalization and incorporation of antioxidant defense proteins. Understanding these molecular mechanisms is crucial for optimizing EV-based regenerative therapies. Insights into EV and ROS signaling modulation pave the way for targeted and efficient regenerative therapies harnessing the potential of EVs.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ellie Yan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yoojung Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yeongseo Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin- gu, Seoul, 05029, Republic of Korea.
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, 21205, USA.
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
8
|
Yadav K, Vijayalakshmi R, Kumar Sahu K, Sure P, Chahal K, Yadav R, Sucheta, Dubey A, Jha M, Pradhan M. Exosome-Based Macromolecular neurotherapeutic drug delivery approaches in overcoming the Blood-Brain barrier for treating brain disorders. Eur J Pharm Biopharm 2024; 199:114298. [PMID: 38642716 DOI: 10.1016/j.ejpb.2024.114298] [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: 02/03/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Delivering drugs to the brain is a complex challenge in medical research, particularly for disorders like Alzheimer's and Parkinson's. The blood-brain barrier restricts the entry of many therapeutic molecules, hindering their effectiveness. Nanoparticles, a potential solution, face issues like toxicity and limited approvals. A new avenue explores the use of small extracellular vesicles (sEVs), i.e., exosomes, as natural carriers for drug delivery. sEVs, tiny structures below 150 nm, show promise due to their minimal immune response and ability to precisely deliver drugs. This review focuses on the potential of sEVs-based drug delivery systems for treating neurological disorders, brain cancers, and other brain-related issues. Notably, bioengineered sEVs-carrying therapeutic compounds exhibit promise in early studies. The unique features of sEVs, such as their small size and natural properties, position them as candidates to overcome challenges in drug delivery to the brain. Ongoing clinical trials and research into sEVs behavior within the body further highlight their potential for revolutionizing drug delivery and addressing complex brain conditions.
Collapse
Affiliation(s)
- Krishna Yadav
- Raipur Institute of Pharmaceutical Education and Research, Sarona, Raipur, Chhattisgarh 492010, India
| | - R Vijayalakshmi
- Department of Pharmaceutical Analysis, GIET School of Pharmacy, Chaitanya Knowledge City, Rajahmundry, AP, 533296, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Pavani Sure
- Department of Pharmaceutics, Vignan Institute of Pharmaceutical Sciences, Hyderabad, Telangana, India
| | - Kavita Chahal
- Department of Botany, Government Model Science College Jabalpur, Madhya Pradesh, India
| | - Renu Yadav
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, Haryana, 122103, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, Haryana, 122103, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangaluru-575018, Karnataka, India
| | - Megha Jha
- Department of Life Science, Mansarovar Global University, Sehore, M.P., India
| | - Madhulika Pradhan
- Gracious College of Pharmacy, Abhanpur, Chhattisgarh, 493661, India.
| |
Collapse
|
9
|
Gotoh S, Kawabori M, Fujimura M. Intranasal administration of stem cell-derived exosomes for central nervous system diseases. Neural Regen Res 2024; 19:1249-1255. [PMID: 37905871 PMCID: PMC11467946 DOI: 10.4103/1673-5374.385875] [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: 06/30/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Exosomes, lipid bilayer-enclosed small cellular vesicles, are actively secreted by various cells and play crucial roles in intercellular communication. These nanosized vesicles transport internalized proteins, mRNA, miRNA, and other bioactive molecules. Recent findings have provided compelling evidence that exosomes derived from stem cells hold great promise as a therapeutic modality for central nervous system disorders. These exosomes exhibit multifaceted properties including anti-apoptotic, anti-inflammatory, neurogenic, and vasculogenic effects. Furthermore, exosomes offer several advantages over stem cell therapy, such as high preservation capacity, low immunogenicity, the ability to traverse the blood-brain barrier, and the potential for drug encapsulation. Consequently, researchers have turned their attention to exosomes as a novel therapeutic avenue. Nonetheless, akin to the limitations of stem cell treatment, the limited accumulation of exosomes in the injured brain poses a challenge to their clinical application. To overcome this hurdle, intranasal administration has emerged as a non-invasive and efficacious route for delivering drugs to the central nervous system. By exploiting the olfactory and trigeminal nerve axons, this approach enables the direct transport of therapeutics to the brain while bypassing the blood-brain barrier. Notably, exosomes, owing to their small size, can readily access the nerve pathways using this method. As a result, intranasal administration has gained increasing recognition as an optimal therapeutic strategy for exosome-based treatments. In this comprehensive review, we aim to provide an overview of both basic and clinical research studies investigating the intranasal administration of exosomes for the treatment of central nervous system diseases. Furthermore, we elucidate the underlying therapeutic mechanisms and offer insights into the prospect of this approach.
Collapse
Affiliation(s)
- Shuho Gotoh
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Masahito Kawabori
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Miki Fujimura
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| |
Collapse
|
10
|
Berríos-Cárcamo P, Núñez S, Castañeda J, Gallardo J, Bono MR, Ezquer F. Two-Month Voluntary Ethanol Consumption Promotes Mild Neuroinflammation in the Cerebellum but Not in the Prefrontal Cortex, Hippocampus, or Striatum of Mice. Int J Mol Sci 2024; 25:4173. [PMID: 38673763 PMCID: PMC11050159 DOI: 10.3390/ijms25084173] [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: 03/01/2024] [Revised: 03/29/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic ethanol exposure often triggers neuroinflammation in the brain's reward system, potentially promoting the drive for ethanol consumption. A main marker of neuroinflammation is the microglia-derived monocyte chemoattractant protein 1 (MCP1) in animal models of alcohol use disorder in which ethanol is forcefully given. However, there are conflicting findings on whether MCP1 is elevated when ethanol is taken voluntarily, which challenges its key role in promoting motivation for ethanol consumption. Here, we studied MCP1 mRNA levels in areas implicated in consumption motivation-specifically, the prefrontal cortex, hippocampus, and striatum-as well as in the cerebellum, a brain area highly sensitive to ethanol, of C57BL/6 mice subjected to intermittent and voluntary ethanol consumption for two months. We found a significant increase in MCP1 mRNA levels in the cerebellum of mice that consumed ethanol compared to controls, whereas no significant changes were observed in the prefrontal cortex, hippocampus, or striatum or in microglia isolated from the hippocampus and striatum. To further characterize cerebellar neuroinflammation, we measured the expression changes in other proinflammatory markers and chemokines, revealing a significant increase in the proinflammatory microRNA miR-155. Notably, other classical proinflammatory markers, such as TNFα, IL6, and IL-1β, remained unaltered, suggesting mild neuroinflammation. These results suggest that the onset of neuroinflammation in motivation-related areas is not required for high voluntary consumption in C57BL/6 mice. In addition, cerebellar susceptibility to neuroinflammation may be a trigger to the cerebellar degeneration that occurs after chronic ethanol consumption in humans.
Collapse
Affiliation(s)
- Pablo Berríos-Cárcamo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago 7610615, Chile; (J.G.); (F.E.)
| | - Sarah Núñez
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones 7510602, Chile;
- Centro Ciencia & Vida, Santiago 8580702, Chile
| | - Justine Castañeda
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile; (J.C.); (M.R.B.)
| | - Javiera Gallardo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago 7610615, Chile; (J.G.); (F.E.)
| | - María Rosa Bono
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile; (J.C.); (M.R.B.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago 7610615, Chile; (J.G.); (F.E.)
- Research Center for the Development of Novel Therapeutics Alternatives for Alcohol Use Disorders, Santiago 7610658, Chile
| |
Collapse
|
11
|
Sheykhhasan M, Heidari F, Farsani ME, Azimzadeh M, Kalhor N, Ababzadeh S, Seyedebrahimi R. Dual Role of Exosome in Neurodegenerative Diseases: A Review Study. Curr Stem Cell Res Ther 2024; 19:852-864. [PMID: 37496136 DOI: 10.2174/1574888x18666230726161035] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/07/2023] [Accepted: 06/19/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Extracellular vesicles (EVs) are one of the crucial means of intercellular communication, which takes many different forms. They are heterogeneous, secreted by a range of cell types, and can be generally classified into microvesicles and exosomes depending on their location and function. Exosomes are small EVs with diameters of about 30-150 nm and diverse cell sources. METHODS The MEDLINE/PubMed database was reviewed for papers written in English and publication dates of recent years, using the search string "Exosome" and "Neurodegenerative diseases." RESULTS The exosomes have attracted interest as a significant biomarker for a better understanding of disease development, gene silencing delivery, and alternatives to stem cell-based therapy because of their low-invasive therapeutic approach, repeatable distribution in the central nervous system (CNS), and high efficiency. Also, they are nanovesicles that carry various substances, which can have an impact on neural plasticity and cognitive functioning in both healthy and pathological circumstances. Therefore, exosomes are conceived as nanovesicles containing proteins, lipids, and nucleic acids. However, their composition varies considerably depending on the cells from which they are produced. CONCLUSION In the present review, we discuss several techniques for the isolation of exosomes from different cell sources. Furthermore, reviewing research on exosomes' possible functions as carriers of bioactive substances implicated in the etiology of neurodegenerative illnesses, we further examine them. We also analyze the preclinical and clinical research that shows exosomes to have therapeutic potential.
Collapse
Affiliation(s)
- Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| | - Fatemeh Heidari
- Department of Anatomy, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Mohsen Eslami Farsani
- Department of Anatomy, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Maryam Azimzadeh
- Department of Medical Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| | - Shima Ababzadeh
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Tissue Engineering, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Reihaneh Seyedebrahimi
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Anatomy, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
12
|
Mellado S, Cuesta CM, Montagud S, Rodríguez‐Arias M, Moreno‐Manzano V, Guerri C, Pascual M. Therapeutic role of mesenchymal stem cell-derived extracellular vesicles in neuroinflammation and cognitive dysfunctions induced by binge-like ethanol treatment in adolescent mice. CNS Neurosci Ther 2023; 29:4018-4031. [PMID: 37381698 PMCID: PMC10651955 DOI: 10.1111/cns.14326] [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: 01/04/2023] [Revised: 04/10/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are heterogeneous membrane vesicles secreted by cells in extracellular spaces that play an important role in intercellular communication under both normal and pathological conditions. Mesenchymal stem cells (MSC) are anti-inflammatory and immunoregulatory cells capable of secreting EVs, which are considered promising molecules for treating immune, inflammatory, and degenerative diseases. Our previous studies demonstrate that, by activating innate immune receptors TLR4 (Toll-like receptor 4), binge-like ethanol exposure in adolescence causes neuroinflammation and neural damage. AIMS To evaluate whether the intravenous administration of MSC-derived EVs is capable of reducing neuroinflammation, myelin and synaptic alterations, and the cognitive dysfunction induced by binge-like ethanol treatment in adolescent mice. MATERIALS & METHODS MSC-derived EVs obtained from adipose tissue were administered in the tail vein (50 microg/dose, one weekly dose) to female WT adolescent mice treated intermittently with ethanol (3.0 g/kg) during two weeks. RESULTS MSC-derived EVs from adipose tissue ameliorate ethanol-induced up-regulation of inflammatory genes (e.g., COX-2, iNOS, MIP-1α, NF-κB, CX3CL1, and MCP-1) in the prefrontal cortex of adolescent mice. Notably, MSC-derived EVs also restore the myelin and synaptic derangements, and the memory and learning impairments, induced by ethanol treatment. Using cortical astroglial cells in culture, our results further confirm that MSC-derived EVs decrease inflammatory genes in ethanol-treated astroglial cells. This, in turn, confirms in vivo findings. CONCLUSION Taken together, these results provide the first evidence for the therapeutic potential of the MSC-derived EVs in the neuroimmune response and cognitive dysfunction induced by binge alcohol drinking in adolescence.
Collapse
Affiliation(s)
- Susana Mellado
- Department of Physiology, School of Medicine and DentistryUniversity of ValenciaValenciaSpain
| | - Carlos M. Cuesta
- Department of Physiology, School of Medicine and DentistryUniversity of ValenciaValenciaSpain
| | - Sandra Montagud
- Department of Psychobiology, Facultad de PsicologíaUniversitat de ValenciaValenciaSpain
| | - Marta Rodríguez‐Arias
- Department of Psychobiology, Facultad de PsicologíaUniversitat de ValenciaValenciaSpain
| | | | | | - María Pascual
- Department of Physiology, School of Medicine and DentistryUniversity of ValenciaValenciaSpain
| |
Collapse
|
13
|
da Silva AV, Serrenho I, Araújo B, Carvalho AM, Baltazar G. Secretome as a Tool to Treat Neurological Conditions: Are We Ready? Int J Mol Sci 2023; 24:16544. [PMID: 38003733 PMCID: PMC10671352 DOI: 10.3390/ijms242216544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/04/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Due to their characteristics, mesenchymal stem cells (MSCs) are considered a potential therapy for brain tissue injury or degeneration. Nevertheless, despite the promising results observed, there has been a growing interest in the use of cell-free therapies in regenerative medicine, such as the use of stem cell secretome. This review provides an in-depth compilation of data regarding the secretome composition, protocols used for its preparation, as well as existing information on the impact of secretome administration on various brain conditions, pointing out gaps and highlighting relevant findings. Moreover, due to the ability of MSCs to respond differently depending on their microenvironment, preconditioning of MSCs has been used to modulate their composition and, consequently, their therapeutic potential. The different strategies used to modulate the MSC secretome were also reviewed. Although secretome administration was effective in improving functional impairments, regeneration, neuroprotection, and reducing inflammation in brain tissue, a high variability in secretome preparation and administration was identified, compromising the transposition of preclinical data to clinical studies. Indeed, there are no reports of the use of secretome in clinical trials. Despite the existing limitations and lack of clinical data, secretome administration is a potential tool for the treatment of various diseases that impact the CNS.
Collapse
Affiliation(s)
- Andreia Valente da Silva
- Health Sciences Research Center (CICS-UBI), University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Inês Serrenho
- Health Sciences Research Center (CICS-UBI), University of Beira Interior, 6201-506 Covilhã, Portugal
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Beatriz Araújo
- Health Sciences Research Center (CICS-UBI), University of Beira Interior, 6201-506 Covilhã, Portugal
| | | | - Graça Baltazar
- Health Sciences Research Center (CICS-UBI), University of Beira Interior, 6201-506 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior, 6201-506 Covilhã, Portugal
| |
Collapse
|
14
|
Ibáñez C, Acuña T, Quintanilla ME, Pérez-Reytor D, Morales P, Karahanian E. Fenofibrate Decreases Ethanol-Induced Neuroinflammation and Oxidative Stress and Reduces Alcohol Relapse in Rats by a PPAR-α-Dependent Mechanism. Antioxidants (Basel) 2023; 12:1758. [PMID: 37760061 PMCID: PMC10525752 DOI: 10.3390/antiox12091758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
High ethanol consumption triggers neuroinflammation, implicated in sustaining chronic alcohol use. This inflammation boosts glutamate, prompting dopamine release in reward centers, driving prolonged drinking and relapse. Fibrate drugs, activating peroxisome proliferator-activated receptor alpha (PPAR-α), counteract neuroinflammation in other contexts, prompting investigation into their impact on ethanol-induced inflammation. Here, we studied, in UChB drinker rats, whether the administration of fenofibrate in the withdrawal stage after chronic ethanol consumption reduces voluntary intake when alcohol is offered again to the animals (relapse-type drinking). Furthermore, we determined if fenofibrate was able to decrease ethanol-induced neuroinflammation and oxidative stress in the brain. Animals treated with fenofibrate decreased alcohol consumption by 80% during post-abstinence relapse. Furthermore, fenofibrate decreased the expression of the proinflammatory cytokines tumor necrosis factor-alpha (TNF-α) and interleukins IL-1β and IL-6, and of an oxidative stress-induced gene (heme oxygenase-1), in the hippocampus, nucleus accumbens, and prefrontal cortex. Animals treated with fenofibrate showed an increase M2-type microglia (with anti-inflammatory proprieties) and a decrease in phagocytic microglia in the hippocampus. A PPAR-α antagonist (GW6471) abrogated the effects of fenofibrate, indicating that they are dependent on PPAR-α activation. These findings highlight the potential of fenofibrate, an FDA-approved dyslipidemia medication, as a supplementary approach to alleviating relapse severity in individuals with alcohol use disorder (AUD) during withdrawal.
Collapse
Affiliation(s)
- Cristina Ibáñez
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.I.); (D.P.-R.)
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 8910060, Chile;
| | - Tirso Acuña
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile;
| | - María Elena Quintanilla
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 8910060, Chile;
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile;
| | - Diliana Pérez-Reytor
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.I.); (D.P.-R.)
| | - Paola Morales
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 8910060, Chile;
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile;
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Eduardo Karahanian
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.I.); (D.P.-R.)
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 8910060, Chile;
| |
Collapse
|
15
|
Fernández-Rodríguez S, Cano-Cebrián MJ, Esposito-Zapero C, Pérez S, Guerri C, Zornoza T, Polache A. N-Acetylcysteine normalizes brain oxidative stress and neuroinflammation observed after protracted ethanol abstinence: a preclinical study in long-term ethanol-experienced male rats. Psychopharmacology (Berl) 2023; 240:725-738. [PMID: 36708386 PMCID: PMC10006045 DOI: 10.1007/s00213-023-06311-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/04/2023] [Indexed: 01/29/2023]
Abstract
RATIONALE Using a preclinical model based on the Alcohol Deprivation Effect (ADE), we have reported that N-Acetylcysteine (NAC) can prevent the relapse-like drinking behaviour in long-term ethanol-experienced male rats. OBJECTIVES To investigate if chronic ethanol intake and protracted abstinence affect several glutamate transporters and whether NAC, administered during the withdrawal period, could restore the ethanol-induced brain potential dysfunctions. Furthermore, the antioxidant and anti-inflammatory effects of NAC during abstinence in rats under the ADE paradigm were also explored. METHODS The expression of GLT1, GLAST and xCT in nucleus accumbens (Nacc) and dorsal striatum (DS) of male Wistar was analysed after water and chronic ethanol intake. We used the model based on the ADE within another cohort of male Wistar rats. During the fourth abstinence period, rats were treated for 9 days with vehicle or NAC (60, 100 mg/kg; s.c.). The effects of NAC treatment on (i) glutamate transporters expression in the Nacc and DS, (ii) the oxidative status in the hippocampus (Hip) and amygdala (AMG) and (iii) some neuroinflammatory markers in prefrontal cortex (PFC) were tested. RESULTS NAC chronic administration during protracted abstinence restored oxidative stress markers (GSSG and GGSH/GSH) in the Hip. Furthermore, NAC was able to normalize some neuroinflammation markers in PFC without normalizing the observed downregulation of GLT1 and GLAST in Nacc. CONCLUSIONS NAC restores brain oxidative stress and neuroinflammation that we previously observed after protracted ethanol abstinence in long-term ethanol-experienced male rats. This NAC effect could be a plausible mechanism for its anti-relapse effect. Also, brain oxidative stress and neuroinflammation could represent and identify plausible targets for searching new anti-relapse pharmacotherapies.
Collapse
Affiliation(s)
- Sandra Fernández-Rodríguez
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - María José Cano-Cebrián
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - Claudia Esposito-Zapero
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Valencia, Spain
| | - Teodoro Zornoza
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain
| | - Ana Polache
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Valencia, Spain.
| |
Collapse
|
16
|
Turano E, Scambi I, Virla F, Bonetti B, Mariotti R. Extracellular Vesicles from Mesenchymal Stem Cells: Towards Novel Therapeutic Strategies for Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24032917. [PMID: 36769247 PMCID: PMC9917806 DOI: 10.3390/ijms24032917] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Neurodegenerative diseases are fatal disorders of the central nervous system (CNS) which currently lack effective treatments. The application of mesenchymal stem cells (MSCs) represents a new promising approach for treating these incurable disorders. Growing evidence suggest that the therapeutic effects of MSCs are due to the secretion of neurotrophic molecules through extracellular vesicles. The extracellular vesicles produced by MSCs (MSC-EVs) have valuable innate properties deriving from parental cells and could be exploited as cell-free treatments for many neurological diseases. In particular, thanks to their small size, they are able to overcome biological barriers and reach lesion sites inside the CNS. They have a considerable pharmacokinetic and safety profile, avoiding the critical issues related to the fate of cells following transplantation. This review discusses the therapeutic potential of MSC-EVs in the treatment of neurodegenerative diseases, focusing on the strategies to further enhance their beneficial effects such as tracking methods, bioengineering applications, with particular attention to intranasal delivery as a feasible strategy to deliver MSC-EVs directly to the CNS in an effective and minimally invasive way. Current progresses and limiting issues to the extent of the use of MSC-EVs treatment for human neurodegenerative diseases will be also revised.
Collapse
Affiliation(s)
- Ermanna Turano
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Ilaria Scambi
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Federica Virla
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Bruno Bonetti
- Neurology Unit, Azienda Ospedaliera Universitaria Integrata Verona, 37124 Verona, Italy
| | - Raffaella Mariotti
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
- Correspondence: ; Tel.: +39-045-802-7164
| |
Collapse
|
17
|
Ratushnyak MG, Zhirnik AS, Smirnova OD, Semochkina YP, Parfenova AA, Goryunov KV, Silachev DN, Moskaleva EY. The Use of Neural Stem Cells-Derived Exosomes to Prevent Late Radiation-Induced Cognitive Impairments in Mice. Bull Exp Biol Med 2023; 174:571-577. [PMID: 36894818 DOI: 10.1007/s10517-023-05749-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Indexed: 03/11/2023]
Abstract
We studied the effect of intranasal administration of neural stem cell (NSC)-derived exosomes on behavior and cognitive functions of mice in the late period after head irradiation in a dose of 8 Gy. The used exosomes had specific markers (CD9+/CD63+, 99.5%; TSG101+, 98.4%) and mean size 105.7±8.8 nm according to dynamic light scattering data and 119.0±12.4 nm according to nanoparticle tracking analysis (NTA). Exosome suspension (2×1012 particles/ml according to NTA measurements) was administered intranasally for 4 weeks starting from 48 h after irradiation in a volume of 5 μl/nostril (2×1010 exosomes/mouse). It was shown that intranasal administration of mouse NSC-derived exosomes prevented delayed radiation-induced behavioral changes and recognition memory impairments in mice after head irradiation.
Collapse
Affiliation(s)
- M G Ratushnyak
- National Research Center "Kurchatov Institute", Moscow, Russia.
| | - A S Zhirnik
- National Research Center "Kurchatov Institute", Moscow, Russia
| | - O D Smirnova
- National Research Center "Kurchatov Institute", Moscow, Russia
| | - Yu P Semochkina
- National Research Center "Kurchatov Institute", Moscow, Russia
| | - A A Parfenova
- National Research Center "Kurchatov Institute", Moscow, Russia
| | - K V Goryunov
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - D N Silachev
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E Yu Moskaleva
- National Research Center "Kurchatov Institute", Moscow, Russia
| |
Collapse
|
18
|
Hajinejad M, Ebrahimzadeh MH, Ebrahimzadeh-Bideskan A, Rajabian A, Gorji A, Sahab Negah S. Exosomes and Nano-SDF Scaffold as a Cell-Free-Based Treatment Strategy Improve Traumatic Brain Injury Mechanisms by Decreasing Oxidative Stress, Neuroinflammation, and Increasing Neurogenesis. Stem Cell Rev Rep 2023; 19:1001-1018. [PMID: 36652144 DOI: 10.1007/s12015-022-10483-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2022] [Indexed: 01/19/2023]
Abstract
Traumatic brain injury (TBI) causes a variety of complex pathological changes in brain parenchymal tissue by increasing neuroinflammatory and apoptosis responses. Currently, there is no treatment to resolve the consequences related to TBI. Recently, an extensive literature has grown up around the theme of bystander effects of stem cells, a mechanism of stem cells without the need for cell transplantation, which is called cell-free therapy. The purpose of this investigation was to determine the efficacy of a cell-free-based therapy strategy using exosomes derived from human neural stem cells (hNSCs) and a novel nano-scaffold in rats subjected to TBI. In this study, a series of in vitro and in vivo experiments from behavior tests to gene expression was performed to define the effect of exosomes in combination with a three-dimensional (3D) nano-scaffold containing a bio-motif of SDF1α (Nano-SDF). Application of exosomes with Nano-SDF significantly decreased oxidative stress in serum and brain samples. Moreover, treatment with exosomes and Nano-SDF significantly reduced the expression of Toll-like receptor 4 and its downstream signaling pathway, including NF-kβ and interleukin-1β. We also found that the cell-free-based therapy strategy could decrease reactive gliosis at the injury site. Interestingly, we showed that exosomes with Nano-SDF increased neurogenesis in the sub-ventricular zone of the lateral ventricle, indicating a bio-bridge mechanism. To sum up, the most obvious finding to emerge from this study is that a cell-free-based therapy strategy can be an effective option for future practice in the course of TBI.
Collapse
Affiliation(s)
- Mehrdad Hajinejad
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Arezoo Rajabian
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, 48149, Munster, Germany
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd, Mashhad, Iran.
| |
Collapse
|
19
|
Baratta AM, Mangieri RA, Aziz HC, Lopez MF, Farris SP, Homanics GE. Effect of chronic intermittent ethanol vapor exposure on RNA content of brain-derived extracellular vesicles. Alcohol 2022; 105:9-24. [PMID: 36055466 PMCID: PMC10173183 DOI: 10.1016/j.alcohol.2022.08.006] [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: 03/17/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 01/26/2023]
Abstract
Extracellular vesicles (EVs) are important players in normal biological function and disease pathogenesis. Of the many biomolecules packaged into EVs, coding and noncoding RNA transcripts are of particular interest for their ability to significantly alter cellular and molecular processes. Here we investigate how chronic ethanol exposure impacts EV RNA cargo and the functional outcomes of these changes. Following chronic intermittent ethanol (CIE) vapor exposure, EVs were isolated from male and female C57BL/6J mouse brain. Total RNA from EVs was analyzed by lncRNA/mRNA microarray to survey changes in RNA cargo following vapor exposure. Differential expression analysis of microarray data revealed a number of lncRNA and mRNA types differentially expressed in CIE compared to control EVs. Weighted gene co-expression network analysis identified multiple male and female specific modules related to neuroinflammation, cell death, demyelination, and synapse organization. To functionally test these changes, whole-cell voltage-clamp recordings were used to assess synaptic transmission. Incubation of nucleus accumbens brain slices with EVs led to a reduction in spontaneous excitatory postsynaptic current amplitude, although no changes in synaptic transmission were observed between control and CIE EV administration. These results indicate that CIE vapor exposure significantly changes the RNA cargo of brain-derived EVs, which have the ability to impact neuronal function.
Collapse
Affiliation(s)
- Annalisa M Baratta
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Regina A Mangieri
- College of Pharmacy, University of Texas at Austin, Texas, United States
| | - Heather C Aziz
- College of Pharmacy, University of Texas at Austin, Texas, United States
| | - Marcelo F Lopez
- Department of Psychiatry and Behavioral Science, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Sean P Farris
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Gregg E Homanics
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.
| |
Collapse
|
20
|
Effect of human mesenchymal stem cell secretome administration on morphine self-administration and relapse in two animal models of opioid dependence. Transl Psychiatry 2022; 12:462. [PMID: 36333316 PMCID: PMC9636200 DOI: 10.1038/s41398-022-02225-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
The present study investigates the possible therapeutic effects of human mesenchymal stem cell-derived secretome on morphine dependence and relapse. This was studied in a new model of chronic voluntary morphine intake in Wistar rats which shows classic signs of morphine intoxication and a severe naloxone-induced withdrawal syndrome. A single intranasal-systemic administration of MSCs secretome fully inhibited (>95%; p < 0.001) voluntary morphine intake and reduced the post-deprivation relapse intake by 50% (p < 0.02). Since several studies suggest a significant genetic contribution to the chronic use of many addictive drugs, the effect of MSCs secretome on morphine self-administration was further studied in rats bred as high alcohol consumers (UChB rats). Sub-chronic intraperitoneal administration of morphine before access to increasing concentrations of morphine solutions and water were available to the animals, led UChB rats to prefer ingesting morphine solutions over water, attaining levels of oral morphine intake in the range of those in the Wistar model. Intranasally administered MSCs secretome to UChB rats dose-dependently inhibited morphine self-administration by 72% (p < 0.001); while a single intranasal dose of MSC-secretome administered during a morphine deprivation period imposed on chronic morphine consumer UChB rats inhibited re-access morphine relapse intake by 80 to 85% (p < 0.0001). Both in the Wistar and the UChB rat models, MSCs-secretome administration reversed the morphine-induced increases in brain oxidative stress and neuroinflammation, considered as key engines perpetuating drug relapse. Overall, present preclinical studies suggest that products secreted by human mesenchymal stem cells may be of value in the treatment of opioid addiction.
Collapse
|
21
|
Bashyal S, Thapa C, Lee S. Recent progresses in exosome-based systems for targeted drug delivery to the brain. J Control Release 2022; 348:723-744. [PMID: 35718214 DOI: 10.1016/j.jconrel.2022.06.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/08/2022] [Indexed: 12/18/2022]
Abstract
Despite the multiple ongoing and novel initiatives for developing brain-targeted drug delivery systems, insurmountable obstacles remain. A perfect drug delivery device that can bypass the brain-blood barrier and boost therapeutic efficacy is urgently needed for clinical applications. Exosomes hold unrivaled benefits as a drug delivery vehicle for treating brain diseases due to their endogenous and innate attributes. Unique properties, such as the ability to penetrate physical barriers, biocompatibility, innate targeting features, ability to leverage natural intracellular trafficking pathways, favored tumor homing, and stability, make exosomes suitable for brain-targeted drug delivery. Herein, we provide an overview of recent exosome-based drug delivery nanoplatforms and discuss how these inherent vesicles can be used to deliver therapeutic agents to the brain to cure neurodegenerative diseases, brain tumors, and other brain disorders. Moreover, we review the current roadblocks associated with exosomes and other brain-targeted drug delivery systems and discuss future directions for achieving successful therapy outcomes.
Collapse
Affiliation(s)
- Santosh Bashyal
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Chhitij Thapa
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Sangkil Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea.
| |
Collapse
|
22
|
Coleman LG. The emerging world of subcellular biological medicine: extracellular vesicles as novel biomarkers, targets, and therapeutics. Neural Regen Res 2022; 17:1020-1022. [PMID: 34558528 PMCID: PMC8552855 DOI: 10.4103/1673-5374.324846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/17/2021] [Accepted: 06/28/2021] [Indexed: 12/27/2022] Open
Affiliation(s)
- Leon G. Coleman
- The University of North Carolina at Chapel Hill, School of Medicine, Department of Pharmacology, Bowles Center for Alcohol Studies, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Li Y, Wu H, Jiang X, Dong Y, Zheng J, Gao J. New idea to promote the clinical applications of stem cells or their extracellular vesicles in central nervous system disorders: combining with intranasal delivery. Acta Pharm Sin B 2022; 12:3215-3232. [PMID: 35967290 PMCID: PMC9366301 DOI: 10.1016/j.apsb.2022.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/27/2022] [Accepted: 02/14/2022] [Indexed: 12/25/2022] Open
Abstract
The clinical translation of stem cells and their extracellular vesicles (EVs)-based therapy for central nervous system (CNS) diseases is booming. Nevertheless, the insufficient CNS delivery and retention together with the invasiveness of current administration routes prevent stem cells or EVs from fully exerting their clinical therapeutic potential. Intranasal (IN) delivery is a possible strategy to solve problems as IN route could circumvent the brain‒blood barrier non-invasively and fit repeated dosage regimens. Herein, we gave an overview of studies and clinical trials involved with IN route and discussed the possibility of employing IN delivery to solve problems in stem cells or EVs-based therapy. We reviewed relevant researches that combining stem cells or EVs-based therapy with IN administration and analyzed benefits brought by IN route. Finally, we proposed possible suggestions to facilitate the development of IN delivery of stem cells or EVs.
Collapse
Affiliation(s)
- Yaosheng Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghui Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinchi Jiang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yunfei Dong
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Juanjuan Zheng
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
- Corresponding author. Tel.: +86 571 88208436.
| |
Collapse
|
24
|
Quintanilla ME, Ezquer F, Morales P, Santapau D, Ezquer M, Herrera‐Marschitz M, Israel Y. A dual mechanism fully blocks ethanol relapse: Role of vagal innervation. Addict Biol 2022; 27:e13140. [PMID: 35229957 DOI: 10.1111/adb.13140] [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: 09/23/2021] [Revised: 12/20/2021] [Accepted: 01/03/2022] [Indexed: 11/01/2022]
Abstract
Previous studies showed that vagotomy markedly inhibits alcohol self-administration. Present studies hypothesised that vagotomy significantly adds to the inhibition of alcohol relapse induced by drugs that reduce the alcohol-induced hyperglutamatergic state (e.g., N-acetylcysteine + acetylsalicylic acid). The alcohol relapse paradigm tested gauges the elevated alcohol intake observed in animals that had consumed ethanol chronically, were subjected to a prolonged alcohol deprivation and are subsequently allowed ethanol re-access. Ethanol-drinker rats (UChB) were exposed to 10% and 20% ethanol and water concurrently for 4 months, were alcohol deprived for 14 days and were thereafter allowed re-access to the ethanol solutions. An initial binge-like drinking episode is observed upon ethanol re-access, followed by a protracted elevated ethanol intake that exceeds the predeprivation intake baseline. Prior to ethanol re-access, animals were (i) administered N-acetylcysteine (40 mg/kg/day) + acetylsalicylic acid (15 mg/kg/day), (ii) were bilaterally vagotomised, (iii) were exposed to both treatments or (iv) received no treatments. The initial binge-like relapse intake and a protracted elevated ethanol intake observed after repeated ethanol deprivations/re-access cycles were inhibited by 50%-70% by the administration of N-acetylcysteine + acetylsalicylic acid and by 40%-70% by vagotomy, while the combined vagotomy plus N-acetylcysteine + acetylsalicylic acid treatment inhibited both the initial binge-like intake and the protracted ethanol intake by >95% (p < 0.001), disclosing a dual mechanism of ethanol relapse and subsequent inhibition beyond that induced by either treatment alone. Future exploration into the mechanism by which vagal activity contributes to ethanol relapse may have translational promise.
Collapse
Affiliation(s)
- María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine University of Chile Santiago Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa Facultad de Medicina Clínica Alemana‐Universidad del Desarrollo Santiago Chile
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders Santiago Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine University of Chile Santiago Chile
- Department of Neuroscience, Faculty of Medicine University of Chile Santiago Chile
| | - Daniela Santapau
- Centro de Medicina Regenerativa Facultad de Medicina Clínica Alemana‐Universidad del Desarrollo Santiago Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa Facultad de Medicina Clínica Alemana‐Universidad del Desarrollo Santiago Chile
| | - Mario Herrera‐Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine University of Chile Santiago Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine University of Chile Santiago Chile
- Centro de Medicina Regenerativa Facultad de Medicina Clínica Alemana‐Universidad del Desarrollo Santiago Chile
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders Santiago Chile
| |
Collapse
|
25
|
Wang R, Wang X, Zhang Y, Zhao H, Cui J, Li J, Di L. Emerging prospects of extracellular vesicles for brain disease theranostics. J Control Release 2022; 341:844-868. [DOI: 10.1016/j.jconrel.2021.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022]
|
26
|
Holbrook OT, Molligoda B, Bushell KN, Gobrogge KL. Behavioral consequences of the downstream products of ethanol metabolism involved in alcohol use disorder. Neurosci Biobehav Rev 2021; 133:104501. [PMID: 34942269 DOI: 10.1016/j.neubiorev.2021.12.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/08/2021] [Accepted: 12/12/2021] [Indexed: 01/04/2023]
Abstract
Research concerning Alcohol Use Disorder (AUD) has previously focused primarily on either the behavioral or chemical consequences experienced following ethanol intake, but these areas of research have rarely been considered in tandem. Compared with other drugs of abuse, ethanol has been shown to have a unique metabolic pathway once it enters the body, which leads to the formation of downstream metabolites which can go on to form biologically active products. These metabolites can mediate a variety of behavioral responses that are commonly observed with AUD, such as ethanol intake, reinforcement, and vulnerability to relapse. The following review considers the preclinical and chemical research implicating these downstream products in AUD and proposes a chemobehavioral model of AUD.
Collapse
Affiliation(s)
- Otto T Holbrook
- Program in Neuroscience, Boston University, Boston, MA, 02215-2425, USA.
| | - Brandon Molligoda
- Program in Neuroscience, Boston University, Boston, MA, 02215-2425, USA.
| | - Kristen N Bushell
- Program in Neuroscience, Boston University, Boston, MA, 02215-2425, USA
| | - Kyle L Gobrogge
- Program in Neuroscience, Boston University, Boston, MA, 02215-2425, USA
| |
Collapse
|
27
|
Peswani Sajnani SL, Zhang Y, Vllasaliu D. Exosome-based therapies for mucosal delivery. Int J Pharm 2021; 608:121087. [PMID: 34530100 DOI: 10.1016/j.ijpharm.2021.121087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
Exosomes are membrane-bound extracellular nanovesicles secreted by most cells and found in multiple sources, including bodily fluids, plants, fruit, and bovine milk. They play an important role as mediators of intercellular communication, having a distinct ability to carry small molecules, proteins, and nucleic acids to recipient cells over large distances. Moreover, competency in crossing usually poorly permeable biological barriers has led to their promising use in diagnostics and in therapeutics, either as therapeutic entities on their own or as drug delivery vehicles, with superior stability, biocompatibility, circulation time and target specificity in comparison to conventional drug delivery systems. The aim of this review is to summarise and critically discuss the current literature on the use of exosomes in a therapeutic setting, with a particular focus on their use as drug delivery vehicles for mucosal drug delivery.
Collapse
Affiliation(s)
- Shilpa Lekhraj Peswani Sajnani
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, SE1 9NH London, United Kingdom.
| | - Yunyue Zhang
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, SE1 9NH London, United Kingdom.
| | - Driton Vllasaliu
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, SE1 9NH London, United Kingdom.
| |
Collapse
|
28
|
Cuitavi J, Lorente JD, Campos-Jurado Y, Polache A, Hipólito L. Neuroimmune and Mu-Opioid Receptor Alterations in the Mesocorticolimbic System in a Sex-Dependent Inflammatory Pain-Induced Alcohol Relapse-Like Rat Model. Front Immunol 2021; 12:689453. [PMID: 34616393 PMCID: PMC8488159 DOI: 10.3389/fimmu.2021.689453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022] Open
Abstract
Evidence concerning the role of alcohol-induced neuroinflammation in alcohol intake and relapse has increased in the last few years. It is also proven that mu-opioid receptors (MORs) mediate the reinforcing properties of alcohol and, interestingly, previous research suggests that neuroinflammation and MORs could be related. Our objective is to study neuroinflammatory states and microglial activation, together with adaptations on MOR expression in the mesocorticolimbic system (MCLS) during the abstinence and relapse phases. To do so, we have used a sex-dependent rat model of complete Freund's adjuvant (CFA)-induced alcohol deprivation effect (ADE). Firstly, our results confirm that only CFA-treated female rats, the only experimental group that showed relapse-like behavior, exhibited specific alterations in the expression of phosphorylated NFκB, iNOS, and COX2 in the PFC and VTA. More interestingly, the analysis of the IBA1 expression revealed a decrease of the microglial activation in PFC during abstinence and an increase of its expression in the relapse phase, together with an augmentation of this activation in the NAc in both phases that only occur in female CFA-treated rats. Additionally, the expression of IL1β also evidenced these dynamic changes through these two phases following similar expression patterns in both areas. Furthermore, the expression of the cytokine IL10 showed a different profile than that of IL1β, indicating anti-inflammatory processes occurring only during abstinence in the PFC of CFA-female rats but neither during the reintroduction phase in PFC nor in the NAc. These data indicate a downregulation of microglial activation and pro-inflammatory processes during abstinence in the PFC, whereas an upregulation can be observed in the NAc during abstinence that is maintained during the reintroduction phase only in CFA-female rats. Secondly, our data reveal a correlation between the alterations observed in IL1β, IBA1 levels, and MOR levels in the PFC and NAc of CFA-treated female rats. Although premature, our data suggest that neuroinflammatory processes, together with neural adaptations involving MOR, might play an important role in alcohol relapse in female rats, so further investigations are warranted.
Collapse
Affiliation(s)
| | | | | | | | - Lucía Hipólito
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Burjassot, Spain
| |
Collapse
|
29
|
Chivero ET, Dagur RS, Peeples ES, Sil S, Liao K, Ma R, Chen L, Gurumurthy CB, Buch S, Hu G. Biogenesis, physiological functions and potential applications of extracellular vesicles in substance use disorders. Cell Mol Life Sci 2021; 78:4849-4865. [PMID: 33821293 PMCID: PMC10563196 DOI: 10.1007/s00018-021-03824-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/02/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
Substance use disorder (SUD) is a growing health problem that affects several millions of people worldwide, resulting in negative socioeconomic impacts and increased health care costs. Emerging evidence suggests that extracellular vesicles (EVs) play a crucial role in SUD pathogenesis. EVs, including exosomes and microvesicles, are membrane-encapsulated particles that are released into the extracellular space by most types of cells. EVs are important players in mediating cell-to-cell communication through transfer of cargo such as proteins, lipids and nucleic acids. The EV cargo can alter the status of recipient cells, thereby contributing to both physiological and pathological processes; some of these play critical roles in SUD. Although the functions of EVs under several pathological conditions have been extensively reviewed, EV functions and potential applications in SUD remain less studied. In this review, we provide an overview of the current knowledge of the role of EVs in SUD, including alcohol, cocaine, heroin, marijuana, nicotine and opiate abuse. The review will focus on the biogenesis and cargo composition of EVs as well as the potential use of EVs as biomarkers of SUD or therapeutic targets in SUD.
Collapse
Affiliation(s)
- Ernest T Chivero
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| | - Raghubendra Singh Dagur
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68105, USA
| | - Eric S Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA, USA
| | - Rong Ma
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Liang Chen
- Department of Computer Science, College of Engineering, Shantou University, Shantou, Guangdong, China
- Key Laboratory of Intelligent Manufacturing Technology, Ministry of Education, Shantou University, Shantou, Guangdong, China
| | - Channabasavaiah B Gurumurthy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| |
Collapse
|
30
|
Transcriptomics identifies STAT3 as a key regulator of hippocampal gene expression and anhedonia during withdrawal from chronic alcohol exposure. Transl Psychiatry 2021; 11:298. [PMID: 34016951 PMCID: PMC8170676 DOI: 10.1038/s41398-021-01421-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
Alcohol use disorder (AUD) is highly comorbid with depression. Withdrawal from chronic alcohol drinking results in depression and understanding brain molecular mechanisms that drive withdrawal-related depression is important for finding new drug targets to treat these comorbid conditions. Here, we performed RNA sequencing of the rat hippocampus during withdrawal from chronic alcohol drinking to discover key signaling pathways involved in alcohol withdrawal-related depressive-like behavior. Data were analyzed by weighted gene co-expression network analysis to identify several modules of co-expressed genes that could have a common underlying regulatory mechanism. One of the hub, or highly interconnected, genes in module 1 that increased during alcohol withdrawal was the transcription factor, signal transducer and activator of transcription 3 (Stat3), a known regulator of immune gene expression. Total and phosphorylated (p)STAT3 protein levels were also increased in the hippocampus during withdrawal after chronic alcohol exposure. Further, pSTAT3 binding was enriched at the module 1 genes Gfap, Tnfrsf1a, and Socs3 during alcohol withdrawal. Notably, pSTAT3 and its target genes were elevated in the postmortem hippocampus of human subjects with AUD when compared with control subjects. To determine the behavioral relevance of STAT3 activation during alcohol withdrawal, we treated rats with the STAT3 inhibitor stattic and tested for sucrose preference as a measure of anhedonia. STAT3 inhibition alleviated alcohol withdrawal-induced anhedonia. These results demonstrate activation of STAT3 signaling in the hippocampus during alcohol withdrawal in rats and in human AUD subjects, and suggest that STAT3 could be a therapeutic target for reducing comorbid AUD and depression.
Collapse
|
31
|
Shekari F, Nazari A, Assar Kashani S, Hajizadeh-Saffar E, Lim R, Baharvand H. Pre-clinical investigation of mesenchymal stromal cell-derived extracellular vesicles: a systematic review. Cytotherapy 2021; 23:277-284. [PMID: 33541780 DOI: 10.1016/j.jcyt.2020.12.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
The therapeutic potential of naturally secreted micro- and nanoscale extracellular vesicles (EVs) makes them attractive candidates for regenerative medicine and pharmaceutical science applications. To date, the results of numerous publications have shown the practicality of using EVs to replace mesenchymal stromal cells (MSCs) or liposomes. This article presents a systematic review of pre-clinical studies conducted over the past decade of MSC-derived EVs (MSC-EVs) used in animal models of disease. The authors searched the relevant literature in the PubMed and Scopus databases (9358 articles), and 690 articles met the inclusion criteria. The eligible articles were placed in the following disease categories: autoimmune, brain, cancer, eye, gastrointestinal, heart, inflammation/transplantation, liver, musculoskeletal, pancreas, spinal cord and peripheral nervous system, respiratory system, reproductive system, skin, urinary system and vascular-related diseases. Next, the eligible articles were assessed for the rate of publication and global distribution, methodology of EV isolation and characterization, route of MSC-EV administration, length of follow-up, source of MSCs and animal species. The current review classifies and critically discusses the technical aspects of these MSC-EV animal studies and discusses potential relationships between methodological details and the effectiveness of MSC-EVs as reported by these pre-clinical studies.
Collapse
Affiliation(s)
- Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran; Department of Biology, University of Science and Culture, Tehran, Iran; Advanced Therapy Medicinal Product Technology Development Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran.
| | - Abdoreza Nazari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran; Advanced Therapy Medicinal Product Technology Development Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Sara Assar Kashani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran; Advanced Therapy Medicinal Product Technology Development Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Rebecca Lim
- Department of Obstetrics and Gynecology, The Ritchie Center, Hudson Institute of Medical Research, Monash University, Clayton, Australia
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran; Department of Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
32
|
Mao R, Shen J, Hu X. RETRACTED: BMSCs-derived exosomal microRNA-let-7a plays a protective role in diabetic nephropathy via inhibition of USP22 expression. Life Sci 2021; 268:118937. [PMID: 33347877 DOI: 10.1016/j.lfs.2020.118937] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/15/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figs. 6B, and 2F, which appear to have the same eyebrow shaped phenotype as many other publications tabulated here (https://docs.google.com/spreadsheets/d/149EjFXVxpwkBXYJOnOHb6RhAqT4a2llhj9LM60MBffM/edit#gid=0). The journal requested the corresponding author comment on these concerns and provide the raw data. However the authors were not able to satisfactorily fulfil this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Ruiyang Mao
- Department of Medicine, Hangzhou Mingzhou Hospital, Hangzhou 311200, Zhejiang, China
| | - Jiaoni Shen
- Department of Medicine, Hangzhou Mingzhou Hospital, Hangzhou 311200, Zhejiang, China.
| | - Xiaoli Hu
- Department of Medicine, Hangzhou Mingzhou Hospital, Hangzhou 311200, Zhejiang, China
| |
Collapse
|
33
|
Crews FT, Zou J, Coleman LG. Extracellular microvesicles promote microglia-mediated pro-inflammatory responses to ethanol. J Neurosci Res 2021; 99:1940-1956. [PMID: 33611821 PMCID: PMC8451840 DOI: 10.1002/jnr.24813] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022]
Abstract
Alcohol use disorder (AUD) pathology features pro-inflammatory gene induction and microglial activation. The underlying cellular processes that promote this activation remain unclear. Previously considered cellular debris, extracellular vesicles (EVs) have emerged as mediators of inflammatory signaling in several disease states. We investigated the role of microvesicles (MVs, 50 nm-100 µm diameter EVs) in pro-inflammatory and microglial functional gene expression using primary organotypic brain slice culture (OBSC). Ethanol caused a unique immune gene signature that featured: temporal induction of pro-inflammatory TNF-α and IL-1β, reduction of homeostatic microglia state gene Tmem119, progressive increases in purinergic receptor P2RY12 and the microglial inhibitory fractalkine receptor CX3CR1, an increase in the microglial presynaptic gene C1q, and a reduction in the phagocytic gene TREM2. MV signaling was implicated in this response as reduction of MV secretion by imipramine blocked pro-inflammatory TNF-α and IL-1β induction by ethanol, and ethanol-conditioned MVs (EtOH-MVs) reproduced the ethanol-associated immune gene signature in naïve OBSC slices. Depletion of microglia prior to ethanol treatment prevented pro-inflammatory activity of EtOH-MVs, as did incubation of EtOH-MVs with the HMGB1 inhibitor glycyrrhizin. Ethanol caused HMGB1 secretion from cultured BV2 microglia in MVs through activation of PI3 kinase. In summary, these studies find MVs modulate pro-inflammatory gene induction and microglial activation changes associated with ethanol. Thus, MVs may represent a novel therapeutic target to reduce neuroinflammation in the setting of alcohol abuse or other diseases that feature a neuroimmune component. [Correction added on 5 April 2021, after first online publication: The copyright line was changed.].
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA.,Department of Pharmacology, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA.,Department of Psychiatry, The University of North Carolina, School of Medicine, Chapel Hill, NC, USA
| | - Jian Zou
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA.,Department of Pharmacology, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
34
|
Xia C, Dai Z, Jin Y, Chen P. Emerging Antioxidant Paradigm of Mesenchymal Stem Cell-Derived Exosome Therapy. Front Endocrinol (Lausanne) 2021; 12:727272. [PMID: 34912294 PMCID: PMC8667174 DOI: 10.3389/fendo.2021.727272] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cell-derived exosomes have been under investigation as potential treatments for a diverse range of diseases, and many animal and clinical trials have achieved encouraging results. However, it is well known that the biological activity of the exosomes is key to their therapeutic properties; however, till date, it has not been completely understood. Previous studies have provided different explanations of therapeutic mechanisms of the exosomes, including anti-inflammatory, immunomodulatory, and anti-aging mechanisms. The pathological effects of oxidative stress often include organ damage, inflammation, and disorders of material and energy metabolism. The evidence gathered from research involving animal models indicates that exosomes have antioxidant properties, which can also explain their anti-inflammatory and cytoprotective effects. In this study, we have summarized the antioxidant effects of exosomes in in vivo and in vitro models, and have evaluated the anti-oxidant mechanisms of exosomes by demonstrating a direct reduction in excessive reactive oxygen species (ROS), promotion of intracellular defence of anti-oxidative stress, immunomodulation by inhibiting excess ROS, and alteration of mitochondrial performance. Exosomes exert their cytoprotective and anti-inflammatory properties by regulating the redox environment and oxidative stress, which explains the therapeutic effects of exosomes in a variety of diseases, mechanisms that can be well preserved among different species.
Collapse
Affiliation(s)
- Chen Xia
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Department of Orthopedic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhanqiu Dai
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Department of Orthopedic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Bengbu Medical College, Bengbu, China
| | - Yongming Jin
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China
| | - Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
- *Correspondence: Pengfei Chen,
| |
Collapse
|
35
|
Ortega A, Martinez-Arroyo O, Forner MJ, Cortes R. Exosomes as Drug Delivery Systems: Endogenous Nanovehicles for Treatment of Systemic Lupus Erythematosus. Pharmaceutics 2020; 13:pharmaceutics13010003. [PMID: 33374908 PMCID: PMC7821934 DOI: 10.3390/pharmaceutics13010003] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Exosomes, nanometer-sized lipid-bilayer-enclosed extracellular vesicles (EVs), have attracted increasing attention due to their inherent ability to shuttle proteins, lipids and genes between cells and their natural affinity to target cells. Their intrinsic features such as stability, biocompatibility, low immunogenicity and ability to overcome biological barriers, have prompted interest in using exosomes as drug delivery vehicles, especially for gene therapy. Evidence indicates that exosomes play roles in both immune stimulation and tolerance, regulating immune signaling and inflammation. To date, exosome-based nanocarriers delivering small molecule drugs have been developed to treat many prevalent autoimmune diseases. This review highlights the key features of exosomes as drug delivery vehicles, such as therapeutic cargo, use of targeting peptide, loading method and administration route with a broad focus. In addition, we outline the current state of evidence in the field of exosome-based drug delivery systems in systemic lupus erythematosus (SLE), evaluating exosomes derived from various cell types and engineered exosomes.
Collapse
Affiliation(s)
- Ana Ortega
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (A.O.); (O.M.-A.); (M.J.F.)
| | - Olga Martinez-Arroyo
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (A.O.); (O.M.-A.); (M.J.F.)
| | - Maria J. Forner
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (A.O.); (O.M.-A.); (M.J.F.)
- Internal Medicine Unit, Hospital Clinico Universitario, 46010 Valencia, Spain
| | - Raquel Cortes
- Cardiometabolic and Renal Risk Research Group, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (A.O.); (O.M.-A.); (M.J.F.)
- Correspondence: ; Tel.: +34-96398-3916; Fax: +34-96398-7860
| |
Collapse
|
36
|
Recent Advances in Extracellular Vesicles as Drug Delivery Systems and Their Potential in Precision Medicine. Pharmaceutics 2020; 12:pharmaceutics12111006. [PMID: 33105857 PMCID: PMC7690579 DOI: 10.3390/pharmaceutics12111006] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane-bilayered nanoparticles released by most cell types. Recently, an enormous number of studies have been published on the potential of EVs as carriers of therapeutic agents. In contrast to systems such as liposomes, EVs exhibit less immunogenicity and higher engineering potential. Here, we review the most relevant publications addressing the potential and use of EVs as a drug delivery system (DDS). The information is divided based on the key steps for designing an EV-mediated delivery strategy. We discuss possible sources and isolation methods of EVs. We address the administration routes that have been tested in vivo and the tissue distribution observed. We describe the current knowledge on EV clearance, a significant challenge towards enhancing bioavailability. Also, EV-engineering approaches are described as alternatives to improve tissue and cell-specificity. Finally, a summary of the ongoing clinical trials is performed. Although the application of EVs in the clinical practice is still at an early stage, a high number of studies in animals support their potential as DDS. Thus, better treatment options could be designed to precisely increase target specificity and therapeutic efficacy while reducing off-target effects and toxicity according to the individual requirements of each patient.
Collapse
|
37
|
Farfán N, Carril J, Redel M, Zamorano M, Araya M, Monzón E, Alvarado R, Contreras N, Tapia-Bustos A, Quintanilla ME, Ezquer F, Valdés JL, Israel Y, Herrera-Marschitz M, Morales P. Intranasal Administration of Mesenchymal Stem Cell Secretome Reduces Hippocampal Oxidative Stress, Neuroinflammation and Cell Death, Improving the Behavioral Outcome Following Perinatal Asphyxia. Int J Mol Sci 2020; 21:ijms21207800. [PMID: 33096871 PMCID: PMC7589575 DOI: 10.3390/ijms21207800] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Perinatal Asphyxia (PA) is a leading cause of motor and neuropsychiatric disability associated with sustained oxidative stress, neuroinflammation, and cell death, affecting brain development. Based on a rat model of global PA, we investigated the neuroprotective effect of intranasally administered secretome, derived from human adipose mesenchymal stem cells (MSC-S), preconditioned with either deferoxamine (an hypoxia-mimetic) or TNF-α+IFN-γ (pro-inflammatory cytokines). PA was generated by immersing fetus-containing uterine horns in a water bath at 37 °C for 21 min. Thereafter, 16 μL of MSC-S (containing 6 μg of protein derived from 2 × 105 preconditioned-MSC), or vehicle, were intranasally administered 2 h after birth to asphyxia-exposed and control rats, evaluated at postnatal day (P) 7. Alternatively, pups received a dose of either preconditioned MSC-S or vehicle, both at 2 h and P7, and were evaluated at P14, P30, and P60. The preconditioned MSC-S treatment (i) reversed asphyxia-induced oxidative stress in the hippocampus (oxidized/reduced glutathione); (ii) increased antioxidative Nuclear Erythroid 2-Related Factor 2 (NRF2) translocation; (iii) increased NQO1 antioxidant protein; (iv) reduced neuroinflammation (decreasing nuclearNF-κB/p65 levels and microglial reactivity); (v) decreased cleaved-caspase-3 cell-death; (vi) improved righting reflex, negative geotaxis, cliff aversion, locomotor activity, anxiety, motor coordination, and recognition memory. Overall, the study demonstrates that intranasal administration of preconditioned MSC-S is a novel therapeutic strategy that prevents the long-term effects of perinatal asphyxia.
Collapse
Affiliation(s)
- Nancy Farfán
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Jaime Carril
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Martina Redel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Marta Zamorano
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Maureen Araya
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Estephania Monzón
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Raúl Alvarado
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Norton Contreras
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (N.C.); (J.L.V.)
| | - Andrea Tapia-Bustos
- School of Pharmacy, Faculty of Medicine, Universidad Andres Bello, Santiago 8370149, Chile;
| | - María Elena Quintanilla
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - José Luis Valdés
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (N.C.); (J.L.V.)
| | - Yedy Israel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (N.C.); (J.L.V.)
- Correspondence: ; Tel.: +56-229786788
| |
Collapse
|
38
|
Odegaard KE, Chand S, Wheeler S, Tiwari S, Flores A, Hernandez J, Savine M, Gowen A, Pendyala G, Yelamanchili SV. Role of Extracellular Vesicles in Substance Abuse and HIV-Related Neurological Pathologies. Int J Mol Sci 2020; 21:E6765. [PMID: 32942668 PMCID: PMC7554956 DOI: 10.3390/ijms21186765] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are a broad, heterogeneous class of membranous lipid-bilayer vesicles that facilitate intercellular communication throughout the body. As important carriers of various types of cargo, including proteins, lipids, DNA fragments, and a variety of small noncoding RNAs, including miRNAs, mRNAs, and siRNAs, EVs may play an important role in the development of addiction and other neurological pathologies, particularly those related to HIV. In this review, we summarize the findings of EV studies in the context of methamphetamine (METH), cocaine, nicotine, opioid, and alcohol use disorders, highlighting important EV cargoes that may contribute to addiction. Additionally, as HIV and substance abuse are often comorbid, we discuss the potential role of EVs in the intersection of substance abuse and HIV. Taken together, the studies presented in this comprehensive review shed light on the potential role of EVs in the exacerbation of substance use and HIV. As a subject of growing interest, EVs may continue to provide information about mechanisms and pathogenesis in substance use disorders and CNS pathologies, perhaps allowing for exploration into potential therapeutic options.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Sowmya V. Yelamanchili
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.E.O.); (S.C.); (S.W.); (S.T.); (A.F.); (J.H.); (M.S.); (A.G.); (G.P.)
| |
Collapse
|
39
|
León-Moreno LC, Castañeda-Arellano R, Aguilar-García IG, Desentis-Desentis MF, Torres-Anguiano E, Gutiérrez-Almeida CE, Najar-Acosta LJ, Mendizabal-Ruiz G, Ascencio-Piña CR, Dueñas-Jiménez JM, Rivas-Carrillo JD, Dueñas-Jiménez SH. Kinematic Changes in a Mouse Model of Penetrating Hippocampal Injury and Their Recovery After Intranasal Administration of Endometrial Mesenchymal Stem Cell-Derived Extracellular Vesicles. Front Cell Neurosci 2020; 14:579162. [PMID: 33192324 PMCID: PMC7533596 DOI: 10.3389/fncel.2020.579162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/14/2020] [Indexed: 12/20/2022] Open
Abstract
Locomotion speed changes appear following hippocampal injury. We used a hippocampal penetrating brain injury mouse model to analyze other kinematic changes. We found a significant decrease in locomotion speed in both open-field and tunnel walk tests. We described a new quantitative method that allows us to analyze and compare the displacement curves between mice steps. In the tunnel walk, we marked mice with indelible ink on the knee, ankle, and metatarsus of the left and right hindlimbs to evaluate both in every step. Animals with hippocampal damage exhibit slower locomotion speed in both hindlimbs. In contrast, in the cortical injured group, we observed significant speed decrease only in the right hindlimb. We found changes in the displacement patterns after hippocampal injury. Mesenchymal stem cell-derived extracellular vesicles had been used for the treatment of several diseases in animal models. Here, we evaluated the effects of intranasal administration of endometrial mesenchymal stem cell-derived extracellular vesicles on the outcome after the hippocampal injury. We report the presence of vascular endothelial growth factor, granulocyte–macrophage colony-stimulating factor, and interleukin 6 in these vesicles. We observed locomotion speed and displacement pattern preservation in mice after vesicle treatment. These mice had lower pyknotic cells percentage and a smaller damaged area in comparison with the nontreated group, probably due to angiogenesis, wound repair, and inflammation decrease. Our results build up on the evidence of the hippocampal role in walk control and suggest that the extracellular vesicles could confer neuroprotection to the damaged hippocampus.
Collapse
Affiliation(s)
- Lilia Carolina León-Moreno
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico.,Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Rolando Castañeda-Arellano
- Laboratory of Tissue Engineering and Transplant, Department of Physiology, cGMP Cell Processing Facility, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Irene Guadalupe Aguilar-García
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | | | - Elizabeth Torres-Anguiano
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Coral Estefanía Gutiérrez-Almeida
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Luis Jesús Najar-Acosta
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Gerardo Mendizabal-Ruiz
- Department of Computer Sciences, University Center of Exact Sciences and Engineering, University of Guadalajara, Guadalajara, Mexico
| | - César Rodolfo Ascencio-Piña
- Department of Computer Sciences, University Center of Exact Sciences and Engineering, University of Guadalajara, Guadalajara, Mexico
| | - Judith Marcela Dueñas-Jiménez
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Jorge David Rivas-Carrillo
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Sergio Horacio Dueñas-Jiménez
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
40
|
Oxidative Stress and Neuroinflammation as a Pivot in Drug Abuse. A Focus on the Therapeutic Potential of Antioxidant and Anti-Inflammatory Agents and Biomolecules. Antioxidants (Basel) 2020; 9:antiox9090830. [PMID: 32899889 PMCID: PMC7555323 DOI: 10.3390/antiox9090830] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
Drug abuse is a major global health and economic problem. However, there are no pharmacological treatments to effectively reduce the compulsive use of most drugs of abuse. Despite exerting different mechanisms of action, all drugs of abuse promote the activation of the brain reward system, with lasting neurobiological consequences that potentiate subsequent consumption. Recent evidence shows that the brain displays marked oxidative stress and neuroinflammation following chronic drug consumption. Brain oxidative stress and neuroinflammation disrupt glutamate homeostasis by impairing synaptic and extra-synaptic glutamate transport, reducing GLT-1, and system Xc− activities respectively, which increases glutamatergic neurotransmission. This effect consolidates the relapse-promoting effect of drug-related cues, thus sustaining drug craving and subsequent drug consumption. Recently, promising results as experimental treatments to reduce drug consumption and relapse have been shown by (i) antioxidant and anti-inflammatory synthetic molecules whose effects reach the brain; (ii) natural biomolecules secreted by mesenchymal stem cells that excel in antioxidant and anti-inflammatory properties, delivered via non-invasive intranasal administration to animal models of drug abuse and (iii) potent anti-inflammatory microRNAs and anti-miRNAs which target the microglia and reduce neuroinflammation and drug craving. In this review, we address the neurobiological consequences of brain oxidative stress and neuroinflammation that follow the chronic consumption of most drugs of abuse, and the current and potential therapeutic effects of antioxidants and anti-inflammatory agents and biomolecules to reduce these drug-induced alterations and to prevent relapse.
Collapse
|
41
|
Meinhardt MW, Güngör C, Skorodumov I, Mertens LJ, Spanagel R. Psilocybin and LSD have no long-lasting effects in an animal model of alcohol relapse. Neuropsychopharmacology 2020; 45:1316-1322. [PMID: 32369828 PMCID: PMC7298021 DOI: 10.1038/s41386-020-0694-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 01/01/2023]
Abstract
For most psychiatric disorders, including alcohol use disorder (AUD), approved pharmacological treatments are limited in their effectiveness, and new drugs that can easily be translated into the clinic are needed. Currently, great hope lies in the potential of psychedelics to effectively treat AUD. The primary hypothesis is that a single session of psychedelic-guided psychotherapy can restore normal brain function in AUD individuals and thereby reduce the risk of relapse in the long run. Here we applied three different treatment schedules with psilocybin/LSD in order to investigate relapse-like drinking in the alcohol deprivation effect (ADE) model. In contrast to the primary hypothesis, psychedelics had no long-lasting effects on the ADE in male and female rats, neither when administered in a high dosage regime that is comparable to the one used in clinical studies, nor in a chronic microdosing scheme. Only sub-chronic treatment with psilocybin produced a short-lasting anti-relapse effect. However, it is not a translatable treatment option to give psychedelics sub-chronically for relapse prevention. In conclusion, our results in the ADE model do not support the hypothesis that microdosing or high doses of psychedelic reduce relapse behavior. This conclusion has to be confirmed by applying other animal models of AUD. It could also well be that animal models of AUD might be unable to fully capture the therapeutic potential of psychedelic drugs and that only future large-scale clinical trials will be able to demonstrate the efficacy of psychedelics as a new treatment option for AUD.
Collapse
Affiliation(s)
- Marcus W Meinhardt
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
| | - Cansu Güngör
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Ivan Skorodumov
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Lea J Mertens
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
42
|
Fructuoso M, Gu YC, Kassis N, de Lagran MM, Dierssen M, Janel N. Ethanol-Induced Changes in Brain of Transgenic Mice Overexpressing DYRK1A. Mol Neurobiol 2020; 57:3195-3205. [PMID: 32504418 DOI: 10.1007/s12035-020-01967-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 05/29/2020] [Indexed: 12/01/2022]
Abstract
Alcoholism is a chronic relapsing disorder defined by loss of control over excessive consumption of ethanol despite damaging effects on the liver and brain. We previously showed that the overexpression in mice of Dyrk1A (TgDyrk1A, for dual-specificity tyrosine (Y) phosphorylation-regulated kinase 1A) reduces the severity of alcohol mediated liver injury. Ethanol consumption has also been associated with increased brain glutamate concentration that led to therapies targeting glutamatergic receptors and normalization of glutamatergic neurotransmission. Interestingly, mice overexpressing Dyrk1A (TgDyrk1A mice) present a reduction of glutamatergic brain transmission, which we propose could be protective against alcohol intake. To answer this question, we investigated the ethanol preference in TgDyrk1A mice using a two-bottle choice paradigm. TgDyrk1A mice showed a non-significant decrease of voluntary ethanol intake and ethanol preference compared with wild-type mice. At the peripheral level, mice overexpressing Dyrk1A show lower ethanol plasma levels, indicating a faster ethanol metabolism. At the end of the protocol, lasting 21 days, brains were extracted for protein analysis. Ethanol reduced levels of the synaptic protein PSD-95 and increased the glutamate decarboxylase GAD65, specifically in the cortex of TgDyrk1A mice. Our results suggest that overexpression of DYRK1A may cause different ethanol-induced changes in the brain.
Collapse
Affiliation(s)
- Marta Fructuoso
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Pompeu Fabra University (Universitat Pompeu Fabra, UPF), 08003, Barcelona, Spain
- Institut du Cerveau et la Moelle épinière, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Paris, France
| | - Yu Chen Gu
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Nadim Kassis
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Maria Martinez de Lagran
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Pompeu Fabra University (Universitat Pompeu Fabra, UPF), 08003, Barcelona, Spain
| | - Mara Dierssen
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Pompeu Fabra University (Universitat Pompeu Fabra, UPF), 08003, Barcelona, Spain
- Human Pharmacology and Clinical Neurosciences Research Group, Neurosciences Research Program, Hospital Del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Department of Statistics and Operations Research, Universitat Politècnica de Catalunya BarcelonaTech, Barcelona, Spain
| | - Nathalie Janel
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France.
| |
Collapse
|
43
|
Upadhya D, Shetty AK. Extracellular Vesicles as Therapeutics for Brain Injury and Disease. Curr Pharm Des 2020; 25:3500-3505. [PMID: 31612823 DOI: 10.2174/1381612825666191014164950] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) are gaining tremendous importance in comprehending central nervous system (CNS) function and treating neurological disorders because of their role in intercellular communication and reparative processes, and suitability as drug delivery vehicles. Since EVs have lipid membranes, they cross the blood-brain barrier easily and communicate with target neurons and glia even deep inside the brain. EVs from various sources have been isolated, characterized, and tailored for promoting beneficial effects in conditions, such as brain injury and disease. Particularly, EVs isolated from mesenchymal stem cells and neural stem cells have shown promise for alleviating brain dysfunction after injury or disease. Such properties of stem cell-derived EVs have great importance for clinical applications, as EV therapy can avoid several concerns typically associated with cell therapy. This minireview confers the competence of EVs for improving brain function by modulating CNS injury and disease.
Collapse
Affiliation(s)
- Dinesh Upadhya
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, United States
| |
Collapse
|
44
|
Quintanilla ME, Ezquer F, Morales P, Santapau D, Berríos-Cárcamo P, Ezquer M, Herrera-Marschitz M, Israel Y. Intranasal mesenchymal stem cell secretome administration markedly inhibits alcohol and nicotine self-administration and blocks relapse-intake: mechanism and translational options. Stem Cell Res Ther 2019; 10:205. [PMID: 31286996 PMCID: PMC6615104 DOI: 10.1186/s13287-019-1304-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/30/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Chronic consumption of most drugs of abuse leads to brain oxidative stress and neuroinflammation, which inhibit the glutamate transporter GLT-1, proposed to perpetuate drug intake. The present study aimed at inhibiting chronic ethanol and nicotine self-administration and relapse by the non-invasive intranasal administration of antioxidant and anti-inflammatory secretome generated by adipose tissue-derived activated mesenchymal stem cells. The anti-addiction mechanism of stem cell secretome is also addressed. METHODS Rats bred for their alcohol preference ingested alcohol chronically or were trained to self-administer nicotine. Secretome of human adipose tissue-derived activated mesenchymal stem cells was administered intranasally to animals, both (i) chronically consuming alcohol or nicotine and (ii) during a protracted deprivation before a drug re-access leading to relapse intake. RESULTS The intranasal administration of secretome derived from activated mesenchymal stem cells inhibited chronic self-administration of ethanol or nicotine by 85% and 75%, respectively. Secretome administration further inhibited by 85-90% the relapse "binge" intake that occurs after a protracted drug deprivation followed by a 60-min drug re-access. Secretome administration fully abolished the oxidative stress induced by chronic ethanol or nicotine self-administration, shown by the normalization of the hippocampal oxidized/reduced glutathione ratio, and the neuroinflammation determined by astrocyte and microglial immunofluorescence. Knockdown of the glutamate transporter GLT-1 by the intracerebral administration of an antisense oligonucleotide fully abolished the inhibitory effect of the secretome on ethanol and nicotine intake. CONCLUSIONS The non-invasive intranasal administration of secretome generated by human adipose tissue-derived activated mesenchymal stem cells markedly inhibits alcohol and nicotine self-administration, an effect mediated by the glutamate GLT-1 transporter. Translational implications are envisioned.
Collapse
Affiliation(s)
- María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Santiago, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, 7710162, Santiago, RM, Chile.
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Daniela Santapau
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, 7710162, Santiago, RM, Chile
| | - Pablo Berríos-Cárcamo
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, 7710162, Santiago, RM, Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, 7710162, Santiago, RM, Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Santiago, Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Santiago, Chile
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
45
|
Bodega G, Alique M, Puebla L, Carracedo J, Ramírez RM. Microvesicles: ROS scavengers and ROS producers. J Extracell Vesicles 2019; 8:1626654. [PMID: 31258880 PMCID: PMC6586107 DOI: 10.1080/20013078.2019.1626654] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022] Open
Abstract
This review analyzes the relationship between microvesicles and reactive oxygen species (ROS). This relationship is bidirectional; on the one hand, the number and content of microvesicles produced by the cells are affected by oxidative stress conditions; on the other hand, microvesicles can directly and/or indirectly modify the ROS content in the extra- as well as the intracellular compartments. In this regard, microvesicles contain a pro-oxidant or antioxidant machinery that may produce or scavenge ROS: direct effect. This mechanism is especially suitable for eliminating ROS in the extracellular compartment. Endothelial microvesicles, in particular, contain a specific and well-developed antioxidant machinery. On the other hand, the molecules included in microvesicles can modify (activate or inhibit) ROS metabolism in their target cells: indirect effect. This can be achieved by the incorporation into the cells of ROS metabolic enzymes included in the microvesicles, or by the regulation of signaling pathways involved in ROS metabolism. Proteins, as well as miRNAs, are involved in this last effect.
Collapse
Affiliation(s)
- G Bodega
- Departamento de Biomedicina y Biotecnología, Universidad de Alcalá, Alcalá de Henares, Spain
| | - M Alique
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Spain
| | - L Puebla
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Spain
| | - J Carracedo
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
| | - R M Ramírez
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Spain
| |
Collapse
|
46
|
Raimondo S, Giavaresi G, Lorico A, Alessandro R. Extracellular Vesicles as Biological Shuttles for Targeted Therapies. Int J Mol Sci 2019; 20:ijms20081848. [PMID: 30991632 PMCID: PMC6514983 DOI: 10.3390/ijms20081848] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/11/2019] [Accepted: 04/13/2019] [Indexed: 12/11/2022] Open
Abstract
The development of effective nanosystems for drug delivery represents a key challenge for the improvement of most current anticancer therapies. Recent progress in the understanding of structure and function of extracellular vesicles (EVs)—specialized membrane-bound nanocarriers for intercellular communication—suggests that they might also serve as optimal delivery systems of therapeutics. In addition to carrying proteins, lipids, DNA and different forms of RNAs, EVs can be engineered to deliver specific bioactive molecules to target cells. Exploitation of their molecular composition and physical properties, together with improvement in bio-techniques to modify their content are critical issues to target them to specific cells/tissues/organs. Here, we will discuss the current developments in the field of animal and plant-derived EVs toward their potential use for delivery of therapeutic agents in different pathological conditions, with a special focus on cancer.
Collapse
Affiliation(s)
- Stefania Raimondo
- Department of BioMedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy.
| | - Gianluca Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy.
| | - Aurelio Lorico
- Touro University Nevada College of Medicine, Henderson, NV 89014, USA.
- Mediterranean Institute of Oncology Foundation, 95029 Viagrande, Italy.
| | - Riccardo Alessandro
- Department of BioMedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), Section of Biology and Genetics, University of Palermo, 90133 Palermo, Italy.
- Institute of Biomedicine and Molecular Immunology "A. Monroy", National Research Council, 90146 Palermo, Italy.
| |
Collapse
|
47
|
Gene and cell therapy on the acquisition and relapse-like binge drinking in a model of alcoholism: translational options. Gene Ther 2019; 26:407-417. [DOI: 10.1038/s41434-019-0064-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/11/2019] [Indexed: 12/11/2022]
|