1
|
Xue Y, Zheng H, Xiong Y, Li K. Extracellular vesicles affecting embryo development in vitro: a potential culture medium supplement. Front Pharmacol 2024; 15:1366992. [PMID: 39359245 PMCID: PMC11445000 DOI: 10.3389/fphar.2024.1366992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
Extracellular vesicles (EVs) are nanometer-sized lipid bilayer vesicles released by cells, playing a crucial role in mediating cellular communication. This review evaluates the effect of EVs on early embryonic development in vitro by systematically searching the literature across three databases, Embase, PubMed, and Scopus, from inception (Embase, 1947; PubMed, 1996; and Scopus, 2004) to 30 June 2024. A total of 28 studies were considered relevant and included in this review. The EVs included in these investigations have been recovered from a range of sources, including oviduct fluid, follicular fluid, uterine fluid, seminal plasma, embryos, oviduct epithelial cells, endometrial epithelial cells, amniotic cells, and endometrial-derived mesenchymal stem cells collected primarily from mice, rabbits, cattle and pigs. This diversity in EV sources highlights the broad interest and potential applications of EVs in embryo culture systems. These studies have demonstrated that supplementation with EVs derived from physiologically normal biofluids and cells to the embryo culture medium system has positive effects on embryonic development. Conversely, EVs derived from cells under pathological conditions have shown a negative impact. This finding underscores the importance of the source and condition of EVs used in culture media. Further, the addition of EVs as a culture medium supplement holds significant therapeutic potential for optimizing in vitro embryo culture systems. In conclusion, this evaluation offers a thorough assessment of the available data on the role of EVs in embryo culture media and highlights the potential and challenges of using EVs in vitro embryo production.
Collapse
Affiliation(s)
- Yamei Xue
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haixia Zheng
- Institute for Reproductive Health, School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Yuping Xiong
- Institute for Reproductive Health, School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Kun Li
- Institute for Reproductive Health, School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
2
|
Lei Q, Divakarla SK, Winsley T, Roux S, Chrzanowski W. Bioprocessing strategies for enhanced probiotic extracellular vesicle production: culture condition modulation. Front Bioeng Biotechnol 2024; 12:1441552. [PMID: 39280339 PMCID: PMC11392866 DOI: 10.3389/fbioe.2024.1441552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Probiotic extracellular vesicles are biochemically active structures responsible for biological effects elicited by probiotic bacteria. Lactobacillus spp., which are abundant in the human body (e.g., gut), are known to have anti-inflammatory and antimicrobial properties, and are commonly used in food products, supplements, and in discovery research. There is increasing evidence that Lactobacillus-derived extracellular vesicles (LREVs) have potent immunomodulatory capacity that is superior to probiotics themselves. However, key mechanistic insights into the process that controls production and thus, the function of LREVs, are lacking. Currently, it is unknown how the probiotic culture microenvironment orchestrates the type, yield and function of LREVs. Here, we investigated how multifactor modulation of the biomanufacturing process controls the yield and biological functionality of the LREVs. To achieve this, we selected Lacticaseibacillus rhamnosus as the candidate probiotic, initially cultivated under traditional culture conditions, i.e., 100% broth concentration and pH 5.5. Subsequently, we systematically modified the culture conditions of the probiotic by adjusting three critical process parameters: (1) culture medium pH (pH 3.5, 5.5 and 7.5), (2) growth time (48 and 72 h), and (3) broth concentration (50% and 10% of original broth concentration). EVs were then isolated separately from each condition. The critical quality attributes (CQA) of LREVs, including physical characteristics (size, distribution, concentration) and biological composition (protein, carbohydrate, lipid), were analysed. Functional impacts of LREVs on human epidermal keratinocytes and Staphylococcus aureus were also assessed as CQA. Our findings show that the production of LREVs is influenced by environmental stresses induced by the culture conditions. Factors like broth concentration, pH levels, and growth time significantly impact stress levels in L. rhamnosus, affecting both the production and composition of LREVs. Additionally, we have observed that LREVs are non-toxicity for keratinocytes, the major cell type of the epidermis, and possess antimicrobial properties against S. aureus, a common human skin pathogen. These properties are prerequisites for the potential application of EVs to treat skin conditions, including infected wounds. However, the functionality of LREVs depends on the culture conditions and stress levels experienced by L. rhamnosus during production. Understanding this relationship between the culture microenvironment, probiotic stress response, and LREV characteristics, can lead to the biomanufacturing of customised probiotic-derived EVs for various medical and industrial applications.
Collapse
Affiliation(s)
- Qingyu Lei
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, VIC, Australia
| | - Shiva Kamini Divakarla
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, VIC, Australia
- BiomeCentric PTY LTD., Molendinar, QLD, Australia
| | | | - Shaun Roux
- BiomeCentric PTY LTD., Molendinar, QLD, Australia
| | - Wojciech Chrzanowski
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, VIC, Australia
- Department of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Division of Clinical Immunology, Karolinska Institute, Stockholm, Sweden
- Division of Biomedical Engineering, Department of Materials Science and Engineering, Uppsala University, Uppsala, Sweden
| |
Collapse
|
3
|
Arteaga-Blanco LA, Temerozo JR, Tiné LPS, Dantas-Pereira L, Sacramento CQ, Fintelman-Rodrigues N, Toja BM, Gomes Dias SS, de Freitas CS, Espírito-Santo CC, Silva YP, Frozza RL, Bozza PT, Menna-Barreto RFS, Souza TML, Bou-Habib DC. Extracellular vesicles from primary human macrophages stimulated with VIP or PACAP mediate anti-SARS-CoV-2 activities in monocytes through NF-κB signaling pathway. Microbes Infect 2024:105400. [PMID: 39069117 DOI: 10.1016/j.micinf.2024.105400] [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: 01/30/2024] [Revised: 06/28/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Infection by SARS-CoV-2 is associated with uncontrolled inflammatory response during COVID-19 severe disease, in which monocytes are one of the main sources of pro-inflammatory mediators leading to acute respiratory distress syndrome. Extracellular vesicles (EVs) from different cells play important roles during SARS-CoV-2 infection, but investigations describing the involvement of EVs from primary human monocyte-derived macrophages (MDM) on the regulation of this infection are not available. Here, we describe the effects of EVs released by MDM stimulated with the neuropeptides VIP and PACAP on SARS-CoV-2-infected monocytes. MDM-derived EVs were isolated by differential centrifugation of medium collected from cells cultured for 24 h in serum-reduced conditions. Based on morphological properties, we distinguished two subpopulations of MDM-EVs, namely large (LEV) and small EVs (SEV). We found that MDM-derived EVs stimulated with the neuropeptides inhibited SARS-CoV-2 RNA synthesis/replication in monocytes, protected these cells from virus-induced cytopathic effects and reduced the production of pro-inflammatory mediators. In addition, EVs derived from VIP- and PACAP-treated MDM prevented the SARS-CoV-2-induced NF-κB activation. Overall, our findings suggest that MDM-EVs are endowed with immunoregulatory properties that might contribute to the antiviral and anti-inflammatory responses in SARS-CoV-2-infected monocytes and expand our knowledge of EV effects during COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Luis A Arteaga-Blanco
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Jairo R Temerozo
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil; National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Lucas P S Tiné
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Luíza Dantas-Pereira
- Laboratory of Cellular Biology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Carolina Q Sacramento
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil; National Institute for Science and Technology on Innovation in Diseases of Neglected Populations, Center for Technological Development in Health, Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Natalia Fintelman-Rodrigues
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil; National Institute for Science and Technology on Innovation in Diseases of Neglected Populations, Center for Technological Development in Health, Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Beatriz M Toja
- Laboratory of Cellular and Molecular Cardiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, 21941-902, Brazil
| | - Suelen Silva Gomes Dias
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Caroline S de Freitas
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | | | - Ygor P Silva
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Rudimar L Frozza
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Rubem F S Menna-Barreto
- Laboratory of Cellular Biology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Thiago Moreno L Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil; National Institute for Science and Technology on Innovation in Diseases of Neglected Populations, Center for Technological Development in Health, Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Dumith Chequer Bou-Habib
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil; National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil.
| |
Collapse
|
4
|
Zhang X, Ma S, Huebner JL, Naz SI, Alnemer N, Soderblom EJ, Aliferis C, Kraus VB. Immune system-related plasma extracellular vesicles in healthy aging. Front Immunol 2024; 15:1355380. [PMID: 38633262 PMCID: PMC11021711 DOI: 10.3389/fimmu.2024.1355380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
Objectives To identify age-related plasma extracellular vehicle (EVs) phenotypes in healthy adults. Methods EV proteomics by high-resolution mass spectrometry to evaluate EV protein stability and discover age-associated EV proteins (n=4 with 4 serial freeze-thaws each); validation by high-resolution flow cytometry and EV cytokine quantification by multiplex ELISA (n=28 healthy donors, aged 18-83 years); quantification of WI-38 fibroblast cell proliferation response to co-culture with PKH67-labeled young and old plasma EVs. The EV samples from these plasma specimens were previously characterized for bilayer structure, intra-vesicle mitochondria and cytokines, and hematopoietic cell-related surface markers. Results Compared with matched exo-EVs (EV-depleted supernatants), endo-EVs (EV-associated) had higher mean TNF-α and IL-27, lower mean IL-6, IL-11, IFN-γ, and IL-17A/F, and similar mean IL-1β, IL-21, and IL-22 concentrations. Some endo-EV and exo-EV cytokine concentrations were correlated, including TNF-α, IL-27, IL-6, IL-1β, and IFN-γ, but not IL-11, IL-17A/F, IL-21 or IL-22. Endo-EV IFN-γ and exo-EV IL-17A/F and IL-21 declined with age. By proteomics and confirmed by flow cytometry, we identified age-associated decline of fibrinogen (FGA, FGB and FGG) in EVs. Age-related EV proteins indicated predominant origins in the liver and innate immune system. WI-38 cells (>95%) internalized similar amounts of young and old plasma EVs, but cells that internalized PKH67-EVs, particularly young EVs, underwent significantly greater cell proliferation. Conclusion Endo-EV and exo-EV cytokines function as different biomarkers. The observed healthy aging EV phenotype reflected a downregulation of EV fibrinogen subpopulations consistent with the absence of a pro-coagulant and pro-inflammatory condition common with age-related disease.
Collapse
Affiliation(s)
- Xin Zhang
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
- Department of Orthopaedic Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Sisi Ma
- Institute for Health Informatics, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Janet L. Huebner
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Syeda Iffat Naz
- Institute for Health Informatics, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Noor Alnemer
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Erik J. Soderblom
- Duke Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Constantin Aliferis
- Institute for Health Informatics, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
- Department of Orthopaedic Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC, United States
| |
Collapse
|
5
|
Dey S, Mohapatra S, Khokhar M, Hassan S, Pandey RK. Extracellular Vesicles in Malaria: Shedding Light on Pathogenic Depths. ACS Infect Dis 2024; 10:827-844. [PMID: 38320272 PMCID: PMC10928723 DOI: 10.1021/acsinfecdis.3c00649] [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: 11/26/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/08/2024]
Abstract
Malaria, a life-threatening infectious disease caused by Plasmodium falciparum, remains a significant global health challenge, particularly in tropical and subtropical regions. The epidemiological data for 2021 revealed a staggering toll, with 247 million reported cases and 619,000 fatalities attributed to the disease. This formidable global health challenge continues to perplex researchers seeking a comprehensive understanding of its pathogenesis. Recent investigations have unveiled the pivotal role of extracellular vesicles (EVs) in this intricate landscape. These tiny, membrane-bound vesicles, secreted by diverse cells, emerge as pivotal communicators in malaria's pathogenic orchestra. This Review delves into the multifaceted roles of EVs in malaria pathogenesis, elucidating their impact on disease progression and immune modulation. Insights into EV involvement offer potential therapeutic and diagnostic strategies. Integrating this information identifies targets to mitigate malaria's global impact. Moreover, this Review explores the potential of EVs as diagnostic biomarkers and therapeutic targets in malaria. By deciphering the intricate dialogue facilitated by these vesicles, new avenues for intervention and novel strategies for disease management may emerge.
Collapse
Affiliation(s)
- Sangita Dey
- CSO
Department, Cellworks Research India Pvt
Ltd, Bengaluru 560066, Karnataka, India
| | - Salini Mohapatra
- Department
of Biotechnology, Chandigarh University, Punjab 140413, India
| | - Manoj Khokhar
- Department
of Biochemistry, All India Institute of
Medical Sciences Jodhpur, Rajasthan 342005, India
| | - Sana Hassan
- Department
of Life Sciences, Manipal Academy of Higher
Education, Dubai 345050, United Arab Emirates
| | - Rajan Kumar Pandey
- Department
of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17177, Sweden
| |
Collapse
|
6
|
Wang YZ, Castillon CCM, Gebis KK, Bartom ET, d'Azzo A, Contractor A, Savas JN. Notch receptor-ligand binding facilitates extracellular vesicle-mediated neuron-to-neuron communication. Cell Rep 2024; 43:113680. [PMID: 38241148 PMCID: PMC10976296 DOI: 10.1016/j.celrep.2024.113680] [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: 06/15/2023] [Revised: 11/15/2023] [Accepted: 01/01/2024] [Indexed: 01/21/2024] Open
Abstract
Extracellular vesicles (EVs) facilitate intercellular communication by transferring cargo between cells in a variety of tissues. However, how EVs achieve cell-type-specific intercellular communication is still largely unknown. We found that Notch1 and Notch2 proteins are expressed on the surface of neuronal EVs that have been generated in response to neuronal excitatory synaptic activity. Notch ligands bind these EVs on the neuronal plasma membrane, trigger their internalization, activate the Notch signaling pathway, and drive the expression of Notch target genes. The generation of these neuronal EVs requires the endosomal sorting complex required for transport-associated protein Alix. Adult Alix conditional knockout mice have reduced hippocampal Notch signaling activation and glutamatergic synaptic protein expression. Thus, EVs facilitate neuron-to-neuron communication via the Notch receptor-ligand system in the brain.
Collapse
Affiliation(s)
- Yi-Zhi Wang
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Charlotte C M Castillon
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kamil K Gebis
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Elizabeth T Bartom
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Alessandra d'Azzo
- Department of Genetics, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anis Contractor
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jeffrey N Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
7
|
Retana Moreira L, Cornet-Gomez A, Sepulveda MR, Molina-Castro S, Alvarado-Ocampo J, Chaves Monge F, Jara Rojas M, Osuna A, Abrahams Sandí E. Providing an in vitro depiction of microglial cells challenged with immunostimulatory extracellular vesicles of Naegleria fowleri. Front Microbiol 2024; 15:1346021. [PMID: 38374922 PMCID: PMC10876093 DOI: 10.3389/fmicb.2024.1346021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024] Open
Abstract
Naegleria fowleri is the causative agent of primary amoebic meningoencephalitis, a rapid and acute infection of the central nervous system with a fatal outcome in >97% of cases. Due to the infrequent report of cases and diagnostic gaps that hinder the possibility of recovering clinic isolates, studies related to pathogenesis of the disease are scarce. However, the secretion of cytolytic molecules has been proposed as a factor involved in the progression of the infection. Several of these molecules could be included in extracellular vesicles (EVs), making them potential virulence factors and even modulators of the immune response in this infection. In this work, we evaluated the immunomodulatory effect of EVs secreted by two clinic isolates of Naegleria fowleri using in vitro models. For this purpose, characterization analyses between EVs produced by both isolates were first performed, for subsequent gene transcription analyses post incubation of these vesicles with primary cultures from mouse cell microglia and BV-2 cells. Analyses of morphological changes induced in primary culture microglia cells by the vesicles were also included, as well as the determination of the presence of nucleic acids of N. fowleri in the EV fractions. Results revealed increased expression of NOS, proinflammatory cytokines IL-6, TNF-α, and IL-23, and the regulatory cytokine IL-10 in primary cultures of microglia, as well as increased expression of NOS and IL-13 in BV-2 cells. Morphologic changes from homeostatic microglia, with small cellular body and long processes to a more amoeboid morphology were also observed after the incubation of these cells with EVs. Regarding the presence of nucleic acids, specific Naegleria fowleri DNA that could be amplified using both conventional and qPCR was confirmed in the EV fractions. Altogether, these results confirm the immunomodulatory effects of EVs of Naegleria fowleri over microglial cells and suggest a potential role of these vesicles as biomarkers of primary acute meningoencephalitis.
Collapse
Affiliation(s)
- Lissette Retana Moreira
- Departamento de Parasitología, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
- Centro de Investigación en Enfermedades Tropicales (CIET), Universidad de Costa Rica, San José, Costa Rica
| | - Alberto Cornet-Gomez
- Grupo de Bioquímica y Parasitología Molecular (CTS 183), Departamento de Parasitología, Campus de Fuentenueva, Instituto de Biotecnología, Universidad de Granada, Granada, Spain
| | - M. Rosario Sepulveda
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Silvia Molina-Castro
- Instituto de Investigaciones en Salud (INISA), Universidad de Costa Rica, San José, Costa Rica
- Departamento de Bioquímica, Escuela de Medicina, Universidad de Costa Rica, San José, Costa Rica
| | - Johan Alvarado-Ocampo
- Centro de Investigación en Enfermedades Tropicales (CIET), Universidad de Costa Rica, San José, Costa Rica
| | - Frida Chaves Monge
- Departamento de Parasitología, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Mariana Jara Rojas
- Departamento de Parasitología, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Antonio Osuna
- Grupo de Bioquímica y Parasitología Molecular (CTS 183), Departamento de Parasitología, Campus de Fuentenueva, Instituto de Biotecnología, Universidad de Granada, Granada, Spain
| | - Elizabeth Abrahams Sandí
- Departamento de Parasitología, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
- Centro de Investigación en Enfermedades Tropicales (CIET), Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
8
|
Krämer-Albers EM, Werner HB. Mechanisms of axonal support by oligodendrocyte-derived extracellular vesicles. Nat Rev Neurosci 2023:10.1038/s41583-023-00711-y. [PMID: 37258632 DOI: 10.1038/s41583-023-00711-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/02/2023]
Abstract
Extracellular vesicles (EVs) have recently emerged as versatile elements of cell communication in the nervous system, mediating tissue homeostasis. EVs influence the physiology of their target cells via horizontal transfer of molecular cargo between cells and by triggering signalling pathways. In this Review, we discuss recent work revealing that EVs mediate interactions between oligodendrocytes and neurons, which are relevant for maintaining the structural integrity of axons. In response to neuronal activity, myelinating oligodendrocytes release EVs, which are internalized by neurons and provide axons with key factors that improve axonal transport, stress resistance and energy homeostasis. Glia-to-neuron transfer of EVs is thus a crucial facet of axonal preservation. When glial support is impaired, axonal integrity is progressively lost, as observed in myelin-related disorders, other neurodegenerative diseases and with normal ageing. We highlight the mechanisms that oligodendroglial EVs use to sustain axonal integrity and function.
Collapse
Affiliation(s)
- Eva-Maria Krämer-Albers
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|