151
|
Patel G, Agnihotri TG, Gitte M, Shinde T, Gomte SS, Goswami R, Jain A. Exosomes: a potential diagnostic and treatment modality in the quest for counteracting cancer. Cell Oncol (Dordr) 2023; 46:1159-1179. [PMID: 37040056 PMCID: PMC10088756 DOI: 10.1007/s13402-023-00810-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Exosomes are nanosized bio vesicles formed when multivesicular bodies and the plasma membrane merge and discharge into bodily fluids. They are well recognized for facilitating intercellular communication by transporting numerous biomolecules, including DNA, RNAs, proteins, and lipids, and have been implicated in varied diseases including cancer. Exosomes may be altered to transport a variety of therapeutic payloads, including as short interfering RNAs, antisense oligonucleotides, chemotherapeutic drugs, and immunological modulators, and can be directed to a specific target. Exosomes also possess the potential to act as a diagnostic biomarker in cancer, in addition to their therapeutic potential. CONCLUSION In this review, the physiological roles played by exosomes were summarized along with their biogenesis process. Different isolation techniques of exosomes including centrifugation-based, size-based, and polymer precipitation-based techniques have also been described in detail with a special focus on cancer therapeutic applications. The review also shed light on techniques of incubation of drugs with exosomes and their characterization methods covering the most advanced techniques. Myriad applications of exosomes in cancer as diagnostic biomarkers, drug delivery carriers, and chemoresistance-related issues have been discussed at length. Furthermore, a brief overview of exosome-based anti-cancer vaccines and a few prominent challenges concerning exosomal delivery have been concluded at the end.
Collapse
Affiliation(s)
- Gayatri Patel
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Manoj Gitte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Tanuja Shinde
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Roshan Goswami
- Biological E Limited, Plot No-1, Phase 2, Kolthur Village, Medchal District, Shameerpet Mdl, Hyderabad, Telangana, 500078, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
152
|
Al-Dhalimy AMB, Salim HM, Shather AH, Naser IH, Hizam MM, Alshujery MK. The pathological and therapeutically role of mesenchymal stem cell (MSC)-derived exosome in degenerative diseases; Particular focus on LncRNA and microRNA. Pathol Res Pract 2023; 250:154778. [PMID: 37683391 DOI: 10.1016/j.prp.2023.154778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/17/2023] [Accepted: 08/20/2023] [Indexed: 09/10/2023]
Abstract
By releasing exosomes, which create the ideal milieu for the resolution of inflammation, mesenchymal stem cells (MSCs) enhance tissue healing and have strong immunomodulatory capabilities. MSCs-derived exosome also can affect tumor progress by a myriad of mechanisms. Exosomes function as a cell-cell communication tool to affect cellular activity in recipient cells and include an array of efficient bioactive chemicals. Understanding the fundamental biology of inflammation ablation, tissue homeostasis, and the creation of therapeutic strategies is particularly interested in the horizontal transfer of exosomal long non-coding RNAs (lncRNA) and microRNAs (miRNAs) to recipient cells, where they affect target gene expression. Herein, we propose an exosomal lncRNA and microRNA profile in neurological, renal, cardiac, lung, and liver diseases as well as skin wounds and arthritis.
Collapse
Affiliation(s)
| | - Haitham Mukhlif Salim
- Ministry of Health, Directorat of the Public Health, Health Promotion Departments, Baghdad, Iraq
| | - A H Shather
- Department of Computer Engineering Technology, Al Kitab University, Altun Kopru, Kirkuk 00964, Iraq
| | - Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University, 51001 Hillah, Babil, Iraq
| | - Manar Mohammed Hizam
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | | |
Collapse
|
153
|
Varela L, van de Lest CHA, Boere J, Libregts SFWM, Lozano-Andrés E, van Weeren PR, Wauben MHM. Acute joint inflammation induces a sharp increase in the number of synovial fluid EVs and modifies their phospholipid profile. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159367. [PMID: 37473834 DOI: 10.1016/j.bbalip.2023.159367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Inflammation is the hallmark of most joint disorders. However, the precise regulation of induction, perpetuation, and resolution of joint inflammation is not entirely understood. Since extracellular vesicles (EVs) are critical for intercellular communication, we aim to unveil their role in these processes. Here, we investigated the EVs' dynamics and phospholipidome profile from synovial fluid (SF) of healthy equine joints and from horses with lipopolysaccharide (LPS)-induced synovitis. LPS injection triggered a sharp increase of SF-EVs at 5-8 h post-injection, which started to decline at 24 h post-injection. Importantly, we identified significant changes in the lipid profile of SF-EVs after synovitis induction. Compared to healthy joint-derived SF-EVs (0 h), SF-EVs collected at 5, 24, and 48 h post-LPS injection were strongly increased in hexosylceramides. At the same time, phosphatidylserine, phosphatidylcholine, and sphingomyelin were decreased in SF-EVs at 5 h and 24 h post-LPS injection. Based on the lipid changes during acute inflammation, we composed specific lipid profiles associated with healthy and inflammatory state-derived SF-EVs. The sharp increase in SF-EVs during acute synovitis and the correlation of specific lipids with either healthy or inflamed states-derived SF-EVs are findings of potential interest for unveiling the role of SF-EVs in joint inflammation, as well as for the identification of EV-biomarkers of joint inflammation.
Collapse
Affiliation(s)
- Laura Varela
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Chris H A van de Lest
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Janneke Boere
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sten F W M Libregts
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Estefanía Lozano-Andrés
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Division of Infectious Diseases & Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - P René van Weeren
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Marca H M Wauben
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
154
|
Vrablova V, Kosutova N, Blsakova A, Bertokova A, Kasak P, Bertok T, Tkac J. Glycosylation in extracellular vesicles: Isolation, characterization, composition, analysis and clinical applications. Biotechnol Adv 2023; 67:108196. [PMID: 37307942 DOI: 10.1016/j.biotechadv.2023.108196] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/14/2023]
Abstract
This review provides a comprehensive overview of our understanding of the role that glycans play in the formation, loading and release of extracellular vesicles (EVs). The capture of EVs (typically with a size of 100-200 nm) is described, including approaches based on glycan recognition with glycan-based analysis offering highly sensitive detection of EVs. Furthermore, detailed information is provided about the use of EV glycans and glycan processing enzymes as potential biomarkers, therapeutic targets or tools applied for regenerative medicine. The review also provides a short introduction into advanced methods for the characterization of EVs, new insights into the biomolecular corona covering EVs and bioanalytical tools available for glycan analysis.
Collapse
Affiliation(s)
- Veronika Vrablova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 845 38, Slovak Republic
| | - Natalia Kosutova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 845 38, Slovak Republic
| | - Anna Blsakova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 845 38, Slovak Republic
| | - Aniko Bertokova
- Glycanostics sro., Kudlakova 7, Bratislava 841 01, Slovak Republic
| | - Peter Kasak
- Centre for Advanced Materials, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Tomas Bertok
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 845 38, Slovak Republic; Glycanostics sro., Kudlakova 7, Bratislava 841 01, Slovak Republic
| | - Jan Tkac
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 845 38, Slovak Republic; Glycanostics sro., Kudlakova 7, Bratislava 841 01, Slovak Republic.
| |
Collapse
|
155
|
Rani S, Lai A, Nair S, Sharma S, Handberg A, Carrion F, Möller A, Salomon C. Extracellular vesicles as mediators of cell-cell communication in ovarian cancer and beyond - A lipids focus. Cytokine Growth Factor Rev 2023; 73:52-68. [PMID: 37423866 DOI: 10.1016/j.cytogfr.2023.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 06/29/2023] [Indexed: 07/11/2023]
Abstract
Extracellular vesicles (EVs) are messengers that carry information in the form of proteins, lipids, and nucleic acids and are not only essential for intercellular communication but also play a critical role in the progression of various pathologies, including ovarian cancer. There has been recent substantial research characterising EV cargo, specifically, the lipid profile of EVs. Lipids are involved in formation and cargo sorting of EVs, their release and cellular uptake. Numerous lipidomic studies demonstrated the enrichment of specific classes of lipids in EVs derived from cancer cells suggesting that the EV associated lipids can potentially be employed as minimally invasive biomarkers for early diagnosis of various malignancies, including ovarian cancer. In this review, we aim to provide a general overview of the heterogeneity of EV, biogenesis, their lipid content, and function in cancer progression focussing on ovarian cancer.
Collapse
Affiliation(s)
- Shikha Rani
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Andrew Lai
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Soumya Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Shayna Sharma
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD 4029, Australia
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Flavio Carrion
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - Andreas Möller
- Department of Otorhinolaryngology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD 4029, Australia; Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
156
|
Moosazadeh Moghaddam M, Fazel P, Fallah A, Sedighian H, Kachuei R, Behzadi E, Imani Fooladi AA. Host and Pathogen-Directed Therapies against Microbial Infections Using Exosome- and Antimicrobial Peptide-derived Stem Cells with a Special look at Pulmonary Infections and Sepsis. Stem Cell Rev Rep 2023; 19:2166-2191. [PMID: 37495772 DOI: 10.1007/s12015-023-10594-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Microbial diseases are a great threat to global health and cause considerable mortality and extensive economic losses each year. The medications for treating this group of diseases (antibiotics, antiviral, antifungal drugs, etc.) directly attack the pathogenic agents by recognizing the target molecules. However, it is necessary to note that excessive use of any of these drugs can lead to an increase in microbial resistance and infectious diseases. New therapeutic methods have been studied recently using emerging drugs such as mesenchymal stem cell-derived exosomes (MSC-Exos) and antimicrobial peptides (AMPs), which act based on two completely different strategies against pathogens including Host-Directed Therapy (HDT) and Pathogen-Directed Therapy (PDT), respectively. In the PDT approach, AMPs interact directly with pathogens to interrupt their intrusion, survival, and proliferation. These drugs interact directly with the cell membrane or intracellular components of pathogens and cause the death of pathogens or inhibit their replication. The mechanism of action of MSC-Exos in HDT is based on immunomodulation and regulation, promotion of tissue regeneration, and reduced host toxicity. This review studies the potential of mesenchymal stem cell-derived exosomes/ATPs therapeutic properties against microbial infectious diseases especially pulmonary infections and sepsis.
Collapse
Affiliation(s)
- Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Parvindokht Fazel
- Department of Microbiology, Fars Science and Research Branch, Islamic Azad University, Shiraz, Iran
| | - Arezoo Fallah
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- Academy of Medical Sciences of the I.R. of Iran, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
157
|
Boussios S, Devo P, Goodall ICA, Sirlantzis K, Ghose A, Shinde SD, Papadopoulos V, Sanchez E, Rassy E, Ovsepian SV. Exosomes in the Diagnosis and Treatment of Renal Cell Cancer. Int J Mol Sci 2023; 24:14356. [PMID: 37762660 PMCID: PMC10531522 DOI: 10.3390/ijms241814356] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Renal cell carcinoma (RCC) is the most prevalent type of kidney cancer originating from renal tubular epithelial cells, with clear cell RCC comprising approximately 80% of cases. The primary treatment modalities for RCC are surgery and targeted therapy, albeit with suboptimal efficacies. Despite progress in RCC research, significant challenges persist, including advanced distant metastasis, delayed diagnosis, and drug resistance. Growing evidence suggests that extracellular vesicles (EVs) play a pivotal role in multiple aspects of RCC, including tumorigenesis, metastasis, immune evasion, and drug response. These membrane-bound vesicles are released into the extracellular environment by nearly all cell types and are capable of transferring various bioactive molecules, including RNA, DNA, proteins, and lipids, aiding intercellular communication. The molecular cargo carried by EVs renders them an attractive resource for biomarker identification, while their multifarious role in the RCC offers opportunities for diagnosis and targeted interventions, including EV-based therapies. As the most versatile type of EVs, exosomes have attracted much attention as nanocarriers of biologicals, with multi-range signaling effects. Despite the growing interest in exosomes, there is currently no widely accepted consensus on their subtypes and properties. The emerging heterogeneity of exosomes presents both methodological challenges and exciting opportunities for diagnostic and clinical interventions. This article reviews the characteristics and functions of exosomes, with a particular reference to the recent advances in their application to the diagnosis and treatment of RCC.
Collapse
Affiliation(s)
- Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK; (A.G.); (E.S.)
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, Strand, London WC2R 2LS, UK
- Kent Medway Medical School, University of Kent, Canterbury CT2 7LX, UK
- AELIA Organization, 9th Km Thessaloniki–Thermi, 57001 Thessaloniki, Greece
| | - Perry Devo
- School of Sciences, Faculty of Engineering and Science, University of Greenwich, Chatham Maritime ME4 4TB, UK; (P.D.); (I.C.A.G.); (S.V.O.)
| | - Iain C. A. Goodall
- School of Sciences, Faculty of Engineering and Science, University of Greenwich, Chatham Maritime ME4 4TB, UK; (P.D.); (I.C.A.G.); (S.V.O.)
| | - Konstantinos Sirlantzis
- School of Engineering, Technology and Design, Canterbury Christ Church University, Canterbury CT1 1QU, UK;
| | - Aruni Ghose
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK; (A.G.); (E.S.)
- Barts Cancer Centre, Barts Health NHS Trust, London EC1A 7BE, UK
- Mount Vernon Cancer Centre, East and North Hertfordshire NHS Trust, Northwood HA6 2RN, UK
- Immuno-Oncology Clinical Network, London, UK
| | - Sayali D. Shinde
- Centre for Tumour Biology, Barts Cancer Institute, Cancer Research UK Barts Centre, Queen Mary University of London, London EC1M 6BQ, UK;
| | | | - Elisabet Sanchez
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK; (A.G.); (E.S.)
| | - Elie Rassy
- Department of Medical Oncology, Gustave Roussy Institut, 94805 Villejuif, France;
| | - Saak V. Ovsepian
- School of Sciences, Faculty of Engineering and Science, University of Greenwich, Chatham Maritime ME4 4TB, UK; (P.D.); (I.C.A.G.); (S.V.O.)
| |
Collapse
|
158
|
Sayson SG, Ashbaugh A, Cushion MT. Extracellular Vesicles from Pneumocystis carinii -Infected Rats Impair Fungal Viability but are Dispensable for Macrophage Functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558454. [PMID: 37786700 PMCID: PMC10541577 DOI: 10.1101/2023.09.19.558454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Pneumocystis spp. are host obligate fungal pathogens that can cause severe pneumonia in mammals and rely heavily on their host for essential nutrients. The lack of a sustainable in vitro culture system poses challenges in understanding their metabolism and the acquisition of essential nutrients from host lungs remains unexplored. Transmission electron micrographs show Extracellular Vesicles (EVs) are found near Pneumocystis spp. within the lung. We hypothesized that EVs transport essential nutrients to the fungi during infection. To investigate this, EVs from P. carinii and P. murina infected rodents were biochemically and functionally characterized. These EVs contained host proteins involved in cellular, metabolic, and immune processes as well as proteins with homologs found in other fungal EV proteomes, indicating Pneumocystis may release EVs. Notably, EV uptake by P. carinii indicated their potential involvement in nutrient acquisition and indicate a possibility for using engineered EVs for efficient therapeutic delivery. However, EVs added to P. carinii in vitro , did not show increased growth or viability, implying that additional nutrients or factors are necessary to support their metabolic requirements. Exposure of macrophages to EVs increased proinflammatory cytokine levels, but did not affect macrophages' ability to kill or phagocytose P. carinii . These findings provide vital insights into P. carinii and host EV interactions, yet the mechanisms underlying P. carinii 's survival in the lung remain uncertain. These studies are the first to isolate, characterize, and functionally assess EVs from Pneumocystis -infected rodents, promising to enhance our understanding of host-pathogen dynamics and therapeutic potential.
Collapse
|
159
|
Li Z, Wang X, Wang X, Yi X, Wong YK, Wu J, Xie F, Hu D, Wang Q, Wang J, Zhong T. Research progress on the role of extracellular vesicles in neurodegenerative diseases. Transl Neurodegener 2023; 12:43. [PMID: 37697342 PMCID: PMC10494410 DOI: 10.1186/s40035-023-00375-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 08/23/2023] [Indexed: 09/13/2023] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease, affect millions of people worldwide. Tremendous efforts have been put into disease-related research, but few breakthroughs have been made in diagnostic and therapeutic approaches. Extracellular vesicles (EVs) are heterogeneous cell-derived membrane structures that arise from the endosomal system or are directly separated from the plasma membrane. EVs contain many biomolecules, including proteins, nucleic acids, and lipids, which can be transferred between different cells, tissues, or organs, thereby regulating cross-organ communication between cells during normal and pathological processes. Recently, EVs have been shown to participate in various aspects of neurodegenerative diseases. Abnormal secretion and levels of EVs are closely related to the pathogenesis of neurodegenerative diseases and contribute to disease progression. Numerous studies have proposed EVs as therapeutic targets or biomarkers for neurodegenerative diseases. In this review, we summarize and discuss the advanced research progress on EVs in the pathological processes of several neurodegenerative diseases. Moreover, we outline the latest research on the roles of EVs in neurodegenerative diseases and their therapeutic potential for the diseases.
Collapse
Affiliation(s)
- Zhengzhe Li
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Xiaoling Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Xiaoxing Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Xiaomei Yi
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Yin Kwan Wong
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, China
| | - Jiyang Wu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Fangfang Xie
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Die Hu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Qi Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Jigang Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, China.
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Tianyu Zhong
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China.
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
160
|
Faria CP, Ferreira B, Lourenço Á, Guerra I, Melo T, Domingues P, Domingues MDRM, Cruz MT, Sousa MDC. Lipidome of extracellular vesicles from Giardia lamblia. PLoS One 2023; 18:e0291292. [PMID: 37683041 PMCID: PMC10490865 DOI: 10.1371/journal.pone.0291292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Extracellular vesicles (EVs) (exossomes, microvesicles and apoptotic bodies) have been well acknowledged as mediators of intercellular communications in prokaryotes and eukaryotes. Lipids are essential molecular components of EVs but at the moment the knowledge about the lipid composition and the function of lipids in EVs is limited and as for now none lipidomic studies in Giardia EVs was described. Therefore, the focus of the current study was to conduct, for the first time, the characterization of the polar lipidome, namely phospholipid and sphingolipid profiles of G. lamblia trophozoites, microvesicles (MVs) and exosomes, using C18-Liquid Chromatography-Mass Spectrometry (C18-LC-MS) and Tandem Mass Spectrometry (MS/MS). A total of 162 lipid species were identified and semi-quantified, in the trophozoites, or in the MVs and exosomes belonging to 8 lipid classes, including the phospholipid classes phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylglycerol (PG), phosphatidylinositol (PI), cardiolipins (CL), the sphingolipid classes sphingomyelin (SM) and ceramides (Cer), and cholesterol (ST), and 3 lipid subclasses that include lyso PC (LPC), lyso PE (LPE) and lyso PG (LPG), but showing different abundances. This work also identified, for the first time, in G. lamblia trophozoites, the lipid classes CL, Cer and ST and subclasses of LPC, LPE and LPG. Univariate and multivariate analysis showed clear discrimination of lipid profiles between trophozoite, exosomes and MVs. The principal component analysis (PCA) plot of the lipidomics dataset showed clear discrimination between the three groups. Future studies focused on the composition and functional properties of Giardia EVs may prove crucial to understand the role of lipids in host-parasite communication, and to identify new targets that could be exploited to develop novel classes of drugs to treat giardiasis.
Collapse
Affiliation(s)
- Clarissa Perez Faria
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | - Ágata Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Inês Guerra
- Department of Chemistry, CICECO Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
- Department of Chemistry, CESAM Centre for Environmental and Marine Studies, University of Aveiro, Aveiro, Portugal
| | - Tânia Melo
- Department of Chemistry, CESAM Centre for Environmental and Marine Studies, University of Aveiro, Aveiro, Portugal
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Maria do Rosário Marques Domingues
- Department of Chemistry, CESAM Centre for Environmental and Marine Studies, University of Aveiro, Aveiro, Portugal
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Maria Teresa Cruz
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Maria do Céu Sousa
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
161
|
Jang J, Yeo S, Baek S, Jung HJ, Lee MS, Choi SH, Choe Y. Abnormal accumulation of extracellular vesicles in hippocampal dystrophic axons and regulation by the primary cilia in Alzheimer's disease. Acta Neuropathol Commun 2023; 11:142. [PMID: 37667395 PMCID: PMC10478284 DOI: 10.1186/s40478-023-01637-3] [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: 06/21/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
Dystrophic neurites (DNs) are abnormal axons and dendrites that are swollen or deformed in various neuropathological conditions. In Alzheimer's disease (AD), DNs play a crucial role in impairing neuronal communication and function, and they may also contribute to the accumulation and spread of amyloid beta (Aβ) in the brain of AD patients. However, it is still a challenge to understand the DNs of specific neurons that are vulnerable to Aβ in the pathogenesis of AD. To shed light on the development of radiating DNs, we examined enriched dystrophic hippocampal axons in a mouse model of AD using a three-dimensional rendering of projecting neurons. We employed the anterograde spread of adeno-associated virus (AAV)1 and conducted proteomic analysis of synaptic compartments obtained from hippocampo-septal regions. Our findings revealed that DNs were formed due to synaptic loss at the axon terminals caused by the accumulation of extracellular vesicle (EV). Abnormal EV-mediated transport and exocytosis were identified in association with primary cilia, indicating their involvement in the accumulation of EVs at presynaptic terminals. To further address the regulation of DNs by primary cilia, we conducted knockdown of the Ift88 gene in hippocampal neurons, which impaired EV-mediated secretion of Aβ and promoted accumulation of axonal spheroids. Using single-cell RNA sequencing, we identified the septal projecting hippocampal somatostatin neurons (SOM) as selectively vulnerable to Aβ with primary cilia dysfunction and vesicle accumulation. Our study suggests that DNs in AD are initiated by the ectopic accumulation of EVs at the neuronal axon terminals, which is affected by neuronal primary cilia.
Collapse
Affiliation(s)
| | - Seungeun Yeo
- Korea Brain Research Institute, Daegu, 41068, Korea
| | | | | | - Mi Suk Lee
- Korea Brain Research Institute, Daegu, 41068, Korea
| | | | - Youngshik Choe
- Korea Brain Research Institute, Daegu, 41068, Korea.
- , Daegu, Korea.
| |
Collapse
|
162
|
Wang Y, Jiang M, Zheng X, He Y, Ma X, Li J, Pu K. Application of exosome engineering modification in targeted delivery of therapeutic drugs. Biochem Pharmacol 2023; 215:115691. [PMID: 37481135 DOI: 10.1016/j.bcp.2023.115691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 07/24/2023]
Abstract
Cancer is the leading cause of premature death in humans. Scientists have developed several therapeutic drugs for cancer treatment. However, drug delivery faces many problems. First, traditional drugs do not target tumors and are prone to causing significant toxic side effects. Second, suitable drug carriers are essential for improving drug delivery to tumors or circulating cancer cells. Exosomes are natural extracellular vesicles with low immunogenicity and prolonged blood circulation in vivo. These characteristics render exosomes ideal drug carriers. This review highlights the properties of exosomes and mechanisms of exosome biogenesis. It also summarizes the engineering modification methods for enhancing exosome yield, targeting, and drug-loading capacity.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Nano-Bio-Chem Centre and Key Laboratory for Nano-Bio Interface Research, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Min Jiang
- Nano-Bio-Chem Centre and Key Laboratory for Nano-Bio Interface Research, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Xuewen Zheng
- Nano-Bio-Chem Centre and Key Laboratory for Nano-Bio Interface Research, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yiran He
- Nano-Bio-Chem Centre and Key Laboratory for Nano-Bio Interface Research, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Xiaochuan Ma
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) & Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Jiong Li
- Nano-Bio-Chem Centre and Key Laboratory for Nano-Bio Interface Research, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Kefeng Pu
- Nano-Bio-Chem Centre and Key Laboratory for Nano-Bio Interface Research, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| |
Collapse
|
163
|
Tamrin SH, Phelps J, Nezhad AS, Sen A. Critical considerations in determining the surface charge of small extracellular vesicles. J Extracell Vesicles 2023; 12:e12353. [PMID: 37632212 PMCID: PMC10457570 DOI: 10.1002/jev2.12353] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 08/27/2023] Open
Abstract
Small extracellular vesicles (EVs) have emerged as a focal point of EV research due to their significant role in a wide range of physiological and pathological processes within living systems. However, uncertainties about the nature of these vesicles have added considerable complexity to the already difficult task of developing EV-based diagnostics and therapeutics. Whereas small EVs have been shown to be negatively charged, their surface charge has not yet been properly quantified. This gap in knowledge has made it challenging to fully understand the nature of these particles and the way they interact with one another, and with other biological structures like cells. Most published studies have evaluated EV charge by focusing on zeta potential calculated using classical theoretical approaches. However, these approaches tend to underestimate zeta potential at the nanoscale. Moreover, zeta potential alone cannot provide a complete picture of the electrical properties of small EVs since it ignores the effect of ions that bind tightly to the surface of these particles. The absence of validated methods to accurately estimate the actual surface charge (electrical valence) and determine the zeta potential of EVs is a significant knowledge gap, as it limits the development of effective label-free methods for EV isolation and detection. In this study, for the first time, we show how the electrical charge of small EVs can be more accurately determined by accounting for the impact of tightly bound ions. This was accomplished by measuring the electrophoretic mobility of EVs, and then analytically correlating the measured values to their charge in the form of zeta potential and electrical valence. In contrast to the currently used theoretical expressions, the employed analytical method in this study enabled a more accurate estimation of EV surface charge, which will facilitate the development of EV-based diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Sara Hassanpour Tamrin
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
- Department of Biomedical Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Biomedical Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
| | - Jolene Phelps
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
- Department of Biomedical Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
| | - Amir Sanati Nezhad
- Department of Biomedical Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Biomedical Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
| | - Arindom Sen
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
- Department of Biomedical Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
| |
Collapse
|
164
|
Cheng WX, Wei SB, Zhou Y, Shao Y, Li MY. Exosomes: potential diagnostic markers and drug carriers for adenomyosis. Front Pharmacol 2023; 14:1216149. [PMID: 37680720 PMCID: PMC10482052 DOI: 10.3389/fphar.2023.1216149] [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: 05/03/2023] [Accepted: 08/14/2023] [Indexed: 09/09/2023] Open
Abstract
Adenomyosis is a common benign gynecological disorder and an important factor leading to infertility in fertile women. Adenomyosis can cause deep lesions and is persistent and refractory in nature due to its tumor-like biological characteristics, such as the ability to implant, adhere, and invade. The pathogenesis of adenomyosis is currently unclear. Therefore, new therapeutic approaches are urgently required. Exosomes are nanoscale vesicles secreted by cells that carry proteins, genetic materials and other biologically active components. Exosomes play an important role in maintaining tissue homeostasis and regulating immune responses and metabolism. A growing body of work has shown that exosomes and their contents are key to the development and progression of adenomyosis. This review discusses the current research progress, future prospects and challenges in this emerging therapeutic tool by providing an overview of the changes in the adenomyosis uterine microenvironment and the biogenesis and functions of exosomes, with particular emphasis on the role of exosomes and their contents in the regulation of cell migration, proliferation, fibrosis formation, neovascularization, and inflammatory responses in adenomyosis.
Collapse
Affiliation(s)
- Wen-Xiu Cheng
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shao-Bin Wei
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yang Zhou
- Trauma Center, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, Shandong, China
| | - Yu Shao
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Mao-Ya Li
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
165
|
Yang Y, Gu H, Zhang K, Guo Z, Wang X, Wei Q, Weng L, Han X, Lv Y, Cao M, Cao P, Huang C, Qiu Z. Exosomal ACADM sensitizes gemcitabine-resistance through modulating fatty acid metabolism and ferroptosis in pancreatic cancer. BMC Cancer 2023; 23:789. [PMID: 37612627 PMCID: PMC10463774 DOI: 10.1186/s12885-023-11239-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/31/2023] [Indexed: 08/25/2023] Open
Abstract
This study aimed to evaluate the potential of exosomes from cancer cells to predict chemoresistance in pancreatic cancer (PC) and explore the molecular mechanisms through RNA-sequencing and mass spectrometry. We sought to understand the connection between the exosomal Medium-chain acyl-CoA dehydrogenase (ACADM) level and the reaction to gemcitabine in vivo and in patients with PC. We employed loss-of-function, gain-of-function, metabolome mass spectrometry, and xenograft models to investigate the effect of exosomal ACADM in chemoresistance in PC. Our results showed that the molecules involved in lipid metabolism in exosomes vary between PC cells with different gemcitabine sensitivity. Exosomal ACADM (Exo-ACADM) was strongly correlated with gemcitabine sensitivity in vivo, which can be used as a predictor for postoperative gemcitabine chemosensitivity in pancreatic patients. Moreover, ACADM was found to regulate the gemcitabine response by affecting ferroptosis through Glutathione peroxidase 4 (GPX4) and mevalonate pathways. It was also observed that ACADM increased the consumption of unsaturated fatty acids and decreased intracellular lipid peroxides and reactive oxygen species (ROS) levels. In conclusion, this research suggests that Exo-ACADM may be a viable biomarker for predicting the responsiveness of patients to chemotherapy.
Collapse
Affiliation(s)
- Yuhan Yang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Haitao Gu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Kundong Zhang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zengya Guo
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiaofeng Wang
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qingyun Wei
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing, 210028, Jiangsu, China
| | - Ling Weng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing, 210028, Jiangsu, China
| | - Xuan Han
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing, 210028, Jiangsu, China
| | - Yan Lv
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing, 210028, Jiangsu, China
| | - Meng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing, 210028, Jiangsu, China.
| | - Peng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing, 210028, Jiangsu, China.
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Chen Huang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| | - Zhengjun Qiu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
166
|
Zou Z, Li H, Xu G, Hu Y, Zhang W, Tian K. Current Knowledge and Future Perspectives of Exosomes as Nanocarriers in Diagnosis and Treatment of Diseases. Int J Nanomedicine 2023; 18:4751-4778. [PMID: 37635911 PMCID: PMC10454833 DOI: 10.2147/ijn.s417422] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/29/2023] [Indexed: 08/29/2023] Open
Abstract
Exosomes, as natural nanocarriers, characterized with low immunogenicity, non-cytotoxicity and targeted delivery capability, which have advantages over synthetic nanocarriers. Recently, exosomes have shown great potential as diagnostic markers for diseases and are also considered as a promising cell-free therapy. Engineered exosomes have significantly enhanced the efficacy and precision of delivering therapeutic agents, and are currently being extensively employed in targeted therapeutic investigations for various ailments, including oncology, inflammatory disorders, and degenerative conditions. Particularly, engineered exosomes enable therapeutic agent loading, targeted modification, evasion of MPS phagocytosis, intelligent control, and bioimaging, and have been developed as multifunctional nano-delivery platforms in recent years. The utilization of bioactive scaffolds that are loaded with exosome delivery has been shown to substantially augment retention, extend exosome release, and enhance efficacy. This approach has advanced from conventional hydrogels to nanocomposite hydrogels, nanofiber hydrogels, and 3D printing, resulting in superior physical and biological properties that effectively address the limitations of natural scaffolds. Additionally, plant-derived exosomes, which can participate in gut flora remodeling via oral administration, are considered as an ideal delivery platform for the treatment of intestinal diseases. Consequently, there is great interest in exosomes and exosomes as nanocarriers for therapeutic and diagnostic applications. This comprehensive review provides an overview of the biogenesis, composition, and isolation methods of exosomes. Additionally, it examines the pathological and diagnostic mechanisms of exosomes in various diseases, including tumors, degenerative disorders, and inflammatory conditions. Furthermore, this review highlights the significance of gut microbial-derived exosomes. Strategies and specific applications of engineered exosomes and bioactive scaffold-loaded exosome delivery are further summarized, especially some new techniques such as large-scale loading technique, macromolecular loading technique, development of multifunctional nano-delivery platforms and nano-scaffold-loaded exosome delivery. The potential benefits of using plant-derived exosomes for the treatment of gut-related diseases are also discussed. Additionally, the challenges, opportunities, and prospects of exosome-based nanocarriers for disease diagnosis and treatment are summarized from both preclinical and clinical viewpoints.
Collapse
Affiliation(s)
- Zaijun Zou
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- School of Graduates, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Han Li
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- School of Graduates, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Gang Xu
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Disease, Dalian, Liaoning Province, 116011, People’s Republic of China
| | - Yunxiang Hu
- School of Graduates, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Weiguo Zhang
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Disease, Dalian, Liaoning Province, 116011, People’s Republic of China
| | - Kang Tian
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Disease, Dalian, Liaoning Province, 116011, People’s Republic of China
| |
Collapse
|
167
|
Murillo Carrasco AG, Otake AH, Macedo-da-Silva J, Feijoli Santiago V, Palmisano G, Andrade LNDS, Chammas R. Deciphering the Functional Status of Breast Cancers through the Analysis of Their Extracellular Vesicles. Int J Mol Sci 2023; 24:13022. [PMID: 37629204 PMCID: PMC10455604 DOI: 10.3390/ijms241613022] [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: 06/30/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer (BC) accounts for the highest incidence of tumor-related mortality among women worldwide, justifying the growing search for molecular tools for the early diagnosis and follow-up of BC patients under treatment. Circulating extracellular vesicles (EVs) are membranous nanocompartments produced by all human cells, including tumor cells. Since minimally invasive methods collect EVs, which represent reservoirs of signals for cell communication, these particles have attracted the interest of many researchers aiming to improve BC screening and treatment. Here, we analyzed the cargoes of BC-derived EVs, both proteins and nucleic acids, which yielded a comprehensive list of potential markers divided into four distinct categories, namely, (i) modulation of aggressiveness and growth; (ii) preparation of the pre-metastatic niche; (iii) epithelial-to-mesenchymal transition; and (iv) drug resistance phenotype, further classified according to their specificity and sensitivity as vesicular BC biomarkers. We discuss the therapeutic potential of and barriers to the clinical implementation of EV-based tests, including the heterogeneity of EVs and the available technologies for analyzing their content, to present a consistent, reproducible, and affordable set of markers for further evaluation.
Collapse
Affiliation(s)
- Alexis Germán Murillo Carrasco
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (A.H.O.); (L.N.d.S.A.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Andreia Hanada Otake
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (A.H.O.); (L.N.d.S.A.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Janaina Macedo-da-Silva
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (J.M.-d.-S.); (V.F.S.); (G.P.)
| | - Veronica Feijoli Santiago
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (J.M.-d.-S.); (V.F.S.); (G.P.)
| | - Giuseppe Palmisano
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (J.M.-d.-S.); (V.F.S.); (G.P.)
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Luciana Nogueira de Sousa Andrade
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (A.H.O.); (L.N.d.S.A.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Roger Chammas
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (A.H.O.); (L.N.d.S.A.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| |
Collapse
|
168
|
Zhou Y, Dong Y, Zhang A, Wu J, Sun Q. The role of mesenchymal stem cells derived exosomes as a novel nanobiotechnology target in the diagnosis and treatment of cancer. Front Bioeng Biotechnol 2023; 11:1214190. [PMID: 37662434 PMCID: PMC10470003 DOI: 10.3389/fbioe.2023.1214190] [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] [Received: 04/29/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs), one of the most common types of stem cells, are involved in the modulation of the tumor microenvironment (TME). With the advancement of nanotechnology, exosomes, especially exosomes secreted by MSCs, have been found to play an important role in the initiation and development of tumors. In recent years, nanobiotechnology and bioengineering technology have been gradually developed to detect and identify exosomes for diagnosis and modify exosomes for tumor treatment. Several novel therapeutic strategies bioengineer exosomes to carry drugs, proteins, and RNAs, and further deliver their encapsulated cargoes to cancer cells through the properties of exosomes. The unique properties of exosomes in cancer treatment include targeting, low immunogenicity, flexibility in modification, and high biological barrier permeability. Nevertheless, the current comprehensive understanding of the roles of MSCs and their secreted exosomes in cancer development remain inadequate. It is necessary to better understand/update the mechanism of action of MSCs-secreted exosomes in cancer development, providing insights for better modification of exosomes through bioengineering technology and nanobiotechnology. Therefore, this review focuses on the role of MSCs-secreted exosomes and bioengineered exosomes in the development, progression, diagnosis, and treatment of cancer.
Collapse
Affiliation(s)
- You Zhou
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yuqing Dong
- China Medical University and Department of Pathology, Shenyang, China
| | - Aixue Zhang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Jibin Wu
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Qiang Sun
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
169
|
Zhang L, Liu J, Zhou C. Current aspects of small extracellular vesicles in pain process and relief. Biomater Res 2023; 27:78. [PMID: 37563666 PMCID: PMC10416402 DOI: 10.1186/s40824-023-00417-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
Small extracellular vesicles (sEVs) have been identified as a noteworthy paracrine mechanism of intercellular communication in diagnosing and managing neurological disorders. Current research suggests that sEVs play a pivotal role in the pathological progression of pain, emphasizing their critical function in the pathological progression of pain in acute and chronic pain models. By facilitating the transfer of diverse molecules, such as proteins, nucleic acids, and metabolites, sEVs can modulate pain signaling transmission in both the central and peripheral nervous systems. Furthermore, the unique molecules conveyed by sEVs in pain disorders indicate their potential as diagnostic biomarkers. The application of sEVs derived from mesenchymal stem cells (MSCs) in regenerative pain medicine has emerged as a promising strategy for pain management. Moreover, modified sEVs have garnered considerable attention in the investigation of pathological processes and therapeutic interventions. This review presents a comprehensive overview of the current knowledge regarding the involvement of sEVs in pain pathogenesis and treatment. Nevertheless, additional research is imperative to facilitate their clinical implementation. Schematic diagram of sEVs in the biogenesis, signal transmission, diagnosis, and treatment of pain disorders. Small extracellular vesicles (sEVs) are secreted by multiple cells, loading with various biomolecules, such as miRNAs, transmembrane proteins, and amino acids. They selectively target other cells and regulating pain signal transmission. The composition of sEVs can serve as valuable biomarkers for pain diagnosis. In particular, mesenchymal stem cell-derived sEVs have shown promise as regenerative medicine for managing multiple pain disorders. Furthermore, by modifying the structure or contents of sEVs, they could potentially be used as a potent analgesic method.
Collapse
Affiliation(s)
- Lanyu Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
170
|
Fitz NF, Sahu A, Lu Y, Ambrosio F, Lefterov I, Koldamova R. Extracellular Vesicles in Young Serum Contribute to the Restoration of Age-Related Brain Transcriptomes and Cognition in Old Mice. Int J Mol Sci 2023; 24:12550. [PMID: 37628730 PMCID: PMC10454174 DOI: 10.3390/ijms241612550] [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: 07/10/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
We have previously demonstrated that circulating extracellular vesicles (EVs) are essential to the beneficial effect of young serum on the skeletal muscle regenerative cascade. Here, we show that infusions of young serum significantly improve age-associated memory deficits, and that these effects are abolished after serum depletion of EVs. RNA-seq analysis of the choroid plexus demonstrates EV-mediated effects on genes involved in barrier function and trans-barrier transport. Comparing the differentially expressed genes to recently published chronological aging clock genes reveals a reversal of transcriptomic aging in the choroid plexus. Following young serum treatment, the hippocampal transcriptome demonstrates significant upregulation of the anti-aging gene Klotho, along with an abrogated effect after EV depletion. Transcriptomic profiling of Klotho knockout and heterozygous mice shows the downregulation of genes associated with transport, exocytosis, and lipid transport, while upregulated genes are associated with activated microglia. The results of our study indicate the significance of EVs as vehicles to deliver signals from the periphery to the brain and the importance of Klotho in maintaining brain homeostasis.
Collapse
Affiliation(s)
- Nicholas F. Fitz
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Amrita Sahu
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Yi Lu
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02115, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA 02129, USA
| | - Iliya Lefterov
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Radosveta Koldamova
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
171
|
Skotland T, Llorente A, Sandvig K. Lipids in Extracellular Vesicles: What Can Be Learned about Membrane Structure and Function? Cold Spring Harb Perspect Biol 2023; 15:a041415. [PMID: 37277192 PMCID: PMC10411865 DOI: 10.1101/cshperspect.a041415] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Extracellular vesicles, such as exosomes, can be used as interesting models to study the structure and function of biological membranes as these vesicles contain only one membrane (i.e., one lipid bilayer). In addition to lipids, they contain proteins, nucleic acids, and various other molecules. The lipid composition of exosomes is here compared to HIV particles and detergent-resistant membranes, which also have a high content of sphingolipids, cholesterol, and phosphatidylserine (PS). We discuss interactions between the lipids in the two bilayers, and especially those between PS 18:0/18:1 in the inner leaflet and the very-long-chain sphingolipids in the outer leaflet, and the importance of cholesterol for these interactions. We also briefly discuss the involvement of ether-linked phospholipids (PLs) in such lipid raft-like structures, and the possible involvement of these and other lipid classes in the formation of exosomes. The urgent need to improve the quality of quantitative lipidomic studies is highlighted.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
- Department of Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, 0167 Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
- Department of Molecular Biosciences, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
172
|
Moorkens K, Leroy JLMR, Quanico J, Baggerman G, Marei WFA. How the Oviduct Lipidomic Profile Changes over Time after the Start of an Obesogenic Diet in an Outbred Mouse Model. BIOLOGY 2023; 12:1016. [PMID: 37508445 PMCID: PMC10376370 DOI: 10.3390/biology12071016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/03/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023]
Abstract
We investigated whether a high-fat/high-sugar (HF/HS) diet alters the lipidomic profile of the oviductal epithelium (OE) and studied the patterns of these changes over time. Female outbred Swiss mice were fed either a control (10% fat) or HF/HS (60% fat, 20% fructose) diet. Mice (n = 3 per treatment per time point) were sacrificed and oviducts were collected at 3 days and 1, 4, 8, 12 and 16 weeks on the diet. Lipids in the OE were imaged using matrix-assisted laser desorption ionisation mass spectrometry imaging. Discriminative m/z values and differentially regulated lipids were determined in the HF/HS versus control OEs at each time point. Feeding the obesogenic diet resulted in acute changes in the lipid profile in the OE already after 3 days, and thus even before the development of an obese phenotype. The changes in the lipid profile of the OE progressively increased and became more persistent after long-term HF/HS diet feeding. Functional annotation revealed a differential abundance of phospholipids, sphingomyelins and lysophospholipids in particular. These alterations appear to be not only caused by the direct accumulation of the excess circulating dietary fat but also a reduction in the de novo synthesis of several lipid classes, due to oxidative stress and endoplasmic reticulum dysfunction. The described diet-induced lipidomic changes suggest alterations in the OE functions and the oviductal microenvironment which may impact crucial reproductive events that take place in the oviduct, such as fertilization and early embryo development.
Collapse
Affiliation(s)
- Kerlijne Moorkens
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Jo L M R Leroy
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Jusal Quanico
- Centre for Proteomics, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Geert Baggerman
- Centre for Proteomics, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
- Health Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400 Mol, Belgium
| | - Waleed F A Marei
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| |
Collapse
|
173
|
Ghadami S, Dellinger K. The lipid composition of extracellular vesicles: applications in diagnostics and therapeutic delivery. Front Mol Biosci 2023; 10:1198044. [PMID: 37520326 PMCID: PMC10381967 DOI: 10.3389/fmolb.2023.1198044] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Extracellular vesicles (EVs), including exosomes, with nanoscale sizes, biological origins, various functions, and unique lipid and protein compositions have been introduced as versatile tools for diagnostic and therapeutic medical applications. Numerous studies have reported the importance of the lipid composition of EVs and its influence on their mechanism of action. For example, changes in the lipidomic profile of EVs have been shown to influence the progression of various diseases, including ovarian malignancies and prostate cancer. In this review, we endeavored to examine differences in the lipid content of EV membranes derived from different cell types to characterize their capabilities as diagnostic tools and treatments for diseases like cancer and Alzheimer's disease. We additionally discuss designing functionalized vesicles, whether synthetically by hybrid methods or by changing the lipid composition of natural EVs. Lastly, we provide an overview of current and potential biomedical applications and perspectives on the future of this growing field.
Collapse
Affiliation(s)
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, United States
| |
Collapse
|
174
|
Guzewska MM, Szuszkiewicz J, Kaczmarek MM. Extracellular vesicles: Focus on peri-implantation period of pregnancy in pigs. Mol Reprod Dev 2023; 90:634-645. [PMID: 36645872 DOI: 10.1002/mrd.23664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 01/18/2023]
Abstract
The establishment of cell-to-cell communication between the endometrium and the developing embryo is the most important step in successful mammalian pregnancy. Close interaction between the uterine luminal epithelium and trophoblast cells requires triggering timely molecular dialog for successful maternal recognition of pregnancy, embryo implantation, and placenta development. Quite recently, extracellular vesicles (EVs) carrying unique molecular cargo emerged as evolutionarily conserved mediators of cell-to-cell communication during early pregnancy. To date, the presence of EVs at the embryo-maternal interface has been demonstrated in numerous mammals, including domestic livestock, such as pigs. However, few studies have focused on revealing the mechanism of EV-mediated crosstalk between developing early embryos and receptive endometrium. Over the past years, it has appeared that understanding the role of EVs in mammalian reproduction can substantially improve our understanding of the biological challenges of successful reproductive performance. This review describes current knowledge of EVs, specifically in relation to the peri-implantation period in pigs, characterized by common features of embryo implantation and high embryonic mortality in mammals.
Collapse
Affiliation(s)
- Maria M Guzewska
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Szuszkiewicz
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Monika M Kaczmarek
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
175
|
Pantazopoulou M, Lamprokostopoulou A, Karampela DS, Alexaki A, Delis A, Coens A, Samiotaki M, Kriebardis AG, Melki R, Pagakis SN, Stefanis L, Vekrellis K. Differential intracellular trafficking of extracellular vesicles in microglia and astrocytes. Cell Mol Life Sci 2023; 80:193. [PMID: 37391572 DOI: 10.1007/s00018-023-04841-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 06/07/2023] [Accepted: 06/15/2023] [Indexed: 07/02/2023]
Abstract
Extracellular vesicles (EVs) have emerged as key players in cell-to-cell communication in both physiological and pathological processes in the Central Nervous System. Thus far, the intracellular pathways involved in uptake and trafficking of EVs within different cell types of the brain are poorly understood. In our study, the endocytic processes and subcellular sorting of EVs were investigated in primary glial cells, particularly linked with the EV-associated α-synuclein (α-syn) transmission. Mouse microglia and astrocytic primary cultures were incubated with DiI-stained mouse brain-derived EVs. The internalization and trafficking pathways were analyzed in cells treated with pharmacological reagents that block the major endocytic pathways. Brain-derived EVs were internalized by both glial cell types; however, uptake was more efficient in microglia than in astrocytes. Colocalization of EVs with early and late endocytic markers (Rab5, Lamp1) indicated that EVs are sorted to endo-lysosomes for subsequent processing. Blocking actin-dependent phagocytosis and/or macropinocytosis with Cytochalasin D or EIPA inhibited EV entry into glial cells, whereas treatment with inhibitors that strip cholesterol off the plasma membrane, induced uptake, however differentially altered endosomal sorting. EV-associated fibrillar α-Syn was efficiently internalized and detected in Rab5- and Lamp1-positive compartments within microglia. Our study strongly suggests that EVs enter glial cells through phagocytosis and/or macropinocytosis and are sorted to endo-lysosomes for subsequent processing. Further, brain-derived EVs serve as scavengers and mediate cell-to-glia transfer of pathological α-Syn which is also targeted to the endolysosomal pathway, suggesting a beneficial role in microglia-mediated clearance of toxic protein aggregates, present in numerous neurodegenerative diseases.
Collapse
Affiliation(s)
- Marina Pantazopoulou
- Biomedical Research Foundation Academy of Athens-BRFAA, Clinical-Experimental Surgery & Translational Research, 4, Soranou Tou Efesiou Street, 11527, Athens, Greece.
| | | | | | - Anastasia Alexaki
- Biomedical Research Foundation Academy of Athens-BRFAA, Centre of Basic Research, Athens, Greece
| | - Anastasios Delis
- Biomedical Research Foundation Academy of Athens-BRFAA, Centre of Basic Research, Athens, Greece
| | - Audrey Coens
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-Aux-Roses Cedex, France
| | - Martina Samiotaki
- Institute for Bioinnovation, Biomedical Sciences Research Center 'Alexander Fleming', Fleming 34, 16672, Vari, Greece
| | - Anastasios G Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece
| | - Ronald Melki
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-Aux-Roses Cedex, France
| | - Stamatis N Pagakis
- Biomedical Research Foundation Academy of Athens-BRFAA, Centre of Basic Research, Athens, Greece
| | - Leonidas Stefanis
- Biomedical Research Foundation Academy of Athens-BRFAA, Clinical-Experimental Surgery & Translational Research, 4, Soranou Tou Efesiou Street, 11527, Athens, Greece
| | - Kostas Vekrellis
- Biomedical Research Foundation Academy of Athens-BRFAA, Centre of Basic Research, Athens, Greece
| |
Collapse
|
176
|
Wang T, Koukoulis TF, Vella LJ, Su H, Purnianto A, Nie S, Ang CS, Ma G, Korhonen PK, Taki AC, Williamson NA, Reid GE, Gasser RB. The Proteome and Lipidome of Extracellular Vesicles from Haemonchus contortus to Underpin Explorations of Host-Parasite Cross-Talk. Int J Mol Sci 2023; 24:10955. [PMID: 37446130 DOI: 10.3390/ijms241310955] [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: 06/13/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Many parasitic worms have a major adverse impact on human and animal populations worldwide due to the chronicity of their infections. There is a growing body of evidence indicating that extracellular vesicles (EVs) are intimately involved in modulating (suppressing) inflammatory/immune host responses and parasitism. As one of the most pathogenic nematodes of livestock animals, Haemonchus contortus is an ideal model system for EV exploration. Here, employing a multi-step enrichment process (in vitro culture, followed by ultracentrifugation, size exclusion and filtration), we enriched EVs from H. contortus and undertook the first comprehensive (qualitative and quantitative) multi-omic investigation of EV proteins and lipids using advanced liquid chromatography-mass spectrometry and informatics methods. We identified and quantified 561 proteins and 446 lipids in EVs and compared these molecules with those of adult worms. We identified unique molecules in EVs, such as proteins linked to lipid transportation and lipid species (i.e., sphingolipids) associated with signalling, indicating the involvement of these molecules in parasite-host cross-talk. This work provides a solid starting point to explore the functional roles of EV-specific proteins and lipids in modulating parasite-host cross-talk, and the prospect of finding ways of disrupting or interrupting this relationship to suppress or eliminate parasite infection.
Collapse
Affiliation(s)
- Tao Wang
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Tiana F Koukoulis
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Laura J Vella
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Huaqi Su
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
- Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Adityas Purnianto
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Shuai Nie
- Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Ching-Seng Ang
- Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Guangxu Ma
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia
- Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Pasi K Korhonen
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Aya C Taki
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Nicholas A Williamson
- Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Gavin E Reid
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, School of Chemistry, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Robin B Gasser
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
177
|
Shi YJ, Fang YX, Tian TG, Chen WP, Sun Q, Guo FQ, Gong PQ, Li CM, Wang H, Hu ZQ, Li XX. Discovery of extracellular vesicle-delivered miR-185-5p in the plasma of patients as an indicator for advanced adenoma and colorectal cancer. J Transl Med 2023; 21:421. [PMID: 37386465 PMCID: PMC10308673 DOI: 10.1186/s12967-023-04249-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/05/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND We aimed to evaluate whether extracellular vesicles (EV)-derived microRNAs (miRNAs) can be used as biomarkers for advanced adenoma (AA) and colorectal cancer (CRC). METHODS We detected the changes in the plasma EV-delivered miRNA profiles in healthy donor (HD), AA patient, and I-II stage CRC patient groups using miRNA deep sequencing assay. We performed the TaqMan miRNA assay using 173 plasma samples (two independent cohorts) from HDs, AA patients, and CRC patients to identify the candidate miRNA(s). The accuracy of candidate miRNA(s) in diagnosing AA and CRC was determined using the area under the receiver-operating characteristic curve (AUC) values. Logistic regression analysis was performed to evaluate the association of candidate miRNA(s) as an independent factor for the diagnosis of AA and CRC. The role of candidate miRNA(s) in the malignant progression of CRC was explored using functional assays. RESULTS We screened and identified four prospective EV-delivered miRNAs, including miR-185-5p, which were significantly upregulated or downregulated in AA vs. HD and CRC vs. AA groups. In two independent cohorts, miR-185-5p was the best potential biomarker with the AUCs of 0.737 (Cohort I) and 0.720 (Cohort II) for AA vs. HD diagnosis, 0.887 (Cohort I) and 0.803 (Cohort II) for CRC vs. HD diagnosis, and 0.700 (Cohort I) and 0.631 (Cohort II) for CRC vs. AA diagnosis. Finally, we demonstrated that the upregulated expression of miR-185-5p promoted the malignant progression of CRC. CONCLUSION EV-delivered miR-185-5p in the plasma of patients is a promising diagnostic biomarker for colorectal AA and CRC. Trial registration The study protocol was approved by the Ethics Committee of Changzheng Hospital, Naval Medical University, China (Ethics No. 2022SL005, Registration No. of China Clinical Trial Registration Center: ChiCTR220061592).
Collapse
Affiliation(s)
- Yun-Jie Shi
- Department of Anorectal Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
- Department of Anorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Yu-Xiang Fang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Tong-Guan Tian
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
| | - Wei-Ping Chen
- Institute of Basic Medicine and Cancer (IBMC), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Qiang Sun
- Department of Gastrointestinal Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Fang-Qi Guo
- Department of Ultrasound, Shanghai Fourth People' Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Pi-Qing Gong
- Department of Anorectal Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Chun-Mei Li
- Institute of Basic Medicine and Cancer (IBMC), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Hao Wang
- Department of Anorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200433, China.
| | - Zhi-Qian Hu
- Department of Anorectal Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China.
- Department of General Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Xin-Xing Li
- Department of General Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
178
|
Zhang Y, Dou Y, Liu Y, Di M, Bian H, Sun X, Yang Q. Advances in Therapeutic Applications of Extracellular Vesicles. Int J Nanomedicine 2023; 18:3285-3307. [PMID: 37346366 PMCID: PMC10281276 DOI: 10.2147/ijn.s409588] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/08/2023] [Indexed: 06/23/2023] Open
Abstract
Extracellular vesicles (EVs) are nanoscale bilayer phospholipid membrane vesicles released by cells. Contained large molecules such as nucleic acid, protein, and lipid, EVs are an integral part of cell communication. The contents of EVs vary based on the cell source and play an important role in both pathological and physiological conditions. EVs can be used as drugs or targets in disease treatment, and changes in the contents of EVs can indicate the progression of diseases. In recent years, with the continuous exploration of the structure, characteristics, and functions of EVs, the potential of engineered EVs for drug delivery and therapy being constantly explored. This review provides a brief overview of the structure, characteristics and functions of EVs, summarizes the advanced application of EVs and outlook on the prospect of it. It is our hope that this review will increase understanding of the current development of medical applications of EVs and help us overcome future challenges.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yang Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Mingyuan Di
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Hanming Bian
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| |
Collapse
|
179
|
Yakovlev V, Lapato DM, Rana P, Ghosh P, Frye R, Roberson-Nay R. Neuron Enriched Exosomal MicroRNA Expression Profiles as a Marker of Early Life Alcohol Consumption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.09.544235. [PMID: 37333185 PMCID: PMC10274862 DOI: 10.1101/2023.06.09.544235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Alcohol consumption may impact and shape brain development through perturbed biological pathways and impaired molecular functions. We investigated the relationship between alcohol consumption rates and neuron-enriched exosomal microRNA (miRNA) expression to better understand the impact of alcohol use on early life brain biology. Methods Neuron-enriched exosomal miRNA expression was measured from plasma samples collected from young people using a commercially available microarray platform while alcohol consumption was measured using the Alcohol Use Disorders Identification Test. Linear regression and network analyses were used to identify significantly differentially expressed miRNAs and to characterize the implicated biological pathways, respectively. Results Compared to alcohol naïve controls, young people reporting high alcohol consumption exhibited significantly higher expression of four neuron-enriched exosomal miRNAs including miR-30a-5p, miR-194-5p, and miR-339-3p, although only miR-30a-5p and miR-194-5p survived multiple test correction. The miRNA-miRNA interaction network inferred by a network inference algorithm did not detect any differentially expressed miRNAs with a high cutoff on edge scores. However, when the cutoff of the algorithm was reduced, five miRNAs were identified as interacting with miR-194-5p and miR-30a-5p. These seven miRNAs were associated with 25 biological functions; miR-194-5p was the most highly connected node and was highly correlated with the other miRNAs in this cluster. Conclusions Our observed association between neuron-enriched exosomal miRNAs and alcohol consumption concurs with results from experimental animal models of alcohol use and suggests that high rates of alcohol consumption during the adolescent/young adult years may impact brain functioning and development by modulating miRNA expression.
Collapse
Affiliation(s)
- Vasily Yakovlev
- Department of Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Dana M Lapato
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Preetam Ghosh
- Department of Computer Science, College of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Rebekah Frye
- Neuroscience Program, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Roxann Roberson-Nay
- Department of Psychiatry, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
180
|
Zhou C, Huang YQ, Da MX, Jin WL, Zhou FH. Adipocyte-derived extracellular vesicles: bridging the communications between obesity and tumor microenvironment. Discov Oncol 2023; 14:92. [PMID: 37289328 PMCID: PMC10250291 DOI: 10.1007/s12672-023-00704-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/26/2023] [Indexed: 06/09/2023] Open
Abstract
By the year 2035 more than 4 billion people might be affected by obesity and being overweight. Adipocyte-derived Extracellular Vesicles (ADEVs/ADEV-singular) are essential for communication between the tumor microenvironment (TME) and obesity, emerging as a prominent mechanism of tumor progression. Adipose tissue (AT) becomes hypertrophic and hyperplastic in an obese state resulting in insulin resistance in the body. This modifies the energy supply to tumor cells and simultaneously stimulates the production of pro-inflammatory adipokines. In addition, obese AT has a dysregulated cargo content of discharged ADEVs, leading to elevated amounts of pro-inflammatory proteins, fatty acids, and carcinogenic microRNAs. ADEVs are strongly associated with hallmarks of cancer (proliferation and resistance to cell death, angiogenesis, invasion, metastasis, immunological response) and may be useful as biomarkers and antitumor therapy strategy. Given the present developments in obesity and cancer-related research, we conclude by outlining significant challenges and significant advances that must be addressed expeditiously to promote ADEVs research and clinical applications.
Collapse
Affiliation(s)
- Chuan Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000 People’s Republic of China
| | - Yu-Qian Huang
- Department of Center of Medical Cosmetology, Chengdu Second People’s Hospital, Chengdu, 610017 People’s Republic of China
| | - Ming-Xu Da
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000 People’s Republic of China
| | - Wei-Lin Jin
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Feng-Hai Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- Department of Urology, Gansu Provincial Hospital, Lanzhou, 730000 People’s Republic of China
| |
Collapse
|
181
|
Debnath K, Heras KL, Rivera A, Lenzini S, Shin JW. Extracellular vesicle-matrix interactions. NATURE REVIEWS. MATERIALS 2023; 8:390-402. [PMID: 38463907 PMCID: PMC10919209 DOI: 10.1038/s41578-023-00551-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/24/2023] [Indexed: 03/12/2024]
Abstract
The extracellular matrix in microenvironments harbors a variety of signals to control cellular functions and the materiality of tissues. Most efforts to synthetically reconstitute the matrix by biomaterial design have focused on decoupling cell-secreted and polymer-based cues. Cells package molecules into nanoscale lipid membrane-bound extracellular vesicles and secrete them. Thus, extracellular vesicles inherently interact with the meshwork of the extracellular matrix. In this Review, we discuss various aspects of extracellular vesicle-matrix interactions. Cells receive feedback from the extracellular matrix and leverage intracellular processes to control the biogenesis of extracellular vesicles. Once secreted, various biomolecular and biophysical factors determine whether extracellular vesicles are locally incorporated into the matrix or transported out of the matrix to be taken up by other cells or deposited into tissues at a distal location. These insights can be utilized to develop engineered biomaterials where EV release and retention can be precisely controlled in host tissue to elicit various biological and therapeutic outcomes.
Collapse
Affiliation(s)
- Koushik Debnath
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Kevin Las Heras
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU)
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Ambar Rivera
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60608, USA
| | - Stephen Lenzini
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
182
|
Martínez-Santillán A, González-Valdez J. Novel Technologies for Exosome and Exosome-like Nanovesicle Procurement and Enhancement. Biomedicines 2023; 11:1487. [PMID: 37239158 PMCID: PMC10216008 DOI: 10.3390/biomedicines11051487] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Exosomes are extracellular nanovesicles commonly produced by mammalian cells that in recent years have risen as a novel strategy for drug delivery systems and cancer therapy because of their innate specificity and high bioavailability. However, there are limitations that undermine their potential. Among them is the lack of mass production capacity with the current available sources and the failure to reach the intended therapeutic effect because of their insufficient uptake or their rapid clearance once administered. This review aims to show the current advances in overcoming these limitations by presenting, firstly, reported strategies to improve exosome and exosome-like nanovesicle extraction from possible novel eukaryotic sources, including animals, plants, and protozoa; and secondly, alternative modification methods that functionalize exosomes by conferring them higher targeting capacity and protection from organism defenses, which results in an increase in the attachment of ligands and cellular uptake of inorganic materials. However, even when these strategies might address some of the obstacles in their procurement and therapeutic use, there are still several aspects that need to be addressed, so several perspectives of the matter are also presented and analyzed throughout this work.
Collapse
Affiliation(s)
| | - José González-Valdez
- School of Engineering and Science, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501, Monterrey 64849, Mexico
| |
Collapse
|
183
|
Spada S. Methodologies to evaluate the radiation-induced changes on extracellular vesicles. Methods Cell Biol 2023; 180:39-48. [PMID: 37890931 DOI: 10.1016/bs.mcb.2023.03.002] [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] [Indexed: 10/29/2023]
Abstract
The extracellular vesicles (EVs) are carriers that actively transfer functional biomolecules between cells affecting the intercellular communication (Pitt, Kroemer, & Zitvogel, 2016). EV signaling has consequences on the targeted cell behavior impacting multiple processes from health to disease, including cancer (Yates et al., 2022). Radiation treatment (RT) is one of the gold standard and effective treatments for cancer, as curative or palliative (Chandra, Keane, Voncken, & Thomas, 2021). RT induces different release of EVs and their cargo is altered. In addition, the uptake of EVs secreted by irradiated cells is affected. Hence, a deep investigation is required to better understand how RT influence the cell-to-cell communication thought signals shuttle by EVs. Here, detailed methods to study the RT effects on EV size and secretion, EV protein expression, EV uptake will be described. Alterations and adaptions might make the protocols applicable to different cell lines, and with different types of RT and dose exposures.
Collapse
Affiliation(s)
- Sheila Spada
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
184
|
Zhong Y, Wang X, Zhao X, Shen J, Wu X, Gao P, Yang P, Chen J, An W. Multifunctional Milk-Derived Small Extracellular Vesicles and Their Biomedical Applications. Pharmaceutics 2023; 15:1418. [PMID: 37242660 PMCID: PMC10223436 DOI: 10.3390/pharmaceutics15051418] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/25/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
In recent years, small extracellular vesicles (sEVs) have been regarded as the next generation of novel delivery systems after lipid nanoparticles because of their advantages and huge prospects in drug delivery. Studies have shown that sEVs are abundant in milk and therefore can be a large and economical source of sEVs. Natural milk-derived small extracellular vesicles (msEVs) have important functions such as immune regulation, anti-bacterial infection, anti-oxidative, etc., and play a beneficial role in human health at multiple levels, including intestinal health, bone/muscle metabolism, and microbiota regulation. In addition, because they can pass the gastrointestinal barrier and have low immunogenicity, good biocompatibility, and stability, msEVs are considered a crucial oral drug delivery vehicle. Moreover, msEVs can be further engineered for targeted delivery to prolong the circulation time or enhance local drug concentrations. However, msEVs separation and purification, complex contents, and quality control hinder their application in drug delivery. This paper provides a comprehensive review of the biogenesis and characteristics, isolation and purification, composition, loading methods, and function of msEVs, based on which their applications in biomedical fields are further explored.
Collapse
Affiliation(s)
- Youxiu Zhong
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xudong Wang
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xian Zhao
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Jiuheng Shen
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xue Wu
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Peifen Gao
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Peng Yang
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Junge Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing 100083, China
| | - Wenlin An
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| |
Collapse
|
185
|
Lee YJ, Park M, Kim HY, Kim JK, Kim WK, Lim SC, Kang KW. Circulating small extracellular vesicles promote proliferation and migration of vascular smooth muscle cells via AXL and MerTK activation. Acta Pharmacol Sin 2023; 44:984-998. [PMID: 36450791 PMCID: PMC10104856 DOI: 10.1038/s41401-022-01029-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022]
Abstract
The proliferation and migration of vascular smooth muscle cells (VSMCs) after vascular injury lead to neointimal hyperplasia, thus aggravating vascular diseases. However, the molecular mechanisms underlying neointima formation are not fully elucidated. Extracellular vesicles (EVs) are mediators of various intercellular communications. The potential of EVs as regulators in cardiovascular diseases has raised significant interest. In the current study we investigated the role of circulating small extracellular vesicles (csEVs), the most abundant EVs (1010 EVs/mL serum) in VSMC functions. csEVs were prepared from bovine, porcine or rat serum. We showed that incubation with csEVs (0.5 × 1010-2 × 1010) dose-dependently enhanced the proliferation and migration of VSMCs via the membrane phosphatidylserine (PS). In rats with ligation of right carotid artery, we demonstrated that application of csEVs in the ligated vessels aggravated neointima formation via interaction of membrane PS with injury. Furthermore, incubation with csEVs markedly enhanced the phosphorylation of AXL and MerTK in VSMCs. Pretreatment with BSM777607 (pan-TAM inhibitor), bemcentinib (AXL inhibitor) or UNC2250 (MerTK inhibitor) blocked csEV-induced proliferation and migration of VSMCs. We revealed that csEV-activated AXL and MerTK shared the downstream signaling pathways of Akt, extracellular signal-regulated kinase (ERK) and focal adhesion kinase (FAK) that mediated the effects of csEVs. We also found that csEVs increased the expression of AXL through activation of transcription factor YAP, which might constitute an AXL-positive feedback loop to amplify the signals. Finally, we demonstrated that dual inhibition of AXL/MerTK by ONO-7475 (0.1 µM) effectively hindered csEV-mediated proliferation and migration of VSMCs in ex vivo mouse aorta injury model. Based on these results, we propose an essential role for csEVs in proliferation and migration of VSMCs and highlight the feasibility of dual AXL/MerTK inhibitors in the treatment of vascular diseases.
Collapse
Affiliation(s)
- Young Joo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Miso Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyun Young Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jin-Ki Kim
- College of Pharmacy, Hanyang University, Gyeonggi-do, 15588, Republic of Korea
| | - Won-Ki Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Sung Chul Lim
- Department of Pathology, Medical school, Chosun University, Gwangju, 61453, Republic of Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
186
|
Xiong L, Wei Y, Jia Q, Chen J, Chen T, Yuan J, Pi C, Liu H, Tang J, Yin S, Zuo Y, Zhang X, Liu F, Yang H, Zhao L. The application of extracellular vesicles in colorectal cancer metastasis and drug resistance: recent advances and trends. J Nanobiotechnology 2023; 21:143. [PMID: 37120534 PMCID: PMC10148416 DOI: 10.1186/s12951-023-01888-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/06/2023] [Indexed: 05/01/2023] Open
Abstract
Colorectal cancer (CRC) has high incidence and mortality rates and is one of the most common cancers of the digestive tract worldwide. Metastasis and drug resistance are the main causes of cancer treatment failure. Studies have recently suggested extracellular vesicles (EVs) as a novel mechanism for intercellular communication. They are vesicular particles, which are secreted and released into biological fluids, such as blood, urine, milk, etc., by a variety of cells and carry numerous biologically active molecules, including proteins, nucleic acids, lipids, metabolites, etc. EVs play a crucial part in the metastasis and drug resistance of CRC by delivering cargo to recipient cells and modulating their behavior. An in-depth exploration of EVs might facilitate a comprehensive understanding of the biological behavior of CRC metastasis and drug resistance, which might provide a basis for developing therapeutic strategies. Therefore, considering the specific biological properties of EVs, researchers have attempted to explore their potential as next-generation delivery systems. On the other hand, EVs have also been demonstrated as biomarkers for the prediction, diagnosis, and presumed prognosis of CRC. This review focuses on the role of EVs in regulating the metastasis and chemoresistance of CRC. Moreover, the clinical applications of EVs are also discussed.
Collapse
Affiliation(s)
- Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, People's Republic of China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, People's Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, People's Republic of China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, People's Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Qiang Jia
- Ethics Committee Office, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jinglin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, People's Republic of China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, People's Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Tao Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, People's Republic of China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, People's Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jiyuan Yuan
- Clinical Trial Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, People's Republic of China
| | - Huiyang Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, People's Republic of China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, People's Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jia Tang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, People's Republic of China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, People's Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Suyu Yin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, People's Republic of China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, People's Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaomei Zhang
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, Institute of Medicinal Chemistry of Chinese Medicine, Chongqing Academy of Chinese Materia Medica, Chongqing, 400065, People's Republic of China
| | - Furong Liu
- Department of Oncology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, China.
| | - Hongru Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Ling Zhao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, People's Republic of China.
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest, Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
187
|
Lopes D, Lopes J, Pereira-Silva M, Peixoto D, Rabiee N, Veiga F, Moradi O, Guo ZH, Wang XD, Conde J, Makvandi P, Paiva-Santos AC. Bioengineered exosomal-membrane-camouflaged abiotic nanocarriers: neurodegenerative diseases, tissue engineering and regenerative medicine. Mil Med Res 2023; 10:19. [PMID: 37101293 PMCID: PMC10134679 DOI: 10.1186/s40779-023-00453-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 04/07/2023] [Indexed: 04/28/2023] Open
Abstract
A bio-inspired strategy has recently been developed for camouflaging nanocarriers with biomembranes, such as natural cell membranes or subcellular structure-derived membranes. This strategy endows cloaked nanomaterials with improved interfacial properties, superior cell targeting, immune evasion potential, and prolonged duration of systemic circulation. Here, we summarize recent advances in the production and application of exosomal membrane-coated nanomaterials. The structure, properties, and manner in which exosomes communicate with cells are first reviewed. This is followed by a discussion of the types of exosomes and their fabrication methods. We then discuss the applications of biomimetic exosomes and membrane-cloaked nanocarriers in tissue engineering, regenerative medicine, imaging, and the treatment of neurodegenerative diseases. Finally, we appraise the current challenges associated with the clinical translation of biomimetic exosomal membrane-surface-engineered nanovehicles and evaluate the future of this technology.
Collapse
Affiliation(s)
- Daniela Lopes
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Joana Lopes
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Diana Peixoto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, 6150, Australia
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Omid Moradi
- Department of Chemistry, Shahr-e-Qods Branch, Islamic Azad University, Tehran, 374-37515, Iran
| | - Zhan-Hu Guo
- Integrated Composites Laboratory (ICL), Department of Mechanical and Construction Engineering, Northumbria University, Newcastle Upon Tyne, NE1 8ST, UK
| | - Xiang-Dong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, 200032, China.
| | - João Conde
- Faculdade de Ciências Médicas, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
- Centre for Toxicogenomics and Human Health, Genetics, Oncology and Human Toxicology, Faculdade de Ciências Médicas, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
| | - Pooyan Makvandi
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal.
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal.
| |
Collapse
|
188
|
Sancho-Albero M, Martín-Pardillos A, Irusta S, Sebastián V, Cebolla VL, Pazo-Cid R, Martín-Duque P, Santamaría J. X-ray Photoelectron Spectroscopy (XPS) Analysis of Nitrogen Environment in Small Extracellular Vesicle Membranes: A Potential Novel Technique with Application for Cancer Screening. Cancers (Basel) 2023; 15:cancers15092479. [PMID: 37173946 PMCID: PMC10177571 DOI: 10.3390/cancers15092479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Small extracellular vesicle (EV) membranes display characteristic protein-lipidic composition features that are related to their cell of origin, providing valuable clues regarding their parental cell composition and real-time state. This could be especially interesting in the case of cancer cell-derived EVs, as their membranes could serve as valuable tools in liquid biopsy applications and to detect changes in the tumor malignancy. X-Ray Photoelectron Spectroscopy (XPS) is a powerful surface analysis technique able to detect every chemical element present, being also sensitive to their chemical environment. Here we explore the use of XPS as a fast technique to characterize EV membrane composition, with possible application in cancer research. Notably, we have focused on the nitrogen environment as an indicator of the relative abundance of pyridine-type bonding, primary, secondary and tertiary amines. Specifically, we have analyzed how tumoral and healthy cells have different nitrogen chemical environments that can indicate the presence or absence of malignancy. In addition, a collection of human serum samples from cancer patients and healthy donors was also analyzed. The differential XPS analysis of EVs collected from patients confirmed that the patterns of amine evolution could be related to markers of cancer disease, opening the possibility of their use as a non-invasive blood biomarker.
Collapse
Affiliation(s)
- María Sancho-Albero
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, 50018 Zaragoza, Spain
- Department of Chemical Engineering and Environmental Technologies, University of Zaragoza, 50018 Zaragoza, Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Ana Martín-Pardillos
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, 50018 Zaragoza, Spain
- Department of Chemical Engineering and Environmental Technologies, University of Zaragoza, 50018 Zaragoza, Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Silvia Irusta
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, 50018 Zaragoza, Spain
- Department of Chemical Engineering and Environmental Technologies, University of Zaragoza, 50018 Zaragoza, Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Víctor Sebastián
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, 50018 Zaragoza, Spain
- Department of Chemical Engineering and Environmental Technologies, University of Zaragoza, 50018 Zaragoza, Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Laboratorio de Miscroscopia Avanzadas, University of Zaragoza, 50018 Zaragoza, Spain
| | | | - Roberto Pazo-Cid
- Medical Oncology Service, Miguel Servet Hospital, 50009 Zaragoza, Spain
| | - Pilar Martín-Duque
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Instituto Aragonés de Ciencias de la Salud, 50009 Zaragoza, Spain
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| | - Jesús Santamaría
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, 50018 Zaragoza, Spain
- Department of Chemical Engineering and Environmental Technologies, University of Zaragoza, 50018 Zaragoza, Spain
- Networking Research Center on Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| |
Collapse
|
189
|
Muraki R, Morita Y, Ida S, Kitajima R, Furuhashi S, Takeda M, Kikuchi H, Hiramatsu Y, Takanashi Y, Hamaya Y, Sugimoto K, Ito J, Kawata K, Kawasaki H, Sato T, Kahyo T, Setou M, Takeuchi H. Phosphatidylcholine in bile-derived small extracellular vesicles as a novel biomarker of cholangiocarcinoma. Cancer Med 2023. [PMID: 37096775 DOI: 10.1002/cam4.5973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/29/2023] [Accepted: 04/08/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Owing to the lack of definite diagnostic modalities, it is challenging to distinguish malignant cases of cholangiocarcinoma (CCA), which often causes biliary tract obstruction, from benign ones. Here, we investigated a novel lipid biomarker of CCA in bile-derived small extracellular vesicles (sEVs) and developed a simple detection method for clinical application. METHODS Bile samples from seven patients with malignant diseases (hilar CCA = 4, distal CCA = 3) and eight patients with benign diseases (gallstones = 6, primary sclerosing cholangitis = 1, autoimmune pancreatitis = 1) were collected through a nasal biliary drainage tube. sEVs were isolated via serial ultracentrifugation and characterized using nanoparticle tracking analysis, transmission electron microscopy, and immunoblotting (with CD9, CD63, CD81, and TSG101). Comprehensive lipidomic analysis was performed using liquid chromatography-tandem mass spectrometry. Using a measurement kit, we further confirmed whether lipid concentrations could be used as a potential CCA marker. RESULTS Lipidomic analysis of bile sEVs in the two groups identified 209 significantly increased lipid species in the malignant group. When focusing on lipid class, phosphatidylcholine (PC) level was 4.98-fold higher in the malignant group than in the benign group (P = 0.037). The receiver operating characteristic (ROC) curve showed a sensitivity of 71.4%, a specificity of 100%, and an area under the curve (AUC) of 0.857 (95% confidence interval [CI]:0.643-1.000). Using a PC assay kit, the ROC curve showed a cutoff value of 16.1 μg/mL, a sensitivity of 71.4%, a specificity of 100%, and an AUC of 0.839 (95% CI: 0.620-1.000). CONCLUSION PC level in sEVs from human bile is a potential diagnostic marker for CCA and can be assessed by a commercially available assay kit.
Collapse
Affiliation(s)
- Ryuta Muraki
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yoshifumi Morita
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shinya Ida
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ryo Kitajima
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Satoru Furuhashi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Makoto Takeda
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hirotoshi Kikuchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yoshihiro Hiramatsu
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Perioperative Functioning Care & Support, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yusuke Takanashi
- First Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasushi Hamaya
- First Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ken Sugimoto
- First Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Jun Ito
- Second Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhito Kawata
- Second Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideya Kawasaki
- Preeminent Medical Photonics Education & Research Center, Institute for NanoSuit Research, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomohito Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoaki Kahyo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroya Takeuchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
190
|
Perpiñá-Clérigues C, Mellado S, Català-Senent JF, Ibáñez F, Costa P, Marcos M, Guerri C, García-García F, Pascual M. Lipidomic landscape of circulating extracellular vesicles isolated from adolescents exposed to ethanol intoxication: a sex difference study. Biol Sex Differ 2023; 14:22. [PMID: 37085905 PMCID: PMC10120207 DOI: 10.1186/s13293-023-00502-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 04/03/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Lipids represent essential components of extracellular vesicles (EVs), playing structural and regulatory functions during EV biogenesis, release, targeting, and cell uptake. Importantly, lipidic dysregulation has been linked to several disorders, including metabolic syndrome, inflammation, and neurological dysfunction. Our recent results demonstrated the involvement of plasma EV microRNAs as possible amplifiers and biomarkers of neuroinflammation and brain damage induced by ethanol intoxication during adolescence. Considering the possible role of plasma EV lipids as regulatory molecules and biomarkers, we evaluated how acute ethanol intoxication differentially affected the lipid composition of plasma EVs in male and female adolescents and explored the participation of the immune response. METHODS Plasma EVs were extracted from humans and wild-type (WT) and Toll-like receptor 4 deficient (TLR4-KO) mice. Preprocessing and exploratory analyses were conducted after the extraction of EV lipids and data acquisition by mass spectrometry. Comparisons between ethanol-intoxicated and control human female and male individuals and ethanol-treated and untreated WT and TLR4-KO female and male mice were used to analyze the differential abundance of lipids. Annotation of lipids into their corresponding classes and a lipid set enrichment analysis were carried out to evaluate biological functions. RESULTS We demonstrated, for the first time, that acute ethanol intoxication induced a higher enrichment of distinct plasma EV lipid species in human female adolescents than in males. We observed a higher content of the PA, LPC, unsaturated FA, and FAHFA lipid classes in females, whereas males showed enrichment in PI. These lipid classes participate in the formation, release, and uptake of EVs and the activation of the immune response. Moreover, we observed changes in EV lipid composition between ethanol-treated WT and TLR4-KO mice (e.g., enrichment of glycerophosphoinositols in ethanol-treated WT males), and the sex-based differences in lipid abundance are more notable in WT mice than in TLR4-KO mice. All data and results generated have been made openly available on a web-based platform ( http://bioinfo.cipf.es/sal ). CONCLUSIONS Our results suggest that binge ethanol drinking in human female adolescents leads to a higher content of plasma EV lipid species associated with EV biogenesis and the propagation of neuroinflammatory responses than in males. In addition, we discovered greater differences in lipid abundance between sexes in WT mice compared to TLR4-KO mice. Our findings also support the potential use of EV-enriched lipids as biomarkers of ethanol-induced neuroinflammation during adolescence.
Collapse
Affiliation(s)
- Carla Perpiñá-Clérigues
- Bioinformatics and Biostatistics Unit, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
| | - Susana Mellado
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain
| | - José F Català-Senent
- Bioinformatics and Biostatistics Unit, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Francesc Ibáñez
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain
| | - Pilar Costa
- Emergency Department, University Hospital of Salamanca-IBSAL, University of Salamanca, 37007, Salamanca, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007, Salamanca, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain
| | - Francisco García-García
- Bioinformatics and Biostatistics Unit, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012, Valencia, Spain.
| | - María Pascual
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain.
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain.
| |
Collapse
|
191
|
Samarpita S, Li X. Leveraging Exosomes as the Next-Generation Bio-Shuttles: The Next Biggest Approach against Th17 Cell Catastrophe. Int J Mol Sci 2023; 24:ijms24087647. [PMID: 37108809 PMCID: PMC10142210 DOI: 10.3390/ijms24087647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
In recent years, the launch of clinical-grade exosomes is rising expeditiously, as they represent a new powerful approach for the delivery of advanced therapies and for diagnostic purposes for various diseases. Exosomes are membrane-bound extracellular vesicles that can act as biological messengers between cells, in the context of health and disease. In comparison to several lab-based drug carriers, exosome exhibits high stability, accommodates diverse cargo loads, elicits low immunogenicity and toxicity, and therefore manifests tremendous perspectives in the development of therapeutics. The efforts made to spur exosomes in drugging the untreatable targets are encouraging. Currently, T helper (Th) 17 cells are considered the most prominent factor in the establishment of autoimmunity and several genetic disorders. Current reports have indicated the importance of targeting the development of Th17 cells and the secretion of its paracrine molecule, interleukin (IL)-17. However, the present-day targeted approaches exhibit drawbacks, such as high cost of production, rapid transformation, poor bioavailability, and importantly, causing opportunistic infections that ultimately hamper their clinical applications. To overcome this hurdle, the potential use of exosomes as vectors seem to be a promising approach for Th17 cell-targeted therapies. With this standpoint, this review discusses this new concept by providing a snapshot of exosome biogenesis, summarizes the current clinical trials of exosomes in several diseases, analyzes the prospect of exosomes as an established drug carrier and delineates the present challenges, with an emphasis on their practical applications in targeting Th17 cells in diseases. We further decode the possible future scope of exosome bioengineering for targeted drug delivery against Th17 cells and its catastrophe.
Collapse
Affiliation(s)
- Snigdha Samarpita
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
192
|
Yao X, Liao B, Chen F, Liu L, Wu K, Hao Y, Li Y, Wang Y, Fan R, Yin J, Liu L, Guo Y. Comparison of proteomic landscape of extracellular vesicles in pleural effusions isolated by three strategies. Front Bioeng Biotechnol 2023; 11:1108952. [PMID: 37122867 PMCID: PMC10130534 DOI: 10.3389/fbioe.2023.1108952] [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: 11/26/2022] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Extracellular vesicles (EVs) derived from pleural effusion (PE) is emerging as disease biomarkers. However, the methods for isolation of EVs from PE (pEVs) were rarely studied. In our study, three methods for isolating pEVs of lung cancer patients were compared, including ultracentrifugation (UC), a combination of UC and size exclusion chromatography (UC-SEC) and a combination of UC and density gradient ultracentrifugation (UC-DGU). The subpopulation of pEVs was identified by nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM), Western blotting (WB) and nano-flow cytometry (nFCM). Additionally, the proteomic landscape of pEVs was analyzed by Label-free proteomics. The results showed that, compared with UC and UC-DGU, the UC-SEC method separated pEVs with the highest purity. In the proteomic analysis, on average, 1595 proteins were identified in the pEVs isolated by UC-SEC, much more than pEVs isolated by UC (1222) or UC-DGU (807). Furthermore, approximately 90% of identified proteins in each method were found in the EVs public database ExoCarta. Consistent with this, GO annotation indicated that the core proteins identified in each method were mainly enriched in "extracellular exosome." Many of the top 100 proteins with high expression in each method were suggested as protein markers to validate the presence of EVs in the MISEV2018 guidelines. In addition, combined with lung tissue-specific proteins and vesicular membrane proteins, we screened out and validated several novel protein markers (CD11C, HLA DPA1 and HLA DRB1), which were enriched in pEVs rather than in plasma EVs. In conclusion, our study shows that the method of UC-SEC could significantly improve the purity of EVs and the performance of mass spectrometry-based proteomic profiling in analyzing pEVs. The exosomal proteins CD11C, HLA DPA1 and HLA DRB1 may act as potential markers of pEVs. The proteomic analysis of pEVs provides important information and new ideas for studying diseases complicated with PE.
Collapse
Affiliation(s)
- Xue Yao
- School of Medicine, Southwest Jiaotong University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Baixue Liao
- School of Medicine, Southwest Jiaotong University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Feng Chen
- Department of Respiratory, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Lüye Liu
- Medical Research Center, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Kaiwen Wu
- School of Medicine, Southwest Jiaotong University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Yaying Hao
- Medical Research Center, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Yanping Li
- Department of Respiratory, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Yuebin Wang
- Department of Respiratory, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Ruiling Fan
- School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Jun Yin
- Department of Respiratory, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Lei Liu
- Medical Research Center, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Yuanbiao Guo
- Medical Research Center, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
193
|
Kang M, Hisey C, Tsai B, Nursalim Y, Blenkiron C, Chamley LW. Placental Extracellular Vesicles Can Be Loaded with Plasmid DNA. Mol Pharm 2023; 20:1898-1913. [PMID: 36919912 PMCID: PMC11407900 DOI: 10.1021/acs.molpharmaceut.2c00533] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Recently, extracellular vesicles (EVs) have garnered considerable interest as potential vehicles for drug delivery, including gene therapy. Although EVs from diverse sources have been investigated, current techniques used in the field for EV generation limit large-scale EV production. The placenta is essentially a tissue transplant and has unique properties that allow it to avoid the maternal immune system making it likely that placental EVs will not generate inflammatory responses and will avoid clearance by the immune system. We propose that placental EVs produced from explant cultures are an efficient method to produce considerable quantities of EVs that would be safe to administer, and we hypothesize that placental EVs can be loaded with large exogenous plasmids. To this end, we trialed three strategies to load plasmid DNA into placental EVs, including loading via electroporation of placental tissue prior to EV isolation and loading directly into placental EVs via electroporation or direct incubation of the EVs in plasmid solution. We report that the placenta releases vast quantities of EVs compared to placental cells in monolayer cultures. We show successful loading of plasmid DNA into both large- and small-EVs following both exogenous loading strategies with more plasmid encapsulated in large-EVs. Importantly, direct incubation did not alter EV size nor quantity. Further, we showed that the loading efficiency into EVs was dependent on the exogenous plasmid DNA dose and the DNA size. These results provide realistic estimates of plasmid loading capacity into placental EVs using current technologies and showcase the potential of placental EVs as DNA delivery vehicles.
Collapse
Affiliation(s)
- Matthew Kang
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
| | - Colin Hisey
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
- Department of biomedical Engineering, The Ohio State University, Columbus, Ohio, 43210 United States
| | - Bridget Tsai
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
| | - Yohanes Nursalim
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
| | - Cherie Blenkiron
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
- Auckland Cancer Society Research Center (ACSRC), University of Auckland, Auckland, 1023 New Zealand
- Molecular Medicine and Pathology, University of Auckland, Auckland, 1023 New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
| |
Collapse
|
194
|
Cortes-Galvez D, Dangerfield JA, Metzner C. Extracellular Vesicles and Their Membranes: Exosomes vs. Virus-Related Particles. MEMBRANES 2023; 13:397. [PMID: 37103824 PMCID: PMC10146078 DOI: 10.3390/membranes13040397] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Cells produce nanosized lipid membrane-enclosed vesicles which play important roles in intercellular communication. Interestingly, a certain type of extracellular vesicle, termed exosomes, share physical, chemical, and biological properties with enveloped virus particles. To date, most similarities have been discovered with lentiviral particles, however, other virus species also frequently interact with exosomes. In this review, we will take a closer look at the similarities and differences between exosomes and enveloped viral particles, with a focus on events taking place at the vesicle or virus membrane. Since these structures present an area with an opportunity for interaction with target cells, this is relevant for basic biology as well as any potential research or medical applications.
Collapse
Affiliation(s)
- Daniela Cortes-Galvez
- AG Histology and Embryology, Institute of Morphology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | | | | |
Collapse
|
195
|
Lau NCH, Yam JWP. From Exosome Biogenesis to Absorption: Key Takeaways for Cancer Research. Cancers (Basel) 2023; 15:cancers15071992. [PMID: 37046653 PMCID: PMC10093369 DOI: 10.3390/cancers15071992] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 03/29/2023] Open
Abstract
Exosomes are mediators of intercellular communication in normal physiology and diseases. While many studies have emerged on the function of exosomal cargoes, questions remain regarding the origin of these exosomes. The packaging and secretion of exosomes in different contexts modify exosomal composition, which may in turn impact delivery, uptake and cargo function in recipient cells. A mechanistic understanding of exosome biology is therefore crucial to investigating exosomal function in complex biological systems and to the development of novel therapeutic approaches. Here, we outline the steps in exosome biogenesis, including endosome formation, MVB formation, cargo sorting and extracellular release, as well as exosome absorption, including targeting, interaction with recipient cells and the fate of internalized exosomes. In addition to providing a framework of exosome dynamics, we summarize current evidence on major pathways and regulatory mechanisms. We also highlight the various mechanisms observed in cancer and point out directions to improve study design in exosome biology. Further research is needed to illuminate the relationship between exosome biogenesis and function, which will aid the development of translational applications.
Collapse
Affiliation(s)
- Nicolas Cheuk Hang Lau
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Judy Wai Ping Yam
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
- Correspondence: ; Tel.: +852-22552681
| |
Collapse
|
196
|
Lu X, Li Y, Li Y, Zhang X, Shi J, Feng H, Gao Y, Yu Z. Advances of multi-omics applications in hepatic precancerous lesions and hepatocellular carcinoma: The role of extracellular vesicles. Front Mol Biosci 2023; 10:1114594. [PMID: 37006626 PMCID: PMC10060991 DOI: 10.3389/fmolb.2023.1114594] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Due to the lack of distinct early symptoms and specific biomarkers, most patients with hepatocellular carcinoma (HCC) are usually diagnosed at advanced stages, rendering the treatment ineffective and useless. Therefore, recognition of the malady at precancerous lesions and early stages is particularly important for improving patient outcomes. The interest in extracellular vesicles (EVs) has been growing in recent years with the accumulating knowledge of their multiple cargoes and related multipotent roles in the modulation of immune response and tumor progression. By virtue of the rapid advancement of high-throughput techniques, multiple omics, including genomics/transcriptomics, proteomics, and metabolomics/lipidomics, have been widely integrated to analyze the role of EVs. Comprehensive analysis of multi-omics data will provide useful insights for discovery of new biomarkers and identification of therapeutic targets. Here, we review the attainment of multi-omics analysis to the finding of the potential role of EVs in early diagnosis and the immunotherapy in HCC.
Collapse
Affiliation(s)
- Xiaona Lu
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuyao Li
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Li
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuemei Zhang
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jia Shi
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hai Feng
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Hai Feng, ; Yueqiu Gao, ; Zhuo Yu,
| | - Yueqiu Gao
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Hai Feng, ; Yueqiu Gao, ; Zhuo Yu,
| | - Zhuo Yu
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Hai Feng, ; Yueqiu Gao, ; Zhuo Yu,
| |
Collapse
|
197
|
Mustonen AM, Lehmonen N, Paakkonen T, Raekallio M, Käkelä R, Niemelä T, Mykkänen A, Sihvo SP, Nieminen P. Equine osteoarthritis modifies fatty acid signatures in synovial fluid and its extracellular vesicles. Arthritis Res Ther 2023; 25:39. [PMID: 36895037 PMCID: PMC9996872 DOI: 10.1186/s13075-023-02998-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/27/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Individual fatty acids (FAs) and their derivatives (lipid mediators) with pro-inflammatory or dual anti-inflammatory and pro-resolving properties have potential to influence the health of joint tissues. Osteoarthritis (OA) is an age-associated chronic joint disease that can be featured with altered FA composition in the synovial fluid (SF) of human patients. The counts and cargo of extracellular vesicles (EVs), membrane-bound particles released by synovial joint cells and transporting bioactive lipids, can also be modified by OA. The detailed FA signatures of SF and its EVs have remained unexplored in the horse - a well-recognized veterinary model for OA research. METHODS The aim of the present study was to compare the FA profiles in equine SF and its ultracentrifuged EV fraction between control, contralateral, and OA metacarpophalangeal joints (n = 8/group). The FA profiles of total lipids were determined by gas chromatography and the data compared with univariate and multivariate analyses. RESULTS The data revealed distinct FA profiles in SF and its EV-enriched pellet that were modified by naturally occurring equine OA. Regarding SFs, linoleic acid (generalized linear model, p = 0.0006), myristic acid (p = 0.003), palmitoleic acid (p < 0.0005), and n-3/n-6 polyunsaturated FA ratio (p < 0.0005) were among the important variables that separated OA from control samples. In EV-enriched pellets, saturated FAs palmitic acid (p = 0.020), stearic acid (p = 0.002), and behenic acid (p = 0.003) indicated OA. The observed FA modifications are potentially detrimental and could contribute to inflammatory processes and cartilage degradation in OA. CONCLUSIONS Equine OA joints can be distinguished from normal joints based on their FA signatures in SF and its EV-enriched pellet. Clarifying the roles of SF and EV FA compositions in the pathogenesis of OA and their potential as joint disease biomarkers and therapeutic targets warrants future studies.
Collapse
Affiliation(s)
- Anne-Mari Mustonen
- grid.9668.10000 0001 0726 2490Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
- grid.9668.10000 0001 0726 2490Department of Environmental and Biological Sciences, Faculty of Science, Forestry and Technology, University of Eastern Finland, P.O. Box 111, FI-80101 Joensuu, Finland
| | - Nina Lehmonen
- grid.7737.40000 0004 0410 2071Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - Tommi Paakkonen
- grid.9668.10000 0001 0726 2490Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Marja Raekallio
- grid.7737.40000 0004 0410 2071Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - Reijo Käkelä
- grid.7737.40000 0004 0410 2071Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, P.O. Box 65, FI-00014 Helsinki, Finland
- grid.484023.9Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, P.O. Box 65, FI-00014 Helsinki, Finland
| | - Tytti Niemelä
- grid.7737.40000 0004 0410 2071Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - Anna Mykkänen
- grid.7737.40000 0004 0410 2071Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - Sanna P. Sihvo
- grid.7737.40000 0004 0410 2071Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, P.O. Box 65, FI-00014 Helsinki, Finland
- grid.484023.9Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, P.O. Box 65, FI-00014 Helsinki, Finland
| | - Petteri Nieminen
- grid.9668.10000 0001 0726 2490Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
198
|
He S, Ding L, Yuan H, Zhao G, Yang X, Wu Y. A review of sensors for classification and subtype discrimination of cancer: Insights into circulating tumor cells and tumor-derived extracellular vesicles. Anal Chim Acta 2023; 1244:340703. [PMID: 36737145 DOI: 10.1016/j.aca.2022.340703] [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: 07/23/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022]
Abstract
Liquid biopsy can reflect the state of tumors in vivo non-invasively, thus providing a strong basis for the early diagnosis, individualized treatment monitoring and prognosis of tumors. Circulating tumor cells (CTCs) and tumor-derived extracellular vesicles (tdEVs) contain information-rich components, such as nucleic acids and proteins, and they are essential markers for liquid biopsies. Their capture and analysis are of great importance for the study of disease occurrence and development and, consequently, have been the subject of many reviews. However, both CTCs and tdEVs carry the biological characteristics of their original tissue, and few reviews have focused on their function in the staging and classification of cancer. In this review, we focus on state-of-the-art sensors based on the simultaneous detection of multiple biomarkers within CTCs and tdEVs, with clinical applications centered on cancer classification and subtyping. We also provide a thorough discussion of the current challenges and prospects for novel sensors with the ultimate goal of cancer classification and staging. It is hoped that these most advanced technologies will bring new insights into the clinical practice of cancer screening and diagnosis.
Collapse
Affiliation(s)
- Sitian He
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Lihua Ding
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Huijie Yuan
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Gaofeng Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China.
| | - Xiaonan Yang
- School of Information Engineering, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
199
|
Zha J, Pan Y, Liu X, Zhu H, Liu Y, Zeng W. Exosomes from hypoxia-pretreated adipose-derived stem cells attenuate ultraviolet light-induced skin injury via delivery of circ-Ash1l. PHOTODERMATOLOGY, PHOTOIMMUNOLOGY & PHOTOMEDICINE 2023; 39:107-115. [PMID: 36582030 DOI: 10.1111/phpp.12857] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/05/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND An increasing number of studies have reported that exosomes from adipose-derived stem cells (ADSCs) have antioxidant and anti-inflammatory properties. In the present study, we aimed at elucidating the potential therapeutic mechanism underlying ADSC exosomes in ultraviolet B-light (UVB)-induced skin injury. METHODS We isolated the exosomes from ADSCs and hypoxia-pretreated ADSCs. High-throughput sequencing was applied to identify differential circRNA expression. Then, a UV-induced murine skin injury model was constructed and the therapeutic effect of exosomes was determined using immunofluorescence and ELISA. The regulatory mechanism was demonstrated using luciferase reporter analysis and an in vitro experiment. RESULTS Exosomes from hypoxia-pretreated ADSCs inhibited UVB light-induced vascular injury by reversing ROS and inflammatory factor expression. High-throughput sequencing showed that exosomes from hypoxia-pretreated ADSCs (HExo) improved UV-induced skin damage via delivery of circ-Ash1l. Downregulation of circ-Ash1l inhibited the therapeutic effect of HExo on UV-induced skin damage. It was further shown that GPX4 and miR-700-5p were circ-Ash1l downstream targets. MiR-700-5p overexpression or GPX4 downregulation inhibited the circ-Ash1l protective effects of UV-induced endothelial progenitor cell (EPC) damage. CONCLUSION Thus, exosomes from hypoxia-pretreated ADSCs attenuated UV light-induced skin injury via circ-Ash1l delivery and ferroptosis inhibition.
Collapse
Affiliation(s)
- Jindong Zha
- Department of Dermatology, Northwest Hospital, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Cosmetic Dermatology, Mylike Cosmetology Hospital of Yunnan, Kunming, China
| | - Yanbing Pan
- Department of Cosmetic Dermatology, Mylike Cosmetology Hospital of Yunnan, Kunming, China
| | - Xiufeng Liu
- Department of Cosmetic Dermatology, Mylike Cosmetology Hospital of Yunnan, Kunming, China
| | - Hong Zhu
- Department of Cosmetic Dermatology, Mylike Cosmetology Hospital of Yunnan, Kunming, China
| | - Yanting Liu
- Department of Dermatology, Northwest Hospital, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weihui Zeng
- Department of Dermatology, Northwest Hospital, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
200
|
Blandin A, Dugail I, Hilairet G, Ponnaiah M, Ghesquière V, Froger J, Ducheix S, Fizanne L, Boursier J, Cariou B, Lhomme M, Le Lay S. Lipidomic analysis of adipose-derived extracellular vesicles reveals specific EV lipid sorting informative of the obesity metabolic state. Cell Rep 2023; 42:112169. [PMID: 36862553 DOI: 10.1016/j.celrep.2023.112169] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/19/2023] [Accepted: 02/10/2023] [Indexed: 03/03/2023] Open
Abstract
Adipose extracellular vesicles (AdEVs) transport lipids that could participate in the development of obesity-related metabolic dysfunctions. This study aims to define mouse AdEV lipid signature by a targeted LC-MS/MS approach in either healthy or obesity context. Distinct clustering of AdEV and visceral adipose tissue (VAT) lipidomes by principal component analysis reveals specific AdEV lipid sorting when compared with secreting VAT. Comprehensive analysis identifies enrichment of ceramides, sphingomyelins, and phosphatidylglycerols species in AdEVs compared with source VAT whose lipid content closely relates to the obesity status and is influenced by the diet. Obesity moreover impacts AdEV lipidome, mirroring lipid alterations retrieved in plasma and VAT. Overall, our study identifies specific lipid fingerprints for plasma, VAT, and AdEVs that are informative of the metabolic status. Lipid species enriched in AdEVs in the obesity context may constitute biomarker candidates or mediators of the obesity-associated metabolic dysfunctions.
Collapse
Affiliation(s)
- Alexia Blandin
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; Université d'Angers, SFR ICAT, F-49 000 Angers, France
| | - Isabelle Dugail
- UMRS 1269 INSERM/Sorbonne University, Nutriomics, 75013 Paris, France
| | | | - Maharajah Ponnaiah
- IHU ICAN (ICAN Omics and ICAN I/O), Foundation for Innovation in Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Valentine Ghesquière
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; Université d'Angers, SFR ICAT, F-49 000 Angers, France
| | - Josy Froger
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; Université d'Angers, SFR ICAT, F-49 000 Angers, France
| | - Simon Ducheix
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| | - Lionel Fizanne
- HIFIH Laboratory UPRES EA3859, SFR 4208, Angers University, Angers, France
| | - Jérôme Boursier
- HIFIH Laboratory UPRES EA3859, SFR 4208, Angers University, Angers, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| | - Marie Lhomme
- IHU ICAN (ICAN Omics and ICAN I/O), Foundation for Innovation in Cardiometabolism and Nutrition, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Soazig Le Lay
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; Université d'Angers, SFR ICAT, F-49 000 Angers, France.
| |
Collapse
|