1
|
Castillo-Galán S, Parra V, Cuenca J. Unraveling the pathogenesis of viral-induced pulmonary arterial hypertension: Possible new therapeutic avenues with mesenchymal stromal cells and their derivatives. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167519. [PMID: 39332781 DOI: 10.1016/j.bbadis.2024.167519] [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: 05/29/2024] [Revised: 08/16/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Pulmonary hypertension (PH) is a severe condition characterized by elevated pressure in the pulmonary artery, where metabolic and mitochondrial dysfunction may contribute to its progression. Within the PH spectrum, pulmonary arterial hypertension (PAH) stands out with its primary pulmonary vasculopathy. PAH's prevalence varies from 0.4 to 1.4 per 100,000 individuals and is associated with diverse conditions, including viral infections such as HIV. Notably, recent observations highlight an increased occurrence of PAH among COVID-19 patients, even in the absence of pre-existing cardiopulmonary disorders. While current treatments offer partial relief, there's a pressing need for innovative therapeutic strategies, among which mesenchymal stromal cells (MSCs) and their derivatives hold promise. This review critically evaluates recent investigations into viral-induced PAH, encompassing pathogens like human immunodeficiency virus, herpesvirus, Cytomegalovirus, Hepatitis B and C viruses, SARS-CoV-2, and Human endogenous retrovirus K (HERKV), with a specific emphasis on mitochondrial dysfunction. Furthermore, we explore the underlying rationale driving novel therapeutic modalities, including MSCs, extracellular vesicles, and mitochondrial interventions, within the framework of PAH management.
Collapse
Affiliation(s)
- Sebastián Castillo-Galán
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| | - Valentina Parra
- Laboratory of Differentiation and Cell Metabolism (D&M), Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; SYSTEMIX Center for Systems Biology, O'Higgins University, Rancagua, Chile
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile; Cells for Cells, Santiago, Chile.
| |
Collapse
|
2
|
Zhand S, Goss DM, Cheng YY, Warkiani ME. Recent Advances in Microfluidics for Nucleic Acid Analysis of Small Extracellular Vesicles in Cancer. Adv Healthc Mater 2024:e2401295. [PMID: 39707658 DOI: 10.1002/adhm.202401295] [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: 04/08/2024] [Revised: 11/28/2024] [Indexed: 12/23/2024]
Abstract
Small extracellular vesicles (sEVs) are membranous vesicles released from cellular structures through plasma membrane budding. These vesicles contain cellular components such as proteins, lipids, mRNAs, microRNAs, long-noncoding RNA, circular RNA, and double-stranded DNA, originating from the cells they are shed from. Ranging in size from ≈25 to 300 nm and play critical roles in facilitating cell-to-cell communication by transporting signaling molecules. The discovery of sEVs in bodily fluids and their involvement in intercellular communication has revolutionized the fields of diagnosis, prognosis, and treatment, particularly in diseases like cancer. Conventional methods for isolating and analyzing sEVs, particularly their nucleic acid content face challenges including high costs, low purity, time-consuming processes, limited standardization, and inconsistent yield. The development of microfluidic devices, enables improved precision in sorting, isolating, and molecular-level separation using small sample volumes, and offers significant potential for the enhanced detection and monitoring of sEVs associated with cancer. These advanced techniques hold great promise for creating next-generation diagnostic and prognostic tools given their possibility of being cost-effective, simple to operate, etc. This comprehensive review explores the current state of research on microfluidic devices for the detection of sEV-derived nucleic acids as biomarkers and their translation into practical point-of-care and clinical applications.
Collapse
Affiliation(s)
- Sareh Zhand
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Dale Mark Goss
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute of Molecular Theranostics, Sechenov First Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
3
|
Karabay AZ, Ozkan T, Karadag Gurel A, Koc A, Hekmatshoar Y, Sunguroglu A, Aktan F, Buyukbingöl Z. Identification of exosomal microRNAs and related hub genes associated with imatinib resistance in chronic myeloid leukemia. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9701-9721. [PMID: 38916832 PMCID: PMC11582232 DOI: 10.1007/s00210-024-03198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024]
Abstract
Chemotherapy resistance is a major obstacle in cancer therapy, and identifying novel druggable targets to reverse this phenomenon is essential. The exosome-mediated transmittance of drug resistance has been shown in various cancer models including ovarian and prostate cancer models. In this study, we aimed to investigate the role of exosomal miRNA transfer in chronic myeloid leukemia drug resistance. For this purpose, firstly exosomes were isolated from imatinib sensitive (K562S) and resistant (K562R) chronic myeloid leukemia (CML) cells and named as Sexo and Rexo, respectively. Then, miRNA microarray was used to compare miRNA profiles of K562S, K562R, Sexo, Rexo, and Rexo-treated K562S cells. According to our results, miR-125b-5p and miR-99a-5p exhibited increased expression in resistant cells, their exosomes, and Rexo-treated sensitive cells compared to their sensitive counterparts. On the other hand, miR-210-3p and miR-193b-3p were determined to be the two miRNAs which exhibited decreased expression profile in resistant cells and their exosomes compared to their sensitive counterparts. Gene targets, signaling pathways, and enrichment analysis were performed for these miRNAs by TargetScan, KEGG, and DAVID. Potential interactions between gene candidates at the protein level were analyzed via STRING and Cytoscape software. Our findings revealed CCR5, GRK2, EDN1, ARRB1, P2RY2, LAMC2, PAK3, PAK4, and GIT2 as novel gene targets that may play roles in exosomal imatinib resistance transfer as well as mTOR, STAT3, MCL1, LAMC1, and KRAS which are already linked to imatinib resistance. MDR1 mRNA exhibited higher expression in Rexo compared to Sexo as well as in K562S cells treated with Rexo compared to K562S cells which may suggest exosomal transfer of MDR1 mRNA.
Collapse
Affiliation(s)
- Arzu Zeynep Karabay
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey.
| | - Tulin Ozkan
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey.
| | - Aynur Karadag Gurel
- Department of Medical Biology, Faculty of Medicine, Usak University, Usak, Turkey.
| | - Asli Koc
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Yalda Hekmatshoar
- Department of Medical Biology, Faculty of Medicine, Altinbas University, Istanbul, Turkey
| | - Asuman Sunguroglu
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Fugen Aktan
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Zeliha Buyukbingöl
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
4
|
Liu H, Ouyang Z, Li S. Advances of M1 macrophages-derived extracellular vesicles in tumor therapy. Biomed Pharmacother 2024; 181:117735. [PMID: 39644871 DOI: 10.1016/j.biopha.2024.117735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024] Open
Abstract
Extracellular vesicles derived from classically activated M1 macrophages (M1 EVs) have shown great potential in both tumor treatment and drug delivery. M1 EVs inherit specific biological messengers from their parent cells and possess the capability to activate immune cells residing in close or distant tumor tissues for antitumor therapy. Moreover, M1 EVs are commonly used as an attractive drug delivery system due to their tumor-targeting ability, biocompatibility, and non-toxic. They can effectively encapsulate various therapeutic cargoes through specific methods such as electroporation, co-incubation, sonication, extrusion, transfection, or click chemistry reaction. In this review, we provide a comprehensive summary of the advancements in M1 EVs for tumor therapy, discussing their application prospects and existing problems.
Collapse
Affiliation(s)
- Houli Liu
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui Province 230012, China.
| | - Zhaorong Ouyang
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui Province 230012, China
| | - Siyu Li
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui Province 230012, China
| |
Collapse
|
5
|
Lee SY, Klingeborn M, Bulte JWM, Chiu DT, Chopp M, Cutler CW, Das S, Egwuagu CE, Fowler CD, Hamm‐Alvarez SF, Lee H, Liu Y, Mead B, Moore TL, Ravindran S, Shetty AK, Skog J, Witwer KW, Djalilian AR, Weaver AM. A perspective from the National Eye Institute Extracellular Vesicle Workshop: Gaps, needs, and opportunities for studies of extracellular vesicles in vision research. J Extracell Vesicles 2024; 13:e70023. [PMID: 39665315 PMCID: PMC11635481 DOI: 10.1002/jev2.70023] [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: 07/01/2024] [Revised: 11/20/2024] [Accepted: 11/24/2024] [Indexed: 12/13/2024] Open
Abstract
With an evolving understanding and new discoveries in extracellular vesicle (EV) biology and their implications in health and disease, the significant diagnostic and therapeutic potential of EVs for vision research has gained recognition. In 2021, the National Eye Institute (NEI) unveiled its Strategic Plan titled 'Vision for the Future (2021-2025),' which listed EV research as a priority within the domain of Regenerative Medicine, a pivotal area outlined in the Plan. In alignment with this prioritization, NEI organized a workshop inviting twenty experts from within and beyond the visual system. The workshop aimed to review current knowledge in EV research and explore gaps, needs and opportunities for EV research in the eye, including EV biology and applications of EVs in diagnosis, therapy and prognosis within the visual system. This perspective encapsulates the workshop's deliberations, highlighting the current landscape and potential implications of EV research in advancing eye health and addressing visual diseases.
Collapse
Affiliation(s)
- Sun Young Lee
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Jeff W. M. Bulte
- Department of Radiology and Radiological Sciences, School of MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Daniel T. Chiu
- Department of Chemistry and BioengineeringUniversity of WashingtonSeattleWashingtonUSA
| | - Michael Chopp
- Department of NeurologyHenry Ford HealthDetroitMichiganUSA
| | | | - Saumya Das
- Cardiovascular Research CenterMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Charles E. Egwuagu
- Molecular Immunology Section, Laboratory of Immunology, National Eye InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Christie D. Fowler
- Department of Neurobiology and BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Sarah F. Hamm‐Alvarez
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Hakho Lee
- Center for System BiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Ben Mead
- School of Optometry and Vision SciencesCardiff UniversityCardiffUK
| | - Tara L. Moore
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Sriram Ravindran
- Department of Oral Biology, College of DentistryUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and GeneticsTexas A&M University School of MedicineCollege StationTexasUSA
| | - Johan Skog
- Exosome Diagnostics, a Bio‐Techne BrandWalthamMassachusettsUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Ali R. Djalilian
- Department of Ophthalmology and Visual SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Alissa M. Weaver
- Department of Cell and Developmental BiologyVanderbilt University School of MedicineNashvilleTennesseeUSA
| |
Collapse
|
6
|
Yang Y, Wu T, Wang Y, Luo D, Zhao Z, Sun H, Zhang M, Zhang B, Han B. Hypoxic tumour-derived exosomal miR-1290 exacerbates the suppression of CD8+ T cells by promoting M2 macrophage polarization. Immunology 2024; 173:672-688. [PMID: 39183579 DOI: 10.1111/imm.13853] [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: 06/26/2023] [Accepted: 07/30/2024] [Indexed: 08/27/2024] Open
Abstract
Hypoxia plays an important role in the metastasis of hepatocellular carcinoma (HCC). Exosomes have been widely studied as mediators of communication between tumours and immune cells. However, the specific mechanism by which hypoxic HCC cell-derived exosomes suppress antitumor immunity is unclear. Hypoxia scores were determined for The Cancer Genome-Liver Hepatocellular Carcinoma (TCGA-LIHC) dataset patients, and HCC patients in the hyperhypoxic group had a higher degree of M2 macrophage infiltration. Patients in the M2 high-invasion group had a lower probability of survival than those in the low-invasion group. In vivo and in vitro experiments demonstrated that exosomes secreted by hypoxic HCC cells promote M2 macrophage polarization. This polarization induces apoptosis in CD8+ T cells. Additionally, it encourages epithelial-mesenchymal transition (EMT), which increases HCC migration. Exosomal miRNA sequencing revealed that miR-1290 was highly expressed in exosomes secreted by hypoxic HCC cells. Mechanistically, miR-1290 in macrophages inhibited Akt2 while upregulating PD-L1 to promote M2 polarization, induce apoptosis in CD8+ T cells, and enhance EMT in HCC. Animal studies found that the miR-1290 antagomir in combination with the immune checkpoint inhibitor produced better antitumor effects than the monotherapies. In conclusion, the secretion of exosome-derived miR-1290 from HCC cells in a hypoxic environment supported immune escape by HCC cells by promoting M2 macrophage polarization to induce apoptosis in CD8+ T cells and enhance EMT that promoted HCC metastasis. Therefore, miR-1290 is an important molecule in antitumor immunity in HCC, and inhibition of miR-1290 could provide a novel immunotherapeutic approach for HCC treatment.
Collapse
Affiliation(s)
- Yeni Yang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tiansong Wu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Youpeng Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dingan Luo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ziyin Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongfa Sun
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mao Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Zhang
- Department of Transplantation, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bing Han
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
7
|
Yang M, Zhou J, Lu L, Deng D, Huang J, Tang Z, Shi X, Lo P, Lovell JF, Zheng Y, Jin H. Tumor cell membrane-based vaccines: A potential boost for cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230171. [PMID: 39713208 PMCID: PMC11655317 DOI: 10.1002/exp.20230171] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/08/2024] [Indexed: 12/24/2024]
Abstract
Because therapeutic cancer vaccines can, in theory, eliminate tumor cells specifically with relatively low toxicity, they have long been considered for application in repressing cancer progression. Traditional cancer vaccines containing a single or a few discrete tumor epitopes have failed in the clinic, possibly due to challenges in epitope selection, target downregulation, cancer cell heterogeneity, tumor microenvironment immunosuppression, or a lack of vaccine immunogenicity. Whole cancer cell or cancer membrane vaccines, which provide a rich source of antigens, are emerging as viable alternatives. Autologous and allogenic cellular cancer vaccines have been evaluated as clinical treatments. Tumor cell membranes (TCMs) are an intriguing antigen source, as they provide membrane-accessible targets and, at the same time, serve as integrated carriers of vaccine adjuvants and other therapeutic agents. This review provides a summary of the properties and technologies for TCM cancer vaccines. Characteristics, categories, mechanisms, and preparation methods are discussed, as are the demonstrable additional benefits derived from combining TCM vaccines with chemotherapy, sonodynamic therapy, phototherapy, and oncolytic viruses. Further research in chemistry, biomedicine, cancer immunology, and bioinformatics to address current drawbacks could facilitate the clinical adoption of TCM vaccines.
Collapse
Affiliation(s)
- Muyang Yang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Jie Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Liseng Lu
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Deqiang Deng
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Zijian Tang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Xiujuan Shi
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Pui‐Chi Lo
- Department of Biomedical SciencesCity University of Hong KongKowloonHong KongChina
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNew YorkUSA
| | - Yongfa Zheng
- Department of OncologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Honglin Jin
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| |
Collapse
|
8
|
Oh C, Mazan-Mamczarz K, Gorospe M, Noh JH, Kim KM. Impact of UPF2 on the levels of CD81 on extracellular vesicles. Front Cell Dev Biol 2024; 12:1469080. [PMID: 39655046 PMCID: PMC11625909 DOI: 10.3389/fcell.2024.1469080] [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: 07/23/2024] [Accepted: 10/29/2024] [Indexed: 12/12/2024] Open
Abstract
Extracellular vesicles (EVs) are involved in cell-to-cell communication. Following uptake, EV cargo molecules, including DNA, RNA, lipids, and proteins, influence gene expression and molecular signaling in recipient cells. Although various studies have identified disease-specific EV molecules, further research into their biogenesis and secretion mechanisms is needed for clinical application. Here, we investigated the role of UPF2 in regulating the biogenesis and components of EVs. Notably, UPF2 promoted the expression of CD81, a membrane protein marker of EVs, as UPF2 silencing decreased CD81 levels in EVs, both inside the cell and secreted. In contrast, the expression levels of CD63 increased, without altering the size or numbers of EVs. In addition, reducing UPF2 levels did not affect the total number of EVs but lowered production of CD81-positive EVs and reduced the efficiency of uptake by recipient cells. Collectively, our findings uncover a novel function for UPF2 in regulating the production of CD81 and changing EV properties.
Collapse
Affiliation(s)
- Chaehwan Oh
- Department of Biological Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Krystyna Mazan-Mamczarz
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Ji Heon Noh
- Molecular Aging Biology Laboratory (MABL), Department of Biochemistry, College of Natural Science, Chungnam National University, Daejeon, Republic of Korea
| | - Kyoung Mi Kim
- Department of Biological Sciences, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
9
|
Yakubov LA, Taranov OS, Sidorov SV, Nikonov SD, Ostanin AA, Chernykh ER, Kolchanov NA, Bogachev SS. The concept of natural genome reconstruction.Part 1. Basic provisions of the "natural genome reconstruction" concept. Changing the genome of hematopoietic stem cells using several natural cellular mechanisms that are inherent in the hematopoietic cell and determine its biological status as "the source of the body's reparative potential". Vavilovskii Zhurnal Genet Selektsii 2024; 28:696-705. [PMID: 39722670 PMCID: PMC11667570 DOI: 10.18699/vjgb-24-78] [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: 06/13/2024] [Revised: 08/21/2024] [Accepted: 09/05/2024] [Indexed: 12/28/2024] Open
Abstract
We present a series of articles proving the existence of a previously unknown mechanism of interaction between hematopoietic stem cells and extracellular double-stranded DNA (and, in particular, double-stranded DNA of the peripheral bloodstream), which explains the possibility of emergence and fixation of genetic information contained in double-stranded DNA of extracellular origin in hematopoietic stem cells. The concept of the possibility of stochastic or targeted changes in the genome of hematopoietic stem cells is formulated based on the discovery of new, previously unknown biological properties of poorly differentiated hematopoietic precursors. The main provisions of the concept are as follows. The hematopoietic stem cell takes up and internalizes fragments of extracellular double-stranded DNA via a natural mechanism. Specific groups of glycocalyx factors, including glycoproteins/proteoglycans, glycosylphosphatidylinositol-anchored proteins and scavenger receptors, take part in the internalization event. The binding sites for DNA fragments are heparin-binding domains and clusters of positively charged amino acid residues that are parts of protein molecules of these factors. Extracellular fragments delivered to the internal compartments of hematopoietic stem cells initiate terminal differentiation, colony formation, and proliferation of hematopoietic precursors. The molecular manifestation of these processes is the emergence and repair of pangenomic single-strand breaks. The occurrence of pangenomic single-strand breaks and restoration of genome (genomic DNA) integrity are associated with activation of a "recombinogenic situation" in the cell; during its active phase, stochastic homologous recombination or other recombination events between extracellular fragments localized in the nucleus and chromosomal DNA are possible. As a result, genetic material of initially extracellular localization either integrates into the recipient genome with the replacement of homologous chromosomal segments, or is transitively present in the nucleus and can manifest itself as a new genetic trait. It is assumed that as a result of stochastic acts of homologous exchange, chromosome loci are corrected in hematopoietic stem cells that have acquired mutations during the existence of the organism, which are the cause of clonal hematopoiesis associated with old age. In this regard, there is a fundamental possibility of changing the hematopoietic status of hematopoietic stem cells in the direction of polyclonality and the original diversity of clones. Such events can form the basis for the rejuvenation of the blood-forming cell system. The results of the laboratory's work indicate that other stem cells in the body capture extracellular DNA fragments too. This fact creates a paradigm for the overall rejuvenation of the body.
Collapse
Affiliation(s)
| | - O S Taranov
- State Scientific Center of Virology and Biotechnology "Vector" of Rospotrebnadzor, Koltsovo, Novosibirsk region, Russia
| | - S V Sidorov
- City Clinical Hospital No. 1, Novosibirsk, Russia
| | - S D Nikonov
- Novosibirsk Tuberculosis Research Institute, Novosibirsk, Russia
| | - A A Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - E R Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - N A Kolchanov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S S Bogachev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
10
|
Abbas A, Almaghrbi H, Giordo R, Zayed H, Pintus G. Pathogenic mechanisms, diagnostic, and therapeutic potential of microvesicles in diabetes and its complications. Arch Biochem Biophys 2024; 761:110168. [PMID: 39349130 DOI: 10.1016/j.abb.2024.110168] [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: 08/06/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
Extracellular vesicles (EVs), particularly microvesicles (MVs), have gained significant attention for their role as mediators of intercellular communication in both physiological and pathological contexts, including diabetes mellitus (DM) and its complications. This review provides a comprehensive analysis of the emerging roles of MVs in the pathogenesis of diabetes and associated complications such as nephropathy, retinopathy, cardiomyopathy, and neuropathy. MVs, through their cargo of proteins, lipids, mRNAs, and miRNAs, regulate critical processes like inflammation, oxidative stress, immune responses, and tissue remodeling, all of which contribute to the progression of diabetes and its complications. We examine the molecular mechanisms underlying MVs' involvement in these pathological processes and discuss their potential as biomarkers and therapeutic tools, particularly for drug delivery. Despite promising evidence, challenges remain in isolating and characterizing MVs, understanding their molecular mechanisms, and validating them for clinical use. Advanced techniques such as single-cell RNA sequencing and proteomics are required to gain deeper insights. Improved isolation and purification methods are essential for translating MVs into clinical applications, with potential to develop novel diagnostic and therapeutic strategies to improve patient outcomes in diabetes.
Collapse
Affiliation(s)
- Alaa Abbas
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Heba Almaghrbi
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Roberta Giordo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, 505055, Dubai, United Arab Emirates; Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, 07100, Sassari, Italy
| | - Hatem Zayed
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, 07100, Sassari, Italy; Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
11
|
Yadav J, Chaudhary A, Tripathi T, Janjua D, Joshi U, Aggarwal N, Chhokar A, Keshavam CC, Senrung A, Bharti AC. Exosomal transcript cargo and functional correlation with HNSCC patients' survival. BMC Cancer 2024; 24:1144. [PMID: 39272022 PMCID: PMC11395213 DOI: 10.1186/s12885-024-12759-9] [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: 06/12/2024] [Accepted: 08/02/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND HPV status in a subset of HNSCC is linked with distinct treatment outcomes. Present investigation aims to elucidate the distinct clinicopathological features of HPV-positive and HPV-negative HNSCC and investigate their association with the HNSCC patient survival. MATERIALS AND METHODS The total RNA of exosomes from HPV-positive (93VU147T) and HPV-negative (OCT-1) HNSCC cells was isolated, and the transcripts were estimated using Illumina HiSeq X. The expression of altered transcripts and their clinical relevance were further analyzed using publicly available cancer transcriptome data from The Cancer Genome Atlas (TCGA). RESULTS Transcriptomic analyses identified 3785 differentially exported transcripts (DETs) in HPV-positive exosomes compared to HPV-negative exosomes. DETs that regulate the protein machinery, cellular redox potential, and various neurological disorder-related pathways were over-represented in HPV-positive exosomes. TCGA database revealed the clinical relevance of altered transcripts. Among commonly exported abundant transcripts, SGK1 and MAD1L1 showed high expression, which has been correlated with poor survival in HNSCC patients. In the top 20 DETs of HPV-negative exosomes, high expression of FADS3, SGK3, and TESK2 correlated with poor survival of the HNSCC patients in the TCGA database. CONCLUSION Overall, our study demonstrates that HPV-positive and HPV-negative cells' exosomes carried differential transcripts cargo that may be related to pathways associated with neurological disorders. Additionally, the altered transcripts identified have clinical relevance, correlating with patient survival in HNSCC, thereby highlighting their potential as biomarkers and as therapeutic targets.
Collapse
Affiliation(s)
- Joni Yadav
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Apoorva Chaudhary
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Tanya Tripathi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Divya Janjua
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Udit Joshi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Nikita Aggarwal
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Arun Chhokar
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Department of Zoology, Deshbandhu College, University of Delhi, Delhi, India
| | - Chetkar Chandra Keshavam
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Anna Senrung
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Department of Zoology, Daulat Ram College, University of Delhi, Delhi, India
| | - Alok Chandra Bharti
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India.
| |
Collapse
|
12
|
Xiao D, Li J, Ren Z, Dai M, Jiang Y, Qiu T, Zhang H, Chen Y, Zhang Y, Zhang Y, Palaniyappan L. Association of cortical morphology, white matter hyperintensity, and glymphatic function in frontotemporal dementia variants. Alzheimers Dement 2024; 20:6045-6059. [PMID: 39129270 PMCID: PMC11497707 DOI: 10.1002/alz.14158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/13/2024] [Accepted: 05/25/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Frontotemporal dementia (FTD) can be phenotypically divided into behavioral variant FTD (bvFTD), nonfluent variant primary progressive aphasia (nfvPPA), and semantic variant PPA (svPPA). However, the neural underpinnings of this phenotypic heterogeneity remain elusive. METHODS Cortical morphology, white matter hyperintensities (WMH), diffusion tensor image analysis along the perivascular space (DTI-ALPS), and their interrelationships were assessed in subtypes of FTD. Neuroimaging-transcriptional analyses on the regional cortical morphological deviances among subtypes were also performed. RESULTS Changes in cortical thickness, surface area, gyrification, WMH, and DTI-ALPS were subtype-specific in FTD. The three morphologic indices are related to whole-brain WMH volume and cognitive performance, while cortical thickness is related to DTI-ALPS. Neuroimaging-transcriptional analyses identified key biological pathways linked to the formation and/or spread of TDP-43/tau pathologies. DISCUSSION We found subtype-specific changes in cortical morphology, WMH, and glymphatic function in FTD. Our findings have the potential to contribute to the development of personalized predictions and treatment strategies for this disorder. HIGHLIGHTS Cortical morphologic changes, white matter hyperintensities (WMH), and glymphatic dysfunction are subtype-specific. Cortical morphologic changes, WMH, and glymphatic dysfunction are inter-correlated. Cortical morphologic changes and WMH burden contribute to cognitive impairments.
Collapse
Affiliation(s)
- Die Xiao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Jianyu Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Zhanbing Ren
- College of Physical Education, Shenzhen UniversityShenzhenP. R. China
| | - Minghui Dai
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Yihan Jiang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Ting Qiu
- Douglas Mental Health University InstituteMcGill UniversityMontrealCanada
| | - Huixiong Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Yifan Chen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Youming Zhang
- Department of RadiologyXiangya HospitalCentral South UniversityChangshaP. R. China
- National Clinical Research Center for Geriatric DiseasesXiangya HospitalCentral South UniversityChangshaHunanP. R. China
| | - Yuanchao Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduP. R. China
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduP. R. China
| | - Lena Palaniyappan
- Douglas Mental Health University InstituteMcGill UniversityMontrealCanada
| | | |
Collapse
|
13
|
Pollalis D, Nair GKG, Leung J, Bloemhof CM, Bailey JK, Pennington BO, Kelly KR, Khan AI, Yeh AK, Sundaram KS, Clegg DO, Peng CC, Xu L, Georgescu C, Wren JD, Lee SY. Dynamics of microRNA secreted via extracellular vesicles during the maturation of embryonic stem cell-derived retinal pigment epithelium. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70001. [PMID: 39281021 PMCID: PMC11393772 DOI: 10.1002/jex2.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 09/18/2024]
Abstract
Retinal pigment epithelial (RPE) cells are exclusive to the retina, critically multifunctional in maintaining the visual functions and health of photoreceptors and the retina. Despite their vital functions throughout lifetime, RPE cells lack regenerative capacity, rendering them vulnerable which can lead to degenerative retinal diseases. With advancements in stem cell technology enabling the differentiation of functional cells from pluripotent stem cells and leveraging the robust autocrine and paracrine functions of RPE cells, extracellular vesicles (EVs) secreted by RPE cells hold significant therapeutic potential in supplementing RPE cell activity. While previous research has primarily focused on the trophic factors secreted by RPE cells, there is a lack of studies investigating miRNA, which serves as a master regulator of gene expression. Profiling and defining the functional role of miRNA contained within RPE-secreted EVs is critical as it constitutes a necessary step in identifying the optimal phenotype of the EV-secreting cell and understanding the biological cargo of EVs to develop EV-based therapeutics. In this study, we present a comprehensive profile of miRNA in small extracellular vesicles (sEVs) secreted during RPE maturation following differentiation from human embryonic stem cells (hESCs); early-stage hESC-RPE (20-21 days in culture), mid-stage hESC-RPE (30-31 days in culture) and late-stage hESC-RPE (60-61 days in culture). This exploration is essential for ongoing efforts to develop and optimize EV-based intraocular therapeutics utilizing RPE-secreted EVs, which may significantly impact the function of dysfunctional RPE cells in retinal diseases.
Collapse
Affiliation(s)
- Dimitrios Pollalis
- USC Roski Eye Institute, Keck School of Medicine University of Southern California Los Angeles California USA
- USC Ginsburg Institute for Biomedical Therapeutics University of Southern California Los Angeles California USA
| | - Gopa Kumar Gopinadhan Nair
- USC Roski Eye Institute, Keck School of Medicine University of Southern California Los Angeles California USA
- USC Ginsburg Institute for Biomedical Therapeutics University of Southern California Los Angeles California USA
| | - Justin Leung
- USC Roski Eye Institute, Keck School of Medicine University of Southern California Los Angeles California USA
- USC Dornsife College of Letters, Arts and Sciences Los Angeles California USA
| | - Clarisa Marie Bloemhof
- USC Roski Eye Institute, Keck School of Medicine University of Southern California Los Angeles California USA
- University of Southern California Los Angeles California USA
- School of Medicine California University of Science and Medicine Colton California USA
| | - Jeffrey K Bailey
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute University of California Santa Barbara California USA
- Department of Molecular Cellular and Developmental Biology University of California Santa Barbara California USA
| | - Britney O Pennington
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute University of California Santa Barbara California USA
- Department of Molecular Cellular and Developmental Biology University of California Santa Barbara California USA
| | - Kaitlin R Kelly
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute University of California Santa Barbara California USA
- Department of Molecular Cellular and Developmental Biology University of California Santa Barbara California USA
| | - Amir I Khan
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute University of California Santa Barbara California USA
- Department of Molecular Cellular and Developmental Biology University of California Santa Barbara California USA
| | - Ashley K Yeh
- Department of Molecular Cellular and Developmental Biology University of California Santa Barbara California USA
- College of Creative Studies, Biology University of California Santa Barbara California USA
| | - Kartik S Sundaram
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute University of California Santa Barbara California USA
- Biomolecular Science and Engineering University of California Santa Barbara California USA
| | - Dennis O Clegg
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute University of California Santa Barbara California USA
- Department of Molecular Cellular and Developmental Biology University of California Santa Barbara California USA
- Biomolecular Science and Engineering University of California Santa Barbara California USA
| | - Chen-Ching Peng
- USC Roski Eye Institute, Keck School of Medicine University of Southern California Los Angeles California USA
- Children's Hospital Los Angeles Vision Center Los Angeles California USA
| | - Liya Xu
- USC Roski Eye Institute, Keck School of Medicine University of Southern California Los Angeles California USA
- Children's Hospital Los Angeles Vision Center Los Angeles California USA
| | - Constantin Georgescu
- Genes & Human Diseases Research Program Oklahoma Medical Research Foundation Oklahoma City Oklahoma USA
| | - Jonathan D Wren
- Genes & Human Diseases Research Program Oklahoma Medical Research Foundation Oklahoma City Oklahoma USA
| | - Sun Young Lee
- USC Roski Eye Institute, Keck School of Medicine University of Southern California Los Angeles California USA
- USC Ginsburg Institute for Biomedical Therapeutics University of Southern California Los Angeles California USA
- Department of Physiology and Neuroscience, Keck School of Medicine University of Southern California Los Angeles California USA
| |
Collapse
|
14
|
Katana Z, Sianidou K, Kaiopoulos G, Deligianni F, Tsetsakos S, Kouvatsi A, Sakellari I, Kritis A, Touraki M, Sotiropoulos D, Xagorari A. Molecular and biochemical evaluation of oxidative effects of cord blood CD34+ MPs on hematopoietic cells. Blood Cells Mol Dis 2024; 108:102871. [PMID: 39013336 DOI: 10.1016/j.bcmd.2024.102871] [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: 03/27/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/18/2024]
Abstract
A graft source for allogeneic hematopoietic stem cell transplantation is umbilical cord blood, which contains umbilical cord blood mononuclear cells (MNCs and mesenchymal stem cells, both an excellent source of extracellular microparticles (MPs). MPs act as cell communication mediators, which are implicated in reactive oxygen species formation or detoxification depending on their origin. Oxidative stress plays a crucial role in both the development of cancer and its treatment by triggering apoptotic mechanisms, in which CD34+ cells are implicated. The aim of this work is to investigate the oxidative stress status and the apoptosis of HL-60 and mononuclear cells isolated from umbilical cord blood (UCB) following a 24- and 48-hour exposure to CD34 + microparticles (CD34 + MPs). The activity of superoxide dismutase, glutathione reductase, and glutathione S-transferase, as well as lipid peroxidation in the cells, were employed as oxidative stress markers. A 24- and 48-hour exposure of leukemic and mononuclear cells to CD34 + -MPs resulted in a statistically significant increase in the antioxidant activity and lipid peroxidation in both cells types. Moreover, CD34 + MPs affect the expression of BCL2 and FAS and related proteins and downregulate the hematopoietic differentiation program in both HL-60 and mononuclear cells. Our results indicate that MPs through activation of antioxidant enzymes in both homozygous and nonhomozygous cells might serve as a means for graft optimization and enhancement.
Collapse
Affiliation(s)
- Zoi Katana
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kyriaki Sianidou
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gregory Kaiopoulos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Fani Deligianni
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sarantis Tsetsakos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Anastasia Kouvatsi
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Sakellari
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Aristeidis Kritis
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Touraki
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Damianos Sotiropoulos
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece
| | - Angeliki Xagorari
- Public Cord Blood Bank, Hematology Department, G.H.G.Papanicolaou, Thessaloniki, Greece.
| |
Collapse
|
15
|
Carbone D, Santos MF, Corbeil D, Vistoli G, Parrino B, Karbanová J, Cascioferro S, Pecoraro C, Bauson J, Eliwat W, Aalam F, Cirrincione G, Lorico A, Diana P. Triazole derivatives inhibit the VOR complex-mediated nuclear transport of extracellular particles: Potential application in cancer and HIV-1 infection. Bioorg Chem 2024; 150:107589. [PMID: 38941696 DOI: 10.1016/j.bioorg.2024.107589] [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: 04/24/2024] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 06/30/2024]
Abstract
Extracellular vesicles (EVs) appear to play an important role in intercellular communication in various physiological processes and pathological conditions such as cancer. Like enveloped viruses, EVs can transport their contents into the nucleus of recipient cells, and a new intracellular pathway has been described to explain the nuclear shuttling of EV cargoes. It involves a tripartite protein complex consisting of vesicle-associated membrane protein-associated protein A (VAP-A), oxysterol-binding protein (OSBP)-related protein-3 (ORP3) and late endosome-associated Rab7 allowing late endosome entry into the nucleoplasmic reticulum. Rab7 binding to ORP3-VAP-A complex can be blocked by the FDA-approved antifungal drug itraconazole. Here, we design a new series of smaller triazole derivatives, which lack the dioxolane moiety responsible for the antifungal function, acting on the hydrophobic sterol-binding pocket of ORP3 and evaluate their structure-activity relationship through inhibition of VOR interactions and nuclear transfer of EV and HIV-1 cargoes. Our investigation reveals that the most effective compounds that prevent nuclear transfer of EV cargo and productive infection by VSV-G-pseudotyped HIV-1 are those with a side chain between 1 and 4 carbons, linear or branched (methyl) on the triazolone region. These potent chemical drugs could find clinical applications either for nuclear transfer of cancer-derived EVs that impact metastasis or viral infection.
Collapse
Affiliation(s)
- Daniela Carbone
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy.
| | - Mark F Santos
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA.
| | - Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany; Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Dresden, Germany.
| | - Giulio Vistoli
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy.
| | - Barbara Parrino
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy.
| | - Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany; Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Dresden, Germany.
| | - Stella Cascioferro
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy.
| | - Camilla Pecoraro
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy.
| | - Jodi Bauson
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA.
| | - Waleed Eliwat
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA.
| | - Feryal Aalam
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA.
| | - Girolamo Cirrincione
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy.
| | - Aurelio Lorico
- Touro University Nevada College of Osteopathic Medicine, Henderson, NV, USA.
| | - Patrizia Diana
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy.
| |
Collapse
|
16
|
Lanci A, Iacono E, Merlo B. Therapeutic Application of Extracellular Vesicles Derived from Mesenchymal Stem Cells in Domestic Animals. Animals (Basel) 2024; 14:2147. [PMID: 39123673 PMCID: PMC11310970 DOI: 10.3390/ani14152147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/12/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Recently, the therapeutic potential of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) has been extensively studied in both human and veterinary medicine. EVs are nano-sized particles containing biological components commonly found in other biological materials. For that reason, EV isolation and characterization are critical to draw precise conclusions during their investigation. Research on EVs within veterinary medicine is still considered in its early phases, yet numerous papers were published in recent years. The conventional adult tissues for deriving MSCs include adipose tissue and bone marrow. Nonetheless, alternative sources such as synovial fluid, endometrium, gingiva, and milk have also been intermittently used. Fetal adnexa are amniotic membrane/fluid, umbilical cord and Wharton's jelly. Cells derived from fetal adnexa exhibit an intermediate state between embryonic and adult cells, demonstrating higher proliferative and differentiative potential and longer telomeres compared to cells from adult tissues. Summarized here are the principal and recent preclinical and clinical studies performed in domestic animals such as horse, cattle, dog and cat. To minimize the use of antibiotics and address the serious issue of antibiotic resistance as a public health concern, they will undoubtedly also be utilized in the future to treat infections in domestic animals. A number of concerns, including large-scale production with standardization of EV separation and characterization techniques, must be resolved for clinical application.
Collapse
Affiliation(s)
- Aliai Lanci
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (E.I.); (B.M.)
| | - Eleonora Iacono
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (E.I.); (B.M.)
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, 40100 Bologna, Italy
| | - Barbara Merlo
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (E.I.); (B.M.)
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, 40100 Bologna, Italy
| |
Collapse
|
17
|
Wardhani K, Levina A, Grau GER, Lay PA. Fluorescent, phosphorescent, magnetic resonance contrast and radioactive tracer labelling of extracellular vesicles. Chem Soc Rev 2024; 53:6779-6829. [PMID: 38828885 DOI: 10.1039/d2cs00238h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
This review focusses on the significance of fluorescent, phosphorescent labelling and tracking of extracellular vesicles (EVs) for unravelling their biology, pathophysiology, and potential diagnostic and therapeutic uses. Various labeling strategies, such as lipid membrane, surface protein, luminal, nucleic acid, radionuclide, quantum dot labels, and metal complex-based stains, are evaluated for visualizing and characterizing EVs. Direct labelling with fluorescent lipophilic dyes is simple but generally lacks specificity, while surface protein labelling offers selectivity but may affect EV-cell interactions. Luminal and nucleic acid labelling strategies have their own advantages and challenges. Each labelling approach has strengths and weaknesses, which require a suitable probe and technique based on research goals, but new tetranuclear polypyridylruthenium(II) complexes as phosphorescent probes have strong phosphorescence, selective staining, and stability. Future research should prioritize the design of novel fluorescent probes and labelling platforms that can significantly enhance the efficiency, accuracy, and specificity of EV labeling, while preserving their composition and functionality. It is crucial to reduce false positive signals and explore the potential of multimodal imaging techniques to gain comprehensive insights into EVs.
Collapse
Affiliation(s)
- Kartika Wardhani
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, 87545, USA
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Georges E R Grau
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Analytical, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
18
|
Merij LB, da Silva LR, Palhinha L, Gomes MT, Dib PRB, Martins-Gonçalves R, Toledo-Quiroga K, Raposo-Nunes MA, Andrade FB, de Toledo Martins S, Nascimento ALR, Rocha VN, Alves LR, Bozza PT, de Oliveira Trugilho MR, Hottz ED. Density-based lipoprotein depletion improves extracellular vesicle isolation and functional analysis. J Thromb Haemost 2024; 22:1372-1388. [PMID: 38278418 DOI: 10.1016/j.jtha.2024.01.010] [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: 05/15/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Blood plasma is the main source of extracellular vesicles (EVs) in clinical studies aiming to identify biomarkers and to investigate pathophysiological processes, especially regarding EV roles in inflammation and thrombosis. However, EV isolation from plasma has faced the fundamental issue of lipoprotein contamination, representing an important bias since lipoproteins are highly abundant and modulate cell signaling, metabolism, and thromboinflammation. OBJECTIVES Here, we aimed to isolate plasma EVs after depleting lipoproteins, thereby improving sample purity and EV thromboinflammatory analysis. METHODS Density-based gradient ultracentrifugation (G-UC) was used for lipoprotein depletion before EV isolation from plasma through size-exclusion chromatography (SEC) or serial centrifugation (SC). Recovered EVs were analyzed by size, concentration, cellular source, ultrastructure, and bottom-up proteomics. RESULTS G-UC efficiently separated lipoproteins from the plasma, allowing subsequent EV isolation through SEC or SC. Combined analysis from EV proteomics, cholesterol quantification, and apoB-100 detection confirmed the significant reduction in lipoproteins from isolated EVs. Proteomic analysis identified similar gene ontology and cellular components in EVs, regardless of lipoprotein depletion, which was consistent with similar EV cellular sources, size, and ultrastructure by flow cytometry and transmission electron microscopy. Importantly, lipoprotein depletion increased the detection of less abundant proteins in EV proteome and enhanced thromboinflammatory responses of platelets and monocytes stimulated in vitro with EV isolates. CONCLUSION Combination of G-UC+SEC significantly reduced EV lipoprotein contamination without interfering in EV cellular source, gene ontology, and ultrastructure, allowing the recovery of highly pure EVs with potential implications for functional assays and proteomic and lipidomic analyses.
Collapse
Affiliation(s)
- Laura Botelho Merij
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Luana Rocha da Silva
- Laboratory of Toxinology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lohanna Palhinha
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Milena Tavares Gomes
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Paula Ribeiro Braga Dib
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Remy Martins-Gonçalves
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kemily Toledo-Quiroga
- Laboratory of Toxinology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Fernanda Brandi Andrade
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Sharon de Toledo Martins
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, ICC-Fiocruz, Curitiba, Paraná, Brazil
| | - Ana Lúcia Rosa Nascimento
- Laboratory of Ultrastructure and Tissue, Department of Histology and Embryology, State University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinicius Novaes Rocha
- Laboratory of Veterinary Pathology and Histology, Department of Veterinary Medicine, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Lysangela Ronalte Alves
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, ICC-Fiocruz, Curitiba, Paraná, Brazil
| | - Patrícia T Bozza
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, ICC-Fiocruz, Curitiba, Paraná, Brazil
| | - Monique Ramos de Oliveira Trugilho
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil; Center for Technological Development in Health, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Eugenio D Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| |
Collapse
|
19
|
Wang R, Shi Y, Lv Y, Xie C, Hu Y. The novel insights of epithelial-derived exosomes in various fibrotic diseases. Biomed Pharmacother 2024; 174:116591. [PMID: 38631144 DOI: 10.1016/j.biopha.2024.116591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
The characteristics of fibrosis include the abnormal accumulation of extracellular matrix proteins and abnormal tissue repair caused by injury, infection, and inflammation, leading to a significant increase in organ failure and mortality. Effective and precise treatments are urgently needed to halt and reverse the progression of fibrotic diseases. Exosomes are tiny vesicles derived from endosomes, spanning from 40 to 160 nanometers in diameter, which are expelled into the extracellular matrix environment by various cell types. They play a crucial role in facilitating cell-to-cell communication by transporting a variety of cargoes, including proteins, RNA, and DNA. Epithelial cells serve as the primary barrier against diverse external stimuli that precipitate fibrotic diseases. Numerous research suggests that exosomes from epithelial cells have a significant impact on several fibrotic diseases. An in-depth comprehension of the cellular and molecular mechanisms of epithelial cell-derived exosomes in fibrosis holds promise for advancing the exploration of novel diagnostic biomarkers and clinical drug targets. In this review, we expand upon the pathogenic mechanisms of epithelium-derived exosomes and highlight their role in the fibrotic process by inducing inflammation and activating fibroblasts. In addition, we are particularly interested in the bioactive molecules carried by epithelial-derived exosomes and their potential value in the diagnosis and treatment of fibrosis and delineate the clinical utility of exosomes as an emerging therapeutic modality, highlighting their potential application in addressing various medical conditions.
Collapse
Affiliation(s)
- Rifu Wang
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yuxin Shi
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yonglin Lv
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Changqing Xie
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, China.
| | - Yanjia Hu
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China.
| |
Collapse
|
20
|
Wang Y, Xiong Y, Shi K, Effah CY, Song L, He L, Liu J. DNA nanostructures for exploring cell-cell communication. Chem Soc Rev 2024; 53:4020-4044. [PMID: 38444346 DOI: 10.1039/d3cs00944k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
The process of coordinating between the same or multiple types of cells to jointly execute various instructions in a controlled and carefully regulated environment is a very appealing field. In order to provide clearer insight into the role of cell-cell interactions and the cellular communication of this process in their local communities, several interdisciplinary approaches have been employed to enhance the core understanding of this phenomenon. DNA nanostructures have emerged in recent years as one of the most promising tools in exploring cell-cell communication and interactions due to their programmability and addressability. Herein, this review is dedicated to offering a new perspective on using DNA nanostructures to explore the progress of cell-cell communication. After briefly outlining the anchoring strategy of DNA nanostructures on cell membranes and the subsequent dynamic regulation of DNA nanostructures, this paper highlights the significant contribution of DNA nanostructures in monitoring cell-cell communication and regulating its interactions. Finally, we provide a quick overview of the current challenges and potential directions for the application of DNA nanostructures in cellular communication and interactions.
Collapse
Affiliation(s)
- Ya Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Yamin Xiong
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kangqi Shi
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Clement Yaw Effah
- The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou 450003, China
| | - Lulu Song
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Leiliang He
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Jianbo Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, China.
| |
Collapse
|
21
|
Pollalis D, Georgescu C, Wren JD, Tombulyan G, Leung JM, Lo PA, Bloemhof CM, Lee RH, Bae E, Bailey JK, Pennington BO, Khan AI, Kelly KR, Yeh AK, Sundaram KS, Humayun M, Louie S, Clegg DO, Lee SY. Rescuing Photoreceptors in RPE Dysfunction-Driven Retinal Degeneration: The Role of Small Extracellular Vesicles Secreted from Retinal Pigment Epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588773. [PMID: 38645051 PMCID: PMC11030310 DOI: 10.1101/2024.04.09.588773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Dysfunction of the retinal pigment epithelium (RPE) is a common shared pathology in major degenerative retinal diseases despite variations in the primary etiologies of each disease. Due to their demanding and indispensable functional roles throughout the lifetime, RPE cells are vulnerable to genetic predisposition, external stress, and aging processes. Building upon recent advancements in stem cell technology for differentiating healthy RPE cells and recognizing the significant roles of small extracellular vesicles (sEV) in cellular paracrine and autocrine actions, we investigated the hypothesis that the RPE-secreted sEV alone can restore essential RPE functions and rescue photoreceptors in RPE dysfunction-driven retinal degeneration. Our findings support the rationale for developing intravitreal treatment of sEV. We demonstrate that intravitreally delivered sEV effectively penetrate the full thickness of the retina. Xenogenic intraocular administration of human-derived EVs did not induce acute immune reactions in rodents. sEV derived from human embryonic stem cell (hESC)-derived fully differentiated RPE cells, but not sEV-depleted conditioned cell culture media (CCM minus sEV), rescued photoreceptors and their function in a Royal College of Surgeons (RCS) rat model. This model is characterized by photoreceptor death and retinal degeneration resulting from a mutation in the MerTK gene in RPE cells. From the bulk RNA sequencing study, we identified 447 differently expressed genes in the retina after hESC-RPE-sEV treatment compared with the untreated control. Furthermore, 394 out of 447 genes (88%) showed a reversal in expression toward the healthy state in Long-Evans (LE) rats after treatment compared to the diseased state. Particularly, detrimental alterations in gene expression in RCS rats, including essential RPE functions such as phototransduction, vitamin A metabolism, and lipid metabolism were partially reversed. Defective photoreceptor outer segment engulfment due to intrinsic MerTK mutation was partially ameliorated. These findings suggest that RPE-secreted sEV may play a functional role similar to that of RPE cells. Our study justifies further exploration to fully unlock future therapeutic interventions with sEV in a broad array of degenerative retinal diseases.
Collapse
|
22
|
Kistenmacher S, Schwämmle M, Martin G, Ulrich E, Tholen S, Schilling O, Gießl A, Schlötzer-Schrehardt U, Bucher F, Schlunck G, Nazarenko I, Reinhard T, Polisetti N. Enrichment, Characterization, and Proteomic Profiling of Small Extracellular Vesicles Derived from Human Limbal Mesenchymal Stromal Cells and Melanocytes. Cells 2024; 13:623. [PMID: 38607062 PMCID: PMC11011788 DOI: 10.3390/cells13070623] [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: 03/11/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
Limbal epithelial progenitor cells (LEPC) rely on their niche environment for proper functionality and self-renewal. While extracellular vesicles (EV), specifically small EVs (sEV), have been proposed to support LEPC homeostasis, data on sEV derived from limbal niche cells like limbal mesenchymal stromal cells (LMSC) remain limited, and there are no studies on sEVs from limbal melanocytes (LM). In this study, we isolated sEV from conditioned media of LMSC and LM using a combination of tangential flow filtration and size exclusion chromatography and characterized them by nanoparticle tracking analysis, transmission electron microscopy, Western blot, multiplex bead arrays, and quantitative mass spectrometry. The internalization of sEV by LEPC was studied using flow cytometry and confocal microscopy. The isolated sEVs exhibited typical EV characteristics, including cell-specific markers such as CD90 for LMSC-sEV and Melan-A for LM-sEV. Bioinformatics analysis of the proteomic data suggested a significant role of sEVs in extracellular matrix deposition, with LMSC-derived sEV containing proteins involved in collagen remodeling and cell matrix adhesion, whereas LM-sEV proteins were implicated in other cellular bioprocesses such as cellular pigmentation and development. Moreover, fluorescently labeled LMSC-sEV and LM-sEV were taken up by LEPC and localized to their perinuclear compartment. These findings provide valuable insights into the complex role of sEV from niche cells in regulating the human limbal stem cell niche.
Collapse
Affiliation(s)
- Sebastian Kistenmacher
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Melanie Schwämmle
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D–79104 Freiburg, Germany
| | - Gottfried Martin
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Eva Ulrich
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Stefan Tholen
- Institute of Surgical Pathology, Faculty of Medicine, Freiburg, Medical Center, University of Freiburg, 79085 Freiburg im Breisgau, Germany
| | - Oliver Schilling
- Institute of Surgical Pathology, Faculty of Medicine, Freiburg, Medical Center, University of Freiburg, 79085 Freiburg im Breisgau, Germany
| | - Andreas Gießl
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlan-gen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlan-gen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Felicitas Bucher
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Thomas Reinhard
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Naresh Polisetti
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| |
Collapse
|
23
|
Ratajczak MZ, Ratajczak J. Leukemogenesis occurs in a microenvironment enriched by extracellular microvesicles/exosomes: recent discoveries and questions to be answered. Leukemia 2024; 38:692-698. [PMID: 38388648 PMCID: PMC10997496 DOI: 10.1038/s41375-024-02188-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024]
Abstract
In single-cell organisms, extracellular microvesicles (ExMVs) were one of the first cell-cell communication platforms that emerged very early during evolution. Multicellular organisms subsequently adapted this mechanism. Evidence indicates that all types of cells secrete these small circular structures surrounded by a lipid membrane that may be encrusted by ligands and receptors interacting with target cells and harboring inside a cargo comprising RNA species, proteins, bioactive lipids, signaling nucleotides, and even entire organelles "hijacked" from the cells of origin. ExMVs are secreted by normal cells and at higher levels by malignant cells, and there are some differences in their cargo. On the one hand, ExMVs secreted from malignant cells interact with cells in the microenvironment, and in return, they are exposed by a "two-way mechanism" to ExMVs secreted by non-leukemic cells. Therefore, leukemogenesis occurs and progresses in ExMVs enriched microenvironments, and this biological fact has pathologic, diagnostic, and therapeutic implications. We are still trying to decipher this intriguing cell-cell communication language better. We will present a current point of view on this topic and review some selected most recent discoveries and papers.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland.
| | - Janina Ratajczak
- Stem Cell Institute at Brown Cancer Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
24
|
Si G, Chen X, Li Y, Yuan X. Exosomes promote pre-metastatic niche formation in colorectal cancer. Heliyon 2024; 10:e27572. [PMID: 38509970 PMCID: PMC10950591 DOI: 10.1016/j.heliyon.2024.e27572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/22/2024] Open
Abstract
It is well known that colorectal cancer (CRC) has a high morbidity rate, a poor prognosis when metastasized, and a greatly shortened 5-year survival rate. Therefore, understanding the mechanism of tumor metastasis is still important. Based on the "seed and soil" theory, the concept of " premetastatic niche (PMN)" was introduced by Kaplan et al. The complex interaction between primary tumors and the metastatic organ provides a beneficial microenvironment for tumor cells to colonize at a distance. With further exploration of the PMN, exosomes have gradually attracted interest from researchers. Exosomes are extracellular vesicles secreted from cells that include various biological information and are involved in communication between cells. As a key molecule in the PMN, exosomes are closely related to tumor metastasis. In this article, we obtained information by conducting a comprehensive search across academic databases including PubMed and Web of Science using relevant keywords. Only recent, peer-reviewed articles published in the English language were considered for inclusion. This study aims to explore in depth how exosomes promote the formation of pre-metastatic microenvironment (PMN) in colorectal cancer and its related mechanisms.
Collapse
Affiliation(s)
- Guifei Si
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, 261000, China
| | - Xuemei Chen
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, 261000, China
| | - Yuquan Li
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, 261000, China
| | - Xuemin Yuan
- Department of Gastroenterology, Linyi People's Hospital, Linyi, Shandong, 276000, China
| |
Collapse
|
25
|
Ebrahim T, Ebrahim AS, Kandouz M. Diversity of Intercellular Communication Modes: A Cancer Biology Perspective. Cells 2024; 13:495. [PMID: 38534339 DOI: 10.3390/cells13060495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/27/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024] Open
Abstract
From the moment a cell is on the path to malignant transformation, its interaction with other cells from the microenvironment becomes altered. The flow of molecular information is at the heart of the cellular and systemic fate in tumors, and various processes participate in conveying key molecular information from or to certain cancer cells. For instance, the loss of tight junction molecules is part of the signal sent to cancer cells so that they are no longer bound to the primary tumors and are thus free to travel and metastasize. Upon the targeting of a single cell by a therapeutic drug, gap junctions are able to communicate death information to by-standing cells. The discovery of the importance of novel modes of cell-cell communication such as different types of extracellular vesicles or tunneling nanotubes is changing the way scientists look at these processes. However, are they all actively involved in different contexts at the same time or are they recruited to fulfill specific tasks? What does the multiplicity of modes mean for the overall progression of the disease? Here, we extend an open invitation to think about the overall significance of these questions, rather than engage in an elusive attempt at a systematic repertory of the mechanisms at play.
Collapse
Affiliation(s)
- Thanzeela Ebrahim
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Abdul Shukkur Ebrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Mustapha Kandouz
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48202, USA
| |
Collapse
|
26
|
McDonald J, Mohak S, Fabian Z. Stem Cell-Derived Extracellular Vesicles in the Treatment of Cardiovascular Diseases. Pharmaceutics 2024; 16:381. [PMID: 38543275 PMCID: PMC10974254 DOI: 10.3390/pharmaceutics16030381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 01/03/2025] Open
Abstract
Cardiovascular disease constitutes a noteworthy public health challenge characterized by a pronounced incidence, frequency, and mortality rate, particularly impacting specific demographic groups, and imposing a substantial burden on the healthcare infrastructure. Certain risk factors, such as age, gender, and smoking, contribute to the prevalence of fatal cardiovascular disease, highlighting the need for targeted interventions. Current challenges in clinical practice involve medication complexities, the lack of a systematic decision-making approach, and prevalent drug therapy problems. Stem cell-derived extracellular vesicles stand as versatile entities with a unique molecular fingerprint, holding significant therapeutic potential across a spectrum of applications, particularly in the realm of cardio-protection. Their lipid, protein, and nucleic acid compositions, coupled with their multifaceted functions, underscore their role as promising mediators in regenerative medicine and pave the way for further exploration of their intricate contributions to cellular physiology and pathology. Here, we overview our current understanding of the possible role of stem cell-derived extracellular vesicles in the clinical management of human cardiovascular pathologies.
Collapse
Affiliation(s)
- Jennifer McDonald
- School of Medicine and Dentistry, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Fylde Road, Preston PR1 2HE, UK;
| | - Sidhesh Mohak
- Department of Internal Medicine, South Texas Health System, McAllen, TX 78503, USA;
| | - Zsolt Fabian
- School of Medicine and Dentistry, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Fylde Road, Preston PR1 2HE, UK;
| |
Collapse
|
27
|
Leung J, Pollalis D, Nair GKG, Bailey JK, Pennington BO, Khan AI, Kelly KR, Yeh AK, Sundaram KS, Clegg DO, Peng CC, Xu L, Lee SY. Isolation and Characterization of Extracellular Vesicles Through Orthogonal Approaches for the Development of Intraocular EV Therapy. Invest Ophthalmol Vis Sci 2024; 65:6. [PMID: 38466285 PMCID: PMC10929743 DOI: 10.1167/iovs.65.3.6] [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: 12/01/2023] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
Purpose Isolating extracellular vesicles (EVs) with high yield, replicable purity, and characterization remains a bottleneck in the development of EV therapeutics. To address these challenges, the current study aims to establish the necessary framework for preclinical and clinical studies in the development of stem cell-derived intraocular EV therapeutics. Methods Small EVs (sEVs) were separated from the conditioned cell culture medium (CCM) of the human embryogenic stem cell-derived fully polarized retinal pigment epithelium (hESC-RPE-sEV) by a commercially available microfluidic tangential flow filtration (TFF) device ExoDisc (ED) or differential ultracentrifugation (dUC). The scaling and concentration capabilities and purity of recovered sEVs were assessed. Size, number, and surface markers of sEVs were determined by orthogonal approaches using multiple devices. Results ED yielded higher numbers of sEVs, ranging from three to eight times higher depending on the measurement device, compared to dUC using the same 5 mL of CCM input. Within the same setting, the purity of ED-recovered hESC-RPE-sEVs was higher than that for dUC-recovered sEVs. ED yielded a higher concentration of particles, which is strongly correlated with the input volume, up to 10 mL (r = 0.98, P = 0.016). Meanwhile, comprehensive characterization profiles of EV surface markers between ED- and dUC-recovered hESC-RPE-sEVs were compatible. Conclusions Our study supports TFF as a valuable strategy for separating sEVs for the development of intraocular EV therapeutics. However, there is a growing need for diverse devices to optimize TFF for use in EV preparation. Using orthogonal approaches in EV characterization remains ideal for reliably characterizing heterogeneous EV.
Collapse
Affiliation(s)
- Justin Leung
- USC Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, United States
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
| | - Dimitrios Pollalis
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Gopa K. G. Nair
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Jeffrey K. Bailey
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Britney O. Pennington
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Amir I. Khan
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Kaitlin R. Kelly
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Ashley K. Yeh
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- College of Creative Studies, Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Kartik S. Sundaram
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Biomolecular Science and Engineering, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Dennis O. Clegg
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
- Biomolecular Science and Engineering, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Chen-Ching Peng
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
- Children's Hospital Los Angeles Vision Center, Los Angeles, California, United States
| | - Liya Xu
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
- Children's Hospital Los Angeles Vision Center, Los Angeles, California, United States
| | - Sun Young Lee
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| |
Collapse
|
28
|
Chak PT, Kam NW, Choi TH, Dai W, Kwong DLW. Unfolding the Complexity of Exosome-Cellular Interactions on Tumour Immunity and Their Clinical Prospects in Nasopharyngeal Carcinoma. Cancers (Basel) 2024; 16:919. [PMID: 38473281 DOI: 10.3390/cancers16050919] [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: 01/26/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial malignancy situated in the posterolateral nasopharynx. NPC poses grave concerns in Southeast Asia due to its late diagnosis. Together with resistance to standard treatment combining chemo- and radiotherapy, NPC presents high metastatic rates and common recurrence. Despite advancements in immune-checkpoint inhibitors (ICIs) and cytotoxic-T-lymphocytes (CTLs)-based cellular therapy, the exhaustive T cell profile and other signs of immunosuppression within the NPC tumour microenvironment (TME) remain as concerns to immunotherapy response. Exosomes, extracellular vesicles of 30-150 nm in diameter, are increasingly studied and linked to tumourigenesis in oncology. These bilipid-membrane-bound vesicles are packaged with a variety of signalling molecules, mediating cell-cell communications. Within the TME, exosomes can originate from tumour, immune, or stromal cells. Although there are studies on tumour-derived exosomes (TEX) in NPC and their effects on tumour processes like angiogenesis, metastasis, therapeutic resistance, there is a lack of research on their involvement in immune evasion. In this review, we aim to enhance the comprehension of how NPC TEX contribute to cellular immunosuppression. Furthermore, considering the detectability of TEX in bodily fluids, we will also discuss the potential development of TEX-related biomarkers for liquid biopsy in NPC as this could facilitate early diagnosis and prognostication of the disease.
Collapse
Affiliation(s)
- Paak-Ting Chak
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ngar-Woon Kam
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, New Territories, Hong Kong 999077, China
| | - Tsz-Ho Choi
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Wei Dai
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
29
|
Gai C, Pomatto MAC, Deregibus MC, Dieci M, Piga A, Camussi G. Edible Plant-Derived Extracellular Vesicles for Oral mRNA Vaccine Delivery. Vaccines (Basel) 2024; 12:200. [PMID: 38400183 PMCID: PMC10893065 DOI: 10.3390/vaccines12020200] [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: 01/05/2024] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Nucleic acid delivery through extracellular vesicles (EVs) is a well-preserved evolutionary mechanism in all life kingdoms including eukaryotes, prokaryotes, and plants. EVs naturally allow horizontal transfer of native as well as exogenous functional mRNAs, which once incorporated in EVs are protected from enzymatic degradation. This observation has prompted researchers to investigate whether EVs from different sources, including plants, could be used for vaccine delivery. Several studies using human or bacterial EVs expressing mRNA or recombinant SARS-CoV-2 proteins showed induction of a humoral and cell mediated immune response. Moreover, EV-based vaccines presenting the natural configuration of viral antigens have demonstrated advantages in conferring long-lasting immunization and lower toxicity than synthetic nanoparticles. Edible plant-derived EVs were shown to be an alternative to human EVs for vaccine delivery, especially via oral administration. EVs obtained from orange juice (oEVs) loaded with SARS-CoV-2 mRNAs protected their cargo from enzymatic degradation, were stable at room temperature for one year, and were able to trigger a SARS-CoV-2 immune response in mice. Lyophilized oEVs containing the S1 mRNA administered to rats via gavage induced a specific humoral immune response with generation of blocking antibodies, including IgA and Th1 lymphocyte activation. In conclusion, mRNA-containing oEVs could be used for developing new oral vaccines due to optimal mucosal absorption, resistance to stress conditions, and ability to stimulate a humoral and cellular immune response.
Collapse
Affiliation(s)
- Chiara Gai
- EvoBiotech s.r.l., 10148 Torino, Italy; (C.G.); (M.A.C.P.); (M.D.); (A.P.)
- Department of Medical Sciences, University of Turin, 10126 Torino, Italy;
| | - Margherita Alba Carlotta Pomatto
- EvoBiotech s.r.l., 10148 Torino, Italy; (C.G.); (M.A.C.P.); (M.D.); (A.P.)
- Department of Medical Sciences, University of Turin, 10126 Torino, Italy;
| | | | - Marco Dieci
- EvoBiotech s.r.l., 10148 Torino, Italy; (C.G.); (M.A.C.P.); (M.D.); (A.P.)
| | - Alessandro Piga
- EvoBiotech s.r.l., 10148 Torino, Italy; (C.G.); (M.A.C.P.); (M.D.); (A.P.)
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, 10126 Torino, Italy;
| |
Collapse
|
30
|
Liu Y, Wu J, Chen L, Zou J, Yang Q, Tian H, Zheng D, Ji Z, Cai J, Li Z, Chen Y. ncRNAs-mediated overexpression of TET3 predicts unfavorable prognosis and correlates with immunotherapy efficacy in breast cancer. Heliyon 2024; 10:e24855. [PMID: 38318018 PMCID: PMC10838756 DOI: 10.1016/j.heliyon.2024.e24855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 01/07/2024] [Accepted: 01/16/2024] [Indexed: 02/07/2024] Open
Abstract
Breast cancer is the most frequent form of cancer in women and the primary cause of cancer-related deaths globally. DNA methylation and demethylation are important processes in human tumorigenesis. Ten-eleven translocation 3 (TET3) is a DNA demethylase. Prior research has demonstrated that TET3 is highly expressed in various human malignant tumors. However, the exact function and mechanism of TET3 in breast cancer remain unclear. In this study, we investigated TET3 expression in breast cancer and its correlation with clinicopathological characteristics of breast cancer patients. The results presented that TET3 expression was significantly increased in breast cancer and associated with the PAM50 subtype. Subsequently, we performed receiver operating characteristic, survival, and Cox hazard regression analyses. These results suggest that TET3 expression is associated with a poor prognosis and may be an indirect independent prognostic indicator in breast cancer. We also established a protein-protein interaction (PPI) network of TET3 and executed enrichment analyses of TET3 co-expressed genes, revealing their primary association with the cell cycle. Moreover, we identified noncoding RNAs (ncRNAs) contributing to TET3 overexpression using expression, correlation, and survival analyses. We identified the LINC01521/hsa-miR-29a-3p axis as the primary TET3 upstream ncRNA-related pathway in breast cancer. Furthermore, TET3 expression was positively associated with immune cell infiltration, immune cell biomarkers, and eight immune checkpoint gene expressions in breast cancer. TET3 expression also correlated with patient responses to immunotherapy. Finally, we conducted subcellular localization and immunohistochemical staining analysis of TET3 in breast cancer. We found that TET3 localized to the nucleoplasm, vesicles, and cytosol in the MCF-7 cell line, and TET3 expression was significantly upregulated in breast cancer tissues compared to para-tumor tissues. Our findings indicate that ncRNA-mediated overexpression of TET3 predicts an unfavorable prognosis and correlates with immunotherapy efficacy in breast cancer.
Collapse
Affiliation(s)
| | | | | | - Juan Zou
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Qiuping Yang
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Huiting Tian
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Daitian Zheng
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zeqi Ji
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jiehui Cai
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhiyang Li
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yexi Chen
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
31
|
Ma X, Chen Z, Chen W, Chen Z, Shang Y, Zhao Y, Li L, Zhou C, He J, Meng X. LncRNA AL139294.1 can be transported by extracellular vesicles to promote the oncogenic behaviour of recipient cells through activation of the Wnt and NF-κB2 pathways in non-small-cell lung cancer. J Exp Clin Cancer Res 2024; 43:20. [PMID: 38229152 DOI: 10.1186/s13046-023-02939-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/22/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) participate in cancer development via cell-to-cell communication. Long non-coding RNAs (lncRNAs), one component of EVs, can play an essential role in non-small-cell lung cancer (NSCLC) through EV-mediated delivery. METHODS The NSCLC-associated lncRNA AL139294.1 in EVs was identified via lncRNA microarray analysis. The role of AL139294.1 in NSCLC was examined in vitro and in vivo. Confocal microscopy was used to observe the encapsulation of AL139294.1 into EVs and its transport to recipient cells. A co-culture device was used to examine the effects of transported AL139294.1 on the oncogenic behaviour of recipient cells. Dual-luciferase reporter assay was performed to verify the direct interaction of miR-204-5p with AL139294.1 and bromodomain-containing protein 4 (BRD4). AL139294.1 and miR-204-5p in EVs were quantified using quantitative polymerase chain reaction. Receiver operating characteristic analyses were conducted to evaluate the diagnostic efficiency. RESULTS The lncRNA AL139294.1 in EVs promoted NSCLC progression in vitro and in vivo. After AL139294.1 was encapsulated into EVs and transported to recipient cells, it promoted the cells' proliferation, migration, and invasion abilities by competitively binding with miR-204-5p to regulate BRD4, leading to the activation of the Wnt and NF-κB2 pathways. Additionally, the expression of serum lncRNA AL139294.1 in EVs was increased, whereas miR-204-5p in EVs was decreased in NSCLC. High levels of lncRNA AL139294.1 and low levels of miR-204-5p in EVs were associated with advanced pathological staging, lymph node metastasis, and distant metastasis, underscoring their promising utility for distinguishing between more and less severe manifestations of the disease. CONCLUSIONS This study reveals a novel lncRNA in EVs associated with NSCLC, namely, AL139294.1, providing valuable insights into the development of NSCLC and introducing potential diagnostic biomarkers for NSCLC.
Collapse
Affiliation(s)
- Xinyi Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Zhenhua Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Wei Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315020, China
| | - Ziyuan Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315020, China
| | - Yue Shang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Yikai Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Leyi Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Chengwei Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315020, China
| | - Jinxian He
- Department of Thoracic Surgery, The Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, 315048, China
| | - Xiaodan Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, China.
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
32
|
Zhang N, Shu L, Liu Z, Shi A, Zhao L, Huang S, Sheng G, Yan Z, Song Y, Huang F, Tang Y, Zhang Z. The role of extracellular vesicles in cholangiocarcinoma tumor microenvironment. Front Pharmacol 2024; 14:1336685. [PMID: 38269274 PMCID: PMC10805838 DOI: 10.3389/fphar.2023.1336685] [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: 11/11/2023] [Accepted: 12/26/2023] [Indexed: 01/26/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a highly aggressive malignant tumor that originates from the biliary system. With restricted treatment options at hand, the challenging aspect of early CCA diagnosis leads to a bleak prognosis. Besides the intrinsic characteristics of tumor cells, the generation and progression of CCA are profoundly influenced by the tumor microenvironment, which engages in intricate interactions with cholangiocarcinoma cells. Of notable significance is the role of extracellular vesicles as key carriers in enabling communication between cancer cells and the tumor microenvironment. This review aims to provide a comprehensive overview of current research examining the interplay between extracellular vesicles and the tumor microenvironment in the context of CCA. Specifically, we will emphasize the significant contributions of extracellular vesicles in molding the CCA microenvironment and explore their potential applications in the diagnosis, prognosis assessment, and therapeutic strategies for this aggressive malignancy.
Collapse
Affiliation(s)
- Nuoqi Zhang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Lizhuang Shu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zengli Liu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
- Department of General Surgery, Qilu Hospital, Shandong University, Qingdao, Shandong, China
| | - Anda Shi
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Liming Zhao
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Shaohui Huang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Guoli Sheng
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zhangdi Yan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Yan Song
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Fan Huang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Yongchang Tang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zongli Zhang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
33
|
Ilvonen P, Pusa R, Härkönen K, Laitinen S, Impola U. Distinct targeting and uptake of platelet and red blood cell-derived extracellular vesicles into immune cells. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e130. [PMID: 38938679 PMCID: PMC11080822 DOI: 10.1002/jex2.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/03/2023] [Accepted: 11/28/2023] [Indexed: 06/29/2024]
Abstract
Blood-derived extracellular vesicles (EVs) hold great therapeutic potential. As blood contains mixed EV populations, it is challenging to study EVs originating from different cells separately. Blood cell concentrates manufactured in blood banks offer an excellent non-invasive source of blood cell-specific EV populations. To study blood cell-specific EVs, we isolated EVs from platelet (TREVs) and red blood cell (EryEVs) concentrates and characterized them using nanoparticle tracking analysis, imaging flow cytometry, electron microscopy and western blot analysis and co-cultured them with peripheral blood mononuclear cells (PBMCs). Our aim was to use imaging flow cytometry to investigate EV interaction with PBMCs as well as study their effects on T-lymphocyte populations to better understand their possible biological functions. As a conclusion, TREVs interacted with PBMCs more than EryEVs. Distinctively, TREVs were uptaken into CD11c+ monocytes rapidly and into CD19+ B-lymphocytes in 24 h. EryEVs were not uptaken into CD11c+ monocytes before the 24-h time point, and they were only seen on the surface of lymphocytes. Neither TREVs nor EryEV were uptaken into CD3+ T-lymphocytes and no effect on T-cell populations was detected. We have previously seen similar differences in targeting PC-3 cancer cells. Further studies are needed to address the functional properties of blood cell concentrate-derived EVs. This study demonstrates that imaging flow cytometry can be used to study the distinctive differences in the interaction and uptake of EVs. Considering our current and previous results, EVs present a new valuable component for the future development of blood-derived therapeutics.
Collapse
Affiliation(s)
| | - Reetta Pusa
- Finnish Red Cross Blood ServiceHelsinkiFinland
| | | | | | - Ulla Impola
- Finnish Red Cross Blood ServiceHelsinkiFinland
| |
Collapse
|
34
|
Wies Mancini VSB, Mattera VS, Pasquini JM, Pasquini LA, Correale JD. Microglia-derived extracellular vesicles in homeostasis and demyelination/remyelination processes. J Neurochem 2024; 168:3-25. [PMID: 38055776 DOI: 10.1111/jnc.16011] [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: 07/12/2023] [Revised: 10/10/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Microglia (MG) play a crucial role as the predominant myeloid cells in the central nervous system and are commonly activated in multiple sclerosis. They perform essential functions under normal conditions, such as actively surveying the surrounding parenchyma, facilitating synaptic remodeling, engulfing dead cells and debris, and protecting the brain against infectious pathogens and harmful self-proteins. Extracellular vesicles (EVs) are diverse structures enclosed by a lipid bilayer that originate from intracellular endocytic trafficking or the plasma membrane. They are released by cells into the extracellular space and can be found in various bodily fluids. EVs have recently emerged as a communication mechanism between cells, enabling the transfer of functional proteins, lipids, different RNA species, and even fragments of DNA from donor cells. MG act as both source and recipient of EVs. Consequently, MG-derived EVs are involved in regulating synapse development and maintaining homeostasis. These EVs also directly influence astrocytes, significantly increasing the release of inflammatory cytokines like IL-1β, IL-6, and TNF-α, resulting in a robust inflammatory response. Furthermore, EVs derived from inflammatory MG have been found to inhibit remyelination, whereas Evs produced by pro-regenerative MG effectively promote myelin repair. This review aims to provide an overview of the current understanding of MG-derived Evs, their impact on neighboring cells, and the cellular microenvironment in normal conditions and pathological states, specifically focusing on demyelination and remyelination processes.
Collapse
Affiliation(s)
- V S B Wies Mancini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - V S Mattera
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - J M Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - L A Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - J D Correale
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Neurología, Fleni, Buenos Aires, Argentina
| |
Collapse
|
35
|
Izquierdo-Altarejos P, Moreno-Manzano V, Felipo V. Pathological and therapeutic effects of extracellular vesicles in neurological and neurodegenerative diseases. Neural Regen Res 2024; 19:55-61. [PMID: 37488844 PMCID: PMC10479838 DOI: 10.4103/1673-5374.375301] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/08/2023] [Accepted: 04/20/2023] [Indexed: 07/26/2023] Open
Abstract
Extracellular vesicles are released by all cell types and contain proteins, microRNAs, mRNAs, and other bioactive molecules. Extracellular vesicles play an important role in intercellular communication and in the modulation of the immune system and neuroinflammation. The cargo of extracellular vesicles (e.g., proteins and microRNAs) is altered in pathological situations. Extracellular vesicles contribute to the pathogenesis of many pathologies associated with sustained inflammation and neuroinflammation, including cancer, diabetes, hyperammonemia and hepatic encephalopathy, and other neurological and neurodegenerative diseases. Extracellular vesicles may cross the blood-brain barrier and transfer pathological signals from the periphery to the brain. This contributes to inducing neuroinflammation and cognitive and motor impairment in hyperammonemia and hepatic encephalopathy and in neurodegenerative diseases. The mechanisms involved are beginning to be understood. For example, increased tumor necrosis factor α in extracellular vesicles from plasma of hyperammonemic rats induces neuroinflammation and motor impairment when injected into normal rats. Identifying the mechanisms by which extracellular vesicles contribute to the pathogenesis of these diseases will help to develop new treatments and diagnostic tools for their easy and early detection. In contrast, extracellular vesicles from mesenchymal stem cells have therapeutic utility in many of the above pathologies, by reducing inflammation and neuroinflammation and improving cognitive and motor function. These extracellular vesicles recapitulate the beneficial effects of mesenchymal stem cells and have advantages as therapeutic tools: they are less immunogenic, may not differentiate to malignant cells, cross the blood-brain barrier, and may reach more easily target organs. Extracellular vesicles from mesenchymal stem cells have beneficial effects in models of ischemic brain injury, Alzheimer's and Parkinson's diseases, hyperammonemia, and hepatic encephalopathy. Extracellular vesicles from mesenchymal stem cells modulate the immune system, promoting the shift from a pro-inflammatory to an anti-inflammatory state. For example, extracellular vesicles from mesenchymal stem cells modulate the Th17/Treg balance, promoting the anti-inflammatory Treg. Extracellular vesicles from mesenchymal stem cells may also act directly in the brain to modulate microglia activation, promoting a shift from a pro-inflammatory to an anti-inflammatory state. This reduces neuroinflammation and improves cognitive and motor function. Two main components of extracellular vesicles from mesenchymal stem cells which contribute to these beneficial effects are transforming growth factor-β and miR-124. Identifying the mechanisms by which extracellular vesicles from mesenchymal stem cells induce the beneficial effects and the main molecules (e.g., proteins and mRNAs) involved may help to improve their therapeutic utility. The aims of this review are to summarize the knowledge of the pathological effects of extracellular vesicles in different pathologies, the therapeutic potential of extracellular vesicles from mesenchymal stem cells to recover cognitive and motor function and the molecular mechanisms for these beneficial effects on neurological function.
Collapse
Affiliation(s)
| | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Laboratory, Centro Investigación Príncipe Felipe, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
36
|
Ning M, Hua S, Ma Y, Liu Y, Wang D, Xu K, Yu H. Microvesicles facilitate the differentiation of mesenchymal stem cells into pancreatic beta-like cells via miR-181a-5p/150-5p. Int J Biol Macromol 2024; 254:127719. [PMID: 37918601 DOI: 10.1016/j.ijbiomac.2023.127719] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/04/2023]
Abstract
Transplantation of pancreatic islet cells is a promising strategy for the long-term treatment of type 1 diabetes (T1D). The stem cell-derived beta cells showed great potential as substitute sources of transplanted pancreatic islet cells. However, the current efficiency of stem cell differentiation still cannot match the requirements for clinical transplantation. Here, we report that microvesicles (MVs) from insulin-producing INS-1 cells could induce mesenchymal stem cell (MSC) differentiation into pancreatic beta-like cells. The combination of MVs with small molecules, nicotinamide and insulin-transferrin-selenium (ITS), dramatically improved the efficiency of MSC differentiation. Notably, the function of MVs in MSC differentiation requires their entry into MSCs through giant pinocytosis. The MVs-treated or MVs combined with small molecules-treated MSCs show pancreatic beta-like cell morphology and response to glucose stimulation in insulin secretion. Using high throughput small RNA-sequencing, we found that MVs induced MSC differentiation into the beta-like cells through miR-181a-5p/150-5p. Together, our findings reveal the role of MVs or the MV-enriched miR-181a-5p/150-5p as a class of biocompatible reagents to differentiate MSCs into functional beta-like cells and demonstrate that the combined usage of MVs or miR-181a-5p/150-5p with small molecules can potentially be used in making pancreatic islet cells for future clinical purposes.
Collapse
Affiliation(s)
- Mingming Ning
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Shanshan Hua
- Department of Spine Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao 266071, China
| | - Ying Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yunpeng Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Dianliang Wang
- Stem cell and tissue engineering research laboratory, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China.
| | - Kai Xu
- Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
37
|
Raza F, Zheng M, Zhong H, Su J, He B, Yuan WE, Qiu M. Engineered tumor microvesicles modified by SP94 peptide for arsenic trioxide targeting drug delivery in liver cancer therapy. BIOMATERIALS ADVANCES 2023; 155:213683. [PMID: 37925825 DOI: 10.1016/j.bioadv.2023.213683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Liver cancer is among the leading cause of cancer related death worldwide. There is growing interest in using traditional Chinese medicines such as arsenic trioxide (ATO) to treat liver cancer. ATO have attracted attention due to its wide range of anti-cancer activities. However, the current ATO formulations are associated with drawbacks such as short half-life, lack of targeting ability towards solid tumors and apparent toxic side effects. Tumor microvesicles (TMVs) has shown encouraging results for the delivery of drugs to solid tumor. In this work, we designed ATO loaded TMVs further modified by SP94 peptide as liver cancer specific ligand (ATO@SP94-TMVs). This drug delivery system utilized SP94 peptide that selectively targets liver cancer cells while TMVs increase the accumulation of ATO at tumor site and activate immune response owing to the associated antigens. ATO@SP94-TMVs exhibited high encapsulation efficiency and tumor microenvironment triggered enhanced release of ATO in vitro. Cytotoxicity and uptake studies revealed remarkable inhibition and specific targeting of H22 cells. In addition, excellent immune response was detected in vitro, enhancing anti-tumor efficacy. Furthermore, a tumor inhibition rate of about 53.23 % was observed in H22 bearing tumor model. Overall, these results confirm that ATO@SP94-TMVs can be a promising nano drug delivery system for the future liver cancer therapy and improve its clinical applications.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mengyuan Zheng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hongyu Zhong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Beixuan He
- Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Wei-En Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
38
|
Santiago JV, Natu A, Ramelow CC, Rayaprolu S, Xiao H, Kumar V, Kumar P, Seyfried NT, Rangaraju S. Identification of State-Specific Proteomic and Transcriptomic Signatures of Microglia-Derived Extracellular Vesicles. Mol Cell Proteomics 2023; 22:100678. [PMID: 37952696 PMCID: PMC10755493 DOI: 10.1016/j.mcpro.2023.100678] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/26/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023] Open
Abstract
Microglia are resident immune cells of the brain that play important roles in mediating inflammatory responses in several neurological diseases via direct and indirect mechanisms. One indirect mechanism may involve extracellular vesicle (EV) release, so that the molecular cargo transported by microglia-derived EVs can have functional effects by facilitating intercellular communication. The molecular composition of microglia-derived EVs, and how microglial activation states impact EV composition and EV-mediated effects in neuroinflammation, remain poorly understood. We hypothesize that microglia-derived EVs have unique molecular profiles that are determined by microglial activation state. Using size-exclusion chromatography to purify EVs from BV2 microglia, combined with proteomic (label-free quantitative mass spectrometry or LFQ-MS) and transcriptomic (mRNA and noncoding RNA seq) methods, we obtained comprehensive molecular profiles of microglia-derived EVs. LFQ-MS identified several classic EV proteins (tetraspanins, ESCRT machinery, and heat shock proteins), in addition to over 200 proteins not previously reported in the literature. Unique mRNA and microRNA signatures of microglia-derived EVs were also identified. After treating BV2 microglia with lipopolysaccharide (LPS), interleukin-10, or transforming growth factor beta, to mimic pro-inflammatory, anti-inflammatory, or homeostatic states, respectively, LFQ-MS and RNA seq revealed novel state-specific proteomic and transcriptomic signatures of microglia-derived EVs. Particularly, LPS treatment had the most profound impact on proteomic and transcriptomic compositions of microglia-derived EVs. Furthermore, we found that EVs derived from LPS-activated microglia were able to induce pro-inflammatory transcriptomic changes in resting responder microglia, confirming the ability of microglia-derived EVs to relay functionally relevant inflammatory signals. These comprehensive microglia-EV molecular datasets represent important resources for the neuroscience and omics communities and provide novel insights into the role of microglia-derived EVs in neuroinflammation.
Collapse
Affiliation(s)
- Juliet V Santiago
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Aditya Natu
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Christina C Ramelow
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Sruti Rayaprolu
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Hailian Xiao
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Vishnu Kumar
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Prateek Kumar
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA
| | - Nicholas T Seyfried
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA; Department of Biochemistry, Emory University, Atlanta, Georgia, USA
| | - Srikant Rangaraju
- Department of Neurology, Emory University, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia, USA.
| |
Collapse
|
39
|
Szatmári T, Balázs K, Csordás IB, Sáfrány G, Lumniczky K. Effect of radiotherapy on the DNA cargo and cellular uptake mechanisms of extracellular vesicles. Strahlenther Onkol 2023; 199:1191-1213. [PMID: 37347291 DOI: 10.1007/s00066-023-02098-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/01/2023] [Indexed: 06/23/2023]
Abstract
In the past decades, plenty of evidence has gathered pointing to the role of extracellular vesicles (EVs) secreted by irradiated cells in the development of radiation-induced non-targeted effects. EVs are complex natural structures composed of a phospholipid bilayer which are secreted by virtually all cells and carry bioactive molecules. They can travel certain distances in the body before being taken up by recipient cells. In this review we discuss the role and fate of EVs in tumor cells and highlight the importance of DNA specimens in EVs cargo in the context of radiotherapy. The effect of EVs depends on their cargo, which reflects physiological and pathological conditions of donor cell types, but also depends on the mode of EV uptake and mechanisms involved in the route of EV internalization. While the secretion and cargo of EVs from irradiated cells has been extensively studied in recent years, their uptake is much less understood. In this review, we will focus on recent knowledge regarding the EV uptake of cancer cells and the effect of radiation in this process.
Collapse
Affiliation(s)
- Tünde Szatmári
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, 1097, Budapest, Hungary.
| | - Katalin Balázs
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, 1097, Budapest, Hungary
| | - Ilona Barbara Csordás
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, 1097, Budapest, Hungary
| | - Géza Sáfrány
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, 1097, Budapest, Hungary
| | - Katalin Lumniczky
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, 1097, Budapest, Hungary
| |
Collapse
|
40
|
Sacerdoti F, Gomez FD, Jancic C, Lombardo T, Pascuale CA, Moretton MA, Chiappetta DA, Ibarra C, Amaral MM. Detection and characterization of circulating microvesicles containing Shiga toxin type 2 in a rat model of Hemolytic Uremic Syndrome. Toxicon 2023; 236:107349. [PMID: 37979924 DOI: 10.1016/j.toxicon.2023.107349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023]
Abstract
Shiga toxin producing Escherichia coli (STEC) are foodborne pathogens that release Shiga toxin (Stx), virulence factor responsible for the development of Hemolytic Uremic Syndrome (HUS). Stx causes endothelial cell damage, which leads to platelets deposition and thrombi formation within the microvasculature. It has been described that Stx activates blood cells and induces the shedding of proinflammatory and prothrombotic microvesicles (MVs) containing the toxin. In this sense, it has been postulated that MVs containing Stx2 (MVs-Stx2+) can contribute to the physiopathology of HUS, allowing Stx2 to reach the target organs while evading the immune system. In this work, we propose that circulating MVs-Stx2+ can be a potential biomarker for the diagnosis and prognosis of STEC infections and HUS progression. We developed a rat HUS model by the intraperitoneal injection of a sublethal dose of Stx2 and observed: decrease in body weight, increase of creatinine and urea levels, decrease of creatinine clearance and histological renal damages. After characterization of renal damages, we investigated circulating total MVs and MVs-Stx2+ by flow cytometry at different times after Stx2 injection. Additionally, we evaluated the correlation of biochemical parameters such as creatinine and urea in plasma with MVs-Stx2+. As a result, we found a significant circulation of MVs-Stx2+ at 72 and 96 h after Stx2 injection, nevertheless no correlation with creatinine and urea plasma levels were detected. Our results suggest that MVs-Stx2+ may be an additional biomarker for the characterization and diagnosis of HUS progression. A further analysis is required in order to validate MVs-Stx2+ as biomarker of the disease.
Collapse
Affiliation(s)
- Flavia Sacerdoti
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas. Laboratorio de Fisiopatogenia. Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay). Buenos Aires, Argentina.
| | - Fernando D Gomez
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas. Laboratorio de Fisiopatogenia. Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay). Buenos Aires, Argentina
| | - Carolina Jancic
- Instituto de Medicina Experimental - CONICET - Academia Nacional de Medicina, Buenos Aires, Argentina; Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tomas Lombardo
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Buenos Aires, Argentina; Instituto de Estudios de la Inmunidad Humoral Dr. R.A. Margni (IDEHU), UBA-CONICET, Argentina
| | - Carla A Pascuale
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA) - CONICET, Argentina
| | - Marcela A Moretton
- Cátedra de Tecnología Farmacéutica I, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina; Universidad de Buenos Aires, Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Diego A Chiappetta
- Cátedra de Tecnología Farmacéutica I, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina; Universidad de Buenos Aires, Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Cristina Ibarra
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas. Laboratorio de Fisiopatogenia. Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay). Buenos Aires, Argentina
| | - María M Amaral
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas. Laboratorio de Fisiopatogenia. Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay). Buenos Aires, Argentina.
| |
Collapse
|
41
|
Young TW, Kappler MP, Hockaden NM, Carpenter RL, Jacobson SC. Characterization of Extracellular Vesicles by Resistive-Pulse Sensing on In-Plane Multipore Nanofluidic Devices. Anal Chem 2023; 95:16710-16716. [PMID: 37916500 PMCID: PMC10841850 DOI: 10.1021/acs.analchem.3c03546] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived, naturally produced, membrane-bound nanoscale particles that are linked to cell-cell communication and the propagation of diseases. Here, we report the design and testing of in-plane nanofluidic devices for resistive-pulse measurements of EVs derived from bovine milk and human breast cancer cells. The devices were fabricated in plane with three nanopores in series to determine the particle volume and diameter, two pore-to-pore regions to measure the electrophoretic mobility and zeta potential, and an in-line filter to prevent cellular debris and aggregates from entering the nanopore region. Devices were tested with and without the channels coated with a short-chain PEG silane to minimize electroosmotic flow and permit an accurate measurement of the electrophoretic mobility and zeta potential of the EVs. To enhance throughput of EVs, vacuum was applied to the waste reservoir to increase particle frequencies up to 1000 min-1. The nanopores had cross-sections 200 nm wide and 200 nm deep and easily resolved EV diameters from 60 to 160 nm. EVs from bovine milk and human breast cancer cells had similar particle size distributions, but their zeta potentials differed by 2-fold, -8 ± 1 and -4 ± 1 mV, respectively.
Collapse
Affiliation(s)
- Tanner W Young
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| | - Michael P Kappler
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| | - Natasha M Hockaden
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-Bloomington, Bloomington, Indiana 47405-7005, United States
| | - Richard L Carpenter
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-Bloomington, Bloomington, Indiana 47405-7005, United States
| | - Stephen C Jacobson
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405-7102, United States
| |
Collapse
|
42
|
Sui C, Liao Z, Bai J, Hu D, Yue J, Yang S. Current knowledge on the role of extracellular vesicles in endometrial receptivity. Eur J Med Res 2023; 28:471. [PMID: 37899459 PMCID: PMC10614333 DOI: 10.1186/s40001-023-01459-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/19/2023] [Indexed: 10/31/2023] Open
Abstract
Endometrial receptivity has been widely understood as the capacity of the endometrium to receive implantable embryos. The establishment of endometrial receptivity involves multiple biological processes including decidualization, tissue remodeling, angiogenesis, immune regulation, and oxidative metabolism. Extracellular vesicles (EVs) are lipid-bilayer-membrane nanosized vesicles mediating cell-to-cell communication. Recently, EVs and their cargo have been proven as functional factors in the establishment of endometrial receptivity. In this review, we comprehensively summarized the alteration of endometrium/embryo-derived EVs during the receptive phase and retrospected the current findings which revealed the pivotal role and potential mechanism of EVs to promote successful implantation. Furthermore, we highlight the potentiality and limitations of EVs being translated into clinical applications such as biomarkers of endometrial receptivity or reproductive therapeutic mediators, and point out the direction for further research.
Collapse
Affiliation(s)
- Cong Sui
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Zhiqi Liao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Jian Bai
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Dan Hu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Jing Yue
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Shulin Yang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
43
|
Wang Y, Zou M, Zhao Y, Kabir MA, Peng X. Exosomal microRNA/miRNA Dysregulation in Respiratory Diseases: From Mycoplasma-Induced Respiratory Disease to COVID-19 and Beyond. Cells 2023; 12:2421. [PMID: 37830635 PMCID: PMC10571955 DOI: 10.3390/cells12192421] [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: 08/03/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
Respiratory diseases represent a significant economic and health burden worldwide, affecting millions of individuals each year in both human and animal populations. MicroRNAs (miRNAs) play crucial roles in gene expression regulation and are involved in various physiological and pathological processes. Exosomal miRNAs and cellular miRNAs have been identified as key regulators of several immune respiratory diseases, such as chronic respiratory diseases (CRD) caused by Mycoplasma gallisepticum (MG), Mycoplasma pneumoniae pneumonia (MMP) caused by the bacterium Mycoplasma pneumoniae, coronavirus disease 2019 (COVID-19), chronic obstructive pulmonary disease (COPD), asthma, and acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Consequently, miRNAs seem to have the potential to serve as diagnostic biomarkers and therapeutic targets in respiratory diseases. In this review, we summarize the current understanding of the functional roles of miRNAs in the above several respiratory diseases and discuss the potential use of miRNAs as stable diagnostic biomarkers and therapeutic targets for several immune respiratory diseases, focusing on the identification of differentially expressed miRNAs and their targeting of various signaling pathways implicated in disease pathogenesis. Despite the progress made, unanswered questions and future research directions are discussed to facilitate personalized and targeted therapies for patients with these debilitating conditions.
Collapse
Affiliation(s)
| | | | | | | | - Xiuli Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (M.Z.); (Y.Z.); (M.A.K.)
| |
Collapse
|
44
|
Neupane KR, Ramon GS, Harvey B, Chun B, Aryal SP, Masud AA, McCorkle JR, Kolesar JM, Kekenes‐Huskey PM, Richards CI. Programming Cell-Derived Vesicles with Enhanced Immunomodulatory Properties. Adv Healthc Mater 2023; 12:e2301163. [PMID: 37377147 PMCID: PMC11070110 DOI: 10.1002/adhm.202301163] [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: 04/12/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 06/29/2023]
Abstract
Tumor-associated macrophages are the predominant immune cells present in the tumor microenvironment and mostly exhibit a pro-tumoral M2-like phenotype. However, macrophage biology is reversible allowing them to acquire an anti-tumoral M1-like phenotype in response to external stimuli. A potential therapeutic strategy for treating cancer may be achieved by modulating macrophages from an M2 to an M1-like phenotype with the tumor microenvironment. Here, programmed nanovesicles are generated as an immunomodulatory therapeutic platform with the capability to re-polarize M2 macrophages toward a proinflammatory phenotype. Programmed nanovesicles are engineered from cellular membranes to have specific immunomodulatory properties including the capability to bidirectionally modulate immune cell polarization. These programmed nanovesicles decorated with specific membrane-bound ligands can be targeted toward specific cell types including immune cells. Macrophage-derived vesicles are engineered to enhance immune cell reprogramming toward a proinflammatory phenotype.
Collapse
Affiliation(s)
- Khaga R. Neupane
- Department of ChemistryUniversity of Kentucky506 Library Drive, 125 Chemistry‐Physics BuildingLexingtonKY40506USA
| | - Geraldine S. Ramon
- Department of Cell and Molecular PhysiologyLoyola University ChicagoChicagoILUSA
| | - Brock Harvey
- Department of ChemistryUniversity of Kentucky506 Library Drive, 125 Chemistry‐Physics BuildingLexingtonKY40506USA
| | - Byeong Chun
- Department of Cell and Molecular PhysiologyLoyola University ChicagoChicagoILUSA
| | - Surya P. Aryal
- Department of ChemistryUniversity of Kentucky506 Library Drive, 125 Chemistry‐Physics BuildingLexingtonKY40506USA
| | - Abdullah A. Masud
- Department of ChemistryUniversity of Kentucky506 Library Drive, 125 Chemistry‐Physics BuildingLexingtonKY40506USA
| | - J. Robert McCorkle
- Department of Pharmacy Practice and ScienceCollege of PharmacyUniversity of KentuckyLexingtonKY40508USA
| | - Jill M. Kolesar
- Department of Pharmacy Practice and ScienceCollege of PharmacyUniversity of KentuckyLexingtonKY40508USA
| | | | - Christopher I. Richards
- Department of ChemistryUniversity of Kentucky506 Library Drive, 125 Chemistry‐Physics BuildingLexingtonKY40506USA
| |
Collapse
|
45
|
Liu J, Hu X. Fungal extracellular vesicle-mediated regulation: from virulence factor to clinical application. Front Microbiol 2023; 14:1205477. [PMID: 37779707 PMCID: PMC10540631 DOI: 10.3389/fmicb.2023.1205477] [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: 04/13/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023] Open
Abstract
Invasive fungal disease (IFD) poses a significant threat to immunocompromised patients and remains a global challenge due to limited treatment options, high mortality and morbidity rates, and the emergence of drug-resistant strains. Despite advancements in antifungal agents and diagnostic techniques, the lack of effective vaccines, standardized diagnostic tools, and efficient antifungal drugs contributes to the ongoing impact of invasive fungal infections (IFI). Recent studies have highlighted the presence of extracellular vesicles (EVs) released by fungi carrying various components such as enzymes, lipids, nucleic acids, and virulence proteins, which play roles in both physiological and pathological processes. These fungal EVs have been shown to interact with the host immune system during the development of fungal infections whereas their functional role and potential application in patients are not yet fully understood. This review summarizes the current understanding of the biologically relevant findings regarding EV in host-pathogen interaction, and aim to describe our knowledge of the roles of EV as diagnostic tools and vaccine vehicles, offering promising prospects for the treatment of IFI patients.
Collapse
Affiliation(s)
| | - Xiaoping Hu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
46
|
Khani-Eshratabadi M, Mousavi SH, Zarrabi M, Motallebzadeh Khanmiri J, Zeinali Bardar Z. Human Umbilical Cord Mesenchymal Stem Cell-Derived Microvesicles Could Induce Apoptosis and Autophagy in Acute Myeloid Leukemia. IRANIAN BIOMEDICAL JOURNAL 2023; 27:247-56. [PMID: 37873637 PMCID: PMC10707811 DOI: 10.61186/ibj.27.5.247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/31/2023] [Indexed: 12/17/2023]
Abstract
Background Microvesicles (MV) have been identified as candidate biomarkers for treating acute myeloid leukemia (AML). This study investigated the effects of human umbilical cord-derived mesenchymal stem cell (hUCMSC)-derived MVs on apoptosis and autophagy in the KG-1 leukemic cell line. Methods The hUCMSCs were cultured and characterized by flow cytometry. MVs were isolated by ultracentrifugation, and the concentration was determined using the Bradford method. The characteristics of MVs were confirmed using transmission electron microscopy, flow cytometry, and dynamic light scattering methods. KG-1 cells were treated with the desired concentrations of MVs for 24 h. The apoptosis induction and reactive oxygen species production were evaluated using flow cytometry. RT-PCR was performed to evaluate apoptosis- and autophagy-related genes expression. Results Following tretment of KG-1 cells with 25, 50, and 100 μg/ml concentrations of MVs, the apoptosis rates were 47.85%, 47.15%, and 51.35% (p < 0.0001), and the autophagy-induced ROS levels were 73.9% (p < 0.0002), 84.8% (p < 0.0001), and 85.4% (p < 0.0001), respectively. BAX and ATG7 gene expression increased significantly at all concentrations compared to the control, and this level was higher at 50 μg/ml than that of the other concentrations. In addition, LC3 and Beclin 1 expression increased significantly in a concentration-dependen manner. Conversely, BCL2 expression decreased compared to the control. Conclusion Our findings indicate that hUCMSC-MVs could induce cell death pathways of autophagy and apoptosis in the KG-1 cell lines and exert potent antiproliferative and proapoptotic effects on KG-1 cells in vitro. Therefore, hUCMSC-MVs may be a potential approach for cancer therapy as a novel cell-to-cell communication strategy.
Collapse
Affiliation(s)
- Mohammad Khani-Eshratabadi
- Department of Hematology and Blood Transfusion Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
- Kashmar School of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hadi Mousavi
- Department of Hematology and Blood Transfusion Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Zarrabi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Jamal Motallebzadeh Khanmiri
- Department of Hematology and Blood Transfusion Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Zeinali Bardar
- Kashmar School of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
47
|
Li Z, Lei Z, Cai Y, Cheng DB, Sun T. MicroRNA therapeutics and nucleic acid nano-delivery systems in bacterial infection: a review. J Mater Chem B 2023; 11:7804-7833. [PMID: 37539650 DOI: 10.1039/d3tb00694h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Bacteria that have worked with humans for thousands of years pose a major threat to human health even today, as drug resistance has become a prominent problem. Compared to conventional drug therapy, nucleic acid-based therapies are a promising and potential therapeutic strategy for diseases in which nucleic acids are delivered through a nucleic acid delivery system to regulate gene expression in specific cells, offering the possibility of curing intractable diseases that are difficult to treat at this stage. Among the many nucleic acid therapeutic ideas, microRNA, a class of small nucleic acids with special properties, has made great strides in biology and medicine in just over two decades, showing promise in preclinical drug development. In this review, we introduce recent advances in nucleic acid delivery systems and their clinical applications, highlighting the potential of nucleic acid therapies, especially miRNAs extracted from traditional herbs, in combination with the existing set of nucleic acid therapeutic systems, to potentially open up a new line of thought in the treatment of cancer, viruses, and especially bacterial infectious diseases.
Collapse
Affiliation(s)
- Ze Li
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Yilun Cai
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| |
Collapse
|
48
|
Cuadrado-Payán E, Ramírez-Bajo MJ, Bañón-Maneus E, Rovira J, Diekmann F, Revuelta I, Cucchiari D. Physiopathological role of extracellular vesicles in alloimmunity and kidney transplantation and their use as biomarkers. Front Immunol 2023; 14:1154650. [PMID: 37662919 PMCID: PMC10469977 DOI: 10.3389/fimmu.2023.1154650] [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: 01/30/2023] [Accepted: 07/21/2023] [Indexed: 09/05/2023] Open
Abstract
Antibody-mediated rejection is the leading cause of kidney graft dysfunction. The process of diagnosing it requires the performance of an invasive biopsy and subsequent histological examination. Early and sensitive biomarkers of graft damage and alloimmunity are needed to identify graft injury and eventually limit the need for a kidney biopsy. Moreover, other scenarios such as delayed graft function or interstitial fibrosis and tubular atrophy face the same problem. In recent years, interest has grown around extracellular vesicles, specifically exosomes actively secreted by immune cells, which are intercellular communicators and have shown biological significance. This review presents their potential as biomarkers in kidney transplantation and alloimmunity.
Collapse
Affiliation(s)
- Elena Cuadrado-Payán
- Department of Nephrology and Kidney Transplantation, Hospital Clínic, Barcelona, Spain
- Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - María José Ramírez-Bajo
- Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
- Red de Investigación Renal (REDINREN), Insituto de Salud Carlos III, Madrid, Spain
| | - Elisenda Bañón-Maneus
- Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
- Red de Investigación Renal (REDINREN), Insituto de Salud Carlos III, Madrid, Spain
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
- Red de Investigación Renal (REDINREN), Insituto de Salud Carlos III, Madrid, Spain
| | - Fritz Diekmann
- Department of Nephrology and Kidney Transplantation, Hospital Clínic, Barcelona, Spain
- Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
- Red de Investigación Renal (REDINREN), Insituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Revuelta
- Department of Nephrology and Kidney Transplantation, Hospital Clínic, Barcelona, Spain
- Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
- Red de Investigación Renal (REDINREN), Insituto de Salud Carlos III, Madrid, Spain
| | - David Cucchiari
- Department of Nephrology and Kidney Transplantation, Hospital Clínic, Barcelona, Spain
- Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| |
Collapse
|
49
|
Poledniczek M, Neumayer C, Kopp CW, Schlager O, Gremmel T, Jozkowicz A, Gschwandtner ME, Koppensteiner R, Wadowski PP. Micro- and Macrovascular Effects of Inflammation in Peripheral Artery Disease-Pathophysiology and Translational Therapeutic Approaches. Biomedicines 2023; 11:2284. [PMID: 37626780 PMCID: PMC10452462 DOI: 10.3390/biomedicines11082284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Inflammation has a critical role in the development and progression of atherosclerosis. On the molecular level, inflammatory pathways negatively impact endothelial barrier properties and thus, tissue homeostasis. Conformational changes and destruction of the glycocalyx further promote pro-inflammatory pathways also contributing to pro-coagulability and a prothrombotic state. In addition, changes in the extracellular matrix composition lead to (peri-)vascular remodelling and alterations of the vessel wall, e.g., aneurysm formation. Moreover, progressive fibrosis leads to reduced tissue perfusion due to loss of functional capillaries. The present review aims at discussing the molecular and clinical effects of inflammatory processes on the micro- and macrovasculature with a focus on peripheral artery disease.
Collapse
Affiliation(s)
- Michael Poledniczek
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria;
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Oliver Schlager
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Thomas Gremmel
- Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria;
- Institute of Cardiovascular Pharmacotherapy and Interventional Cardiology, Karl Landsteiner Society, 3100 St. Pölten, Austria
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, 31-007 Krakow, Poland;
| | - Michael E. Gschwandtner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Renate Koppensteiner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| |
Collapse
|
50
|
Wang Z, Tan W, Li B, Zou J, Li Y, Xiao Y, He Y, Yoshida S, Zhou Y. Exosomal non-coding RNAs in angiogenesis: Functions, mechanisms and potential clinical applications. Heliyon 2023; 9:e18626. [PMID: 37560684 PMCID: PMC10407155 DOI: 10.1016/j.heliyon.2023.e18626] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/11/2023] Open
Abstract
Exosomes are extracellular vesicles that can be produced by most cells. Exosomes act as important intermediaries in intercellular communication, and participate in a variety of biological activities between cells. Non-coding RNAs (ncRNAs) usually refer to RNAs that do not encode proteins. Although ncRNAs have no protein-coding capacity, they are able to regulate gene expression at multiple levels. Angiogenesis is the formation of new blood vessels from pre-existing vessels, which is an important physiological process. However, abnormal angiogenesis could induce many diseases such as atherosclerosis, diabetic retinopathy and cancer. Many studies have shown that ncRNAs can stably exist in exosomes and play a wide range of physiological and pathological roles including regulation of angiogenesis. In brief, some specific ncRNAs can be enriched in exosomes secreted by cells and absorbed by recipient cells through the exosome pathway, thus activating relevant signaling pathways in target cells and playing a role in regulating angiogenesis. In this review, we describe the physiological and pathological functions of exosomal ncRNAs in angiogenesis, summarize their role in angiogenesis-related diseases, and illustrate potential clinical applications like novel drug therapy strategies and diagnostic markers in exosome research as inspiration for future investigations.
Collapse
Affiliation(s)
- Zicong Wang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Wei Tan
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jingling Zou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yun Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yangyan Xiao
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yan He
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| |
Collapse
|